1
|
Mousa A, Cuomo-Dannenburg G, Thompson HA, Bell DJ, D'Alessandro U, Gosling R, Nahum A, Barnes KI, Raman J, Workmann L, Foo YS, Flegg JA, Hocke EF, Hansson H, Chopo-Pizarro A, Beshir KB, Alifrangis M, Chico RM, Sutherland CJ, Okell LC, Roper C. Impact of dhps mutations on sulfadoxine-pyrimethamine protective efficacy and implications for malaria chemoprevention. Nat Commun 2025; 16:4268. [PMID: 40341172 PMCID: PMC12062426 DOI: 10.1038/s41467-025-58326-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/18/2025] [Indexed: 05/10/2025] Open
Abstract
Sulfadoxine-pyrimethamine (SP) is recommended for perennial malaria chemoprevention in young children in high burden areas across Africa. Mutations in the dihydropteroate synthase (dhps) gene (437 G/540E/581 G) associated with sulfadoxine resistance vary regionally, but their effect on SP protective efficacy is unclear. We retrospectively analyse time to microscopy and PCR-confirmed re-infection in seven efficacy trials including 1639 participants in 12 sites across Africa. We estimate the duration of SP protection against parasites with different genotypes using a Bayesian mathematical model that accounts for variation in transmission intensity and genotype frequencies. The longest duration of SP protection is >42 days against dhps sulfadoxine-susceptible parasites and 30.3 days (95%Credible Interval (CrI):17.1-45.1) against the West-African genotype dhps GKA (437G-K540-A581). A shorter duration of protection is estimated against parasites with additional mutations in the dhps gene, with 16.5 days (95%CrI:11.2-37.4) protection against parasites with the east-African genotype dhps GEA (437G-540E-A581) and 11.7 days (95%CrI:8.0-21.9) against highly resistant parasites carrying the dhps GEG (437G-540E-581G) genotype. Using these estimates and modelled genotype frequencies we map SP protection across Africa. This approach and our estimated parameters can be directly applied to any setting using local genomic surveillance data to inform decision-making on where to scale-up SP-based chemoprevention or consider alternatives.
Collapse
Affiliation(s)
- Andria Mousa
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK.
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, W12 0BZ, London, UK.
| | - Gina Cuomo-Dannenburg
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, W12 0BZ, London, UK
| | - Hayley A Thompson
- Malaria and Neglected Tropical Diseases, PATH, WA 98121, Seattle, WA, US
| | - David J Bell
- Department of Infectious Disease, NHS Greater Glasgow and Clyde, G51 4TF, Glasgow, UK
| | - Umberto D'Alessandro
- MRC Unit The Gambia at the London School of Hygiene and Tropical Medicine, PO Box 273, Serrekunda, Gambia
| | - Roly Gosling
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
- Malaria Elimination Initiative, Institute of Global Health Sciences, University of California San Francisco, CA, 94158, San Francisco, USA
| | - Alain Nahum
- Centre de recherche entomologique de Cotonou, 2604, Cotonou, Benin
| | - Karen I Barnes
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, 7925, Cape Town, South Africa
| | - Jaishree Raman
- South African National Institute for Communicable Diseases, 2192, Johannesburg, South Africa
- Wits Research Institute for Malaria, Faculty of Health Sciences, University of Witwatersrand, 2193, Johannesburg, South Africa
| | - Lesley Workmann
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, 7925, Cape Town, South Africa
| | - Yong See Foo
- School of Mathematics and Statistics, The University of Melbourne, 3052, Parkville, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, The University of Melbourne, 3052, Parkville, Australia
| | - Emma Filtenborg Hocke
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Helle Hansson
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Ana Chopo-Pizarro
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Khalid B Beshir
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Michael Alifrangis
- Department of Immunology and Microbiology, Centre for translational Medicine and Parasitology, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - R Matthew Chico
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Colin J Sutherland
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| | - Lucy C Okell
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, W12 0BZ, London, UK
| | - Cally Roper
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, UK
| |
Collapse
|
2
|
Sen S, Braunack-Mayer L, Kelly SL, Masserey T, Malinga J, Moehrle JJ, Penny MA. Public health impact of current and proposed age-expanded perennial malaria chemoprevention: a modelling study. Sci Rep 2025; 15:10488. [PMID: 40140443 PMCID: PMC11947144 DOI: 10.1038/s41598-025-93623-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
In 2022, the World Health Organization extended their guidelines for perennial malaria chemoprevention (PMC) from infants to children up to 24 months old. However, evidence for PMC's public health impact is primarily limited to children under 15 months. Further research is needed to assess the public health impact and cost-effectiveness of PMC, and the added benefit of further age-expansion. We integrated an individual-based model of malaria with pharmacological models of drug action to address these questions for PMC and a proposed age-expanded schedule (referred as PMC+, for children 03-36 months). Across malaria prevalence settings of 5-70% and different drug sensitivity assumptions, we predicted PMC and PMC+'s median efficacy (interquartile range) of 18.6% (12.2-25.0%) and 21.9% (14.3-29.5%) against clinical disease and 9.0% (2.0-16.0%) and 10.8% (3.2-18.4%) against severe malaria, respectively, in children under three years. PMC's total impact outweighed the risk of delayed malaria in children up to age five and remained cost-effective across currently recommended transmission intensities (over 10% prevalence) when delivered through the existing Expanded Program on Immunization channels. Further empirical evidence of likely added benefit, operational feasibility and sustainability of age-expanded PMC will be essential to complement our model-based findings.
Collapse
Affiliation(s)
- Swapnoleena Sen
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Lydia Braunack-Mayer
- The Kids Research Institute Australia, Nedlands, WA, Australia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Sherrie L Kelly
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Thiery Masserey
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Josephine Malinga
- The Kids Research Institute Australia, Nedlands, WA, Australia
- Centre for Child Health Research, The University of Western Australia, Crawley, WA, Australia
| | | | - Melissa A Penny
- The Kids Research Institute Australia, Nedlands, WA, Australia.
- Centre for Child Health Research, The University of Western Australia, Crawley, WA, Australia.
| |
Collapse
|
3
|
Masserey T, Braunack-Mayer L, Miller RS, Möhrle JJ, Penny MA. A roadmap for understanding sulfadoxine-pyrimethamine in malaria chemoprevention. Parasitology 2025; 152:133-142. [PMID: 39844654 PMCID: PMC12089447 DOI: 10.1017/s0031182025000071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/24/2025]
Abstract
Sulfadoxine-pyrimethamine (SP) is the standard of care for Plasmodium falciparum malaria chemoprevention among pregnant women, infants and children. Developing alternative chemoprevention products and other prevention products, such as vaccines and monoclonal antibodies, requires significant investment. However, knowledge gaps surrounding the activity of SP and resistance put these investments at risk. Therefore, we reviewed SP's combined antimalarial action, including the individual antiplasmodial components, other antimicrobial effects, impact on malaria immunity development and continued effectiveness in settings with high SP resistance. We created a roadmap of non-clinical and clinical evidence to better understand the effectiveness of SP for chemoprevention and inform the development of new prevention tools.
Collapse
Affiliation(s)
- Thiery Masserey
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Lydia Braunack-Mayer
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | | | - Jörg J Möhrle
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
- The Kids Research Institute Australia, Nedlands, WA, Australia
- Centre for Child Health Research, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
4
|
Masserey T, Lee T, Kelly SL, Hastings IM, Penny MA. Seasonal malaria chemoprevention and the spread of Plasmodium falciparum quintuple-mutant parasites resistant to sulfadoxine-pyrimethamine: a modelling study. THE LANCET. MICROBE 2024; 5:100892. [PMID: 38996497 DOI: 10.1016/s2666-5247(24)00115-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 07/14/2024]
Abstract
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.
Collapse
Affiliation(s)
- Thiery Masserey
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Tamsin Lee
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Sherrie L Kelly
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | | | - Melissa A Penny
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland; Centre for Child Health Research, University of Western Australia, Crawley, WA, Australia; Telethon Kids Institute, Nedlands, WA, Australia.
| |
Collapse
|
5
|
Cheung YB, Ma X, Lam KF, Yung CF, Milligan P. Estimation of trajectory of protective efficacy in infectious disease prevention trials using recurrent event times. Stat Med 2024; 43:1759-1773. [PMID: 38396234 DOI: 10.1002/sim.10049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/20/2023] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
In studies of infectious disease prevention, the level of protective efficacy of medicinal products such as vaccines and prophylactic drugs tends to vary over time. Many products require administration of multiple doses at scheduled times, as opposed to one-off or continual intervention. Accurate information on the trajectory of the level of protective efficacy over time facilitates informed clinical recommendations and implementation strategies, for example, with respect to the timing of administration of the doses. Based on concepts from pharmacokinetic and pharmacodynamic modeling, we propose a non-linear function for modeling the trajectory after each dose. The cumulative effect of multiple doses of the products is captured by an additive series of the function. The model has the advantages of parsimony and interpretability, while remaining flexible in capturing features of the trajectories. We incorporate this series into the Andersen-Gill model for analysis of recurrent event time data and compare it with alternative parametric and non-parametric functions. We use data on clinical malaria disease episodes from a trial of four doses of an anti-malarial drug combination for chemoprevention to illustrate, and evaluate the performance of the methods using simulation. The proposed method out-performed the alternatives in the analysis of real data in terms of Akaike and Bayesian Information Criterion. It also accurately captured the features of the protective efficacy trajectory such as the area under curve in simulations. The proposed method has strong potential to enhance the evaluation of disease prevention measures and improve their implementation strategies.
Collapse
Affiliation(s)
- Yin Bun Cheung
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
- Programme in Health Services & Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Xiangmei Ma
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - K F Lam
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong, China
| | - Chee Fu Yung
- Infectious Disease Service, KK Women's and Children's Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Academic Medicine Department, Duke-NUS Medical School, Singapore, Singapore
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
6
|
Ibinaiye T, Rotimi K, Balogun A, Aidenagbon A, Oguoma C, Rassi C, Baker K, Oresanya O, Nnaji C. Receipt of seasonal malaria chemoprevention by age-ineligible children and associated factors in nine implementation states in Nigeria. Malar J 2024; 23:91. [PMID: 38555455 PMCID: PMC10981804 DOI: 10.1186/s12936-024-04916-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/22/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND As part of implementation quality standards, community distributors are expected to ensure that only age-eligible children (aged 3-59 months) receive seasonal malaria chemoprevention (SMC) medicines during monthly campaigns. There is uncertainty about the extent to which SMC medicines are administered to ineligible children. This study aimed to assess the magnitude of this occurrence, while exploring the factors associated with it across nine states where SMC was delivered in Nigeria during the 2022 round. METHODS This analysis was based on data from representative end-of-round SMC household surveys conducted in nine SMC-implementing states in Nigeria. Data of 3299 age-ineligible children aged > 5 years and their caregivers were extracted from the survey dataset. Prevalence of receipt of SMC medicines by ineligible children was described by child-, caregiver- and SMC-related factors. Mixed-effects multivariable logistic regression models were fitted to explore the factors associated with ineligible receipt of SMC medicines. RESULTS 30.30% (95% CI 27.80-32.90) of ineligible children sampled received at least one dose of SMC medicines in 2022, the majority (60.60%) of whom were aged 5-6 years while the rest were aged 7-10 years. There were lower odds of an age-ineligible child receiving SMC among caregivers who had knowledge of SMC age eligibility (OR: 0.53, 95% CI 0.37-0.77, p < 0.001), compared with those who were knowledgeable of age eligibility. Higher odds of receipt of SMC were found among age-ineligible children whose caregivers had higher confidence in the protective effect of SMC against malaria (OR: 2.01, 95% CI 1.07-3.72, p = 0.030), compared with those whose caregivers were less confident. Compared with ineligible children of younger caregivers (aged < 20 years), those whose caregivers were older had lower odds of receiving SMC than those whose caregivers were younger; with lower odds among children of caregivers aged 20-39 years (OR: 0.50, 95% CI 0.30-0.82, p = 0.006). CONCLUSIONS This study contributes important evidence on the magnitude of the receipt of SMC medicines by age-ineligible children, while identifying individual and contextual factors associated with it. The findings provide potentially useful insights that can help inform and guide context-specific SMC implementation quality improvement efforts.
Collapse
Affiliation(s)
- Taiwo Ibinaiye
- Malaria Consortium, 33 Pope John Paul Street, Maitama, Abuja, FCT, Nigeria.
| | - Kunle Rotimi
- Malaria Consortium, 33 Pope John Paul Street, Maitama, Abuja, FCT, Nigeria
| | | | - Adaeze Aidenagbon
- Malaria Consortium, 33 Pope John Paul Street, Maitama, Abuja, FCT, Nigeria
| | - Chibuzo Oguoma
- Malaria Consortium, 33 Pope John Paul Street, Maitama, Abuja, FCT, Nigeria
| | - Christian Rassi
- Malaria Consortium, The Green House, 244-254 Cambridge Heath Road, London, E2 9DA, UK
| | - Kevin Baker
- Malaria Consortium, The Green House, 244-254 Cambridge Heath Road, London, E2 9DA, UK
- Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | - Olusola Oresanya
- Malaria Consortium, 33 Pope John Paul Street, Maitama, Abuja, FCT, Nigeria
| | - Chuks Nnaji
- Malaria Consortium, The Green House, 244-254 Cambridge Heath Road, London, E2 9DA, UK
| |
Collapse
|
7
|
Braunack-Mayer L, Malinga J, Masserey T, Nekkab N, Sen S, Schellenberg D, Tchouatieu AM, Kelly SL, Penny MA. Design and selection of drug properties to increase the public health impact of next-generation seasonal malaria chemoprevention: a modelling study. Lancet Glob Health 2024; 12:e478-e490. [PMID: 38365418 PMCID: PMC10882206 DOI: 10.1016/s2214-109x(23)00550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/02/2023] [Accepted: 11/20/2023] [Indexed: 02/18/2024]
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) is recommended for disease control in settings with moderate to high Plasmodium falciparum transmission and currently depends on the administration of sulfadoxine-pyrimethamine plus amodiaquine. However, poor regimen adherence and the increased frequency of parasite mutations conferring sulfadoxine-pyrimethamine resistance might threaten the effectiveness of SMC. Guidance is needed to de-risk the development of drug compounds for malaria prevention. We aimed to provide guidance for the early prioritisation of new and alternative SMC drugs and their target product profiles. METHODS In this modelling study, we combined an individual-based malaria transmission model that has explicit parasite growth with drug pharmacokinetic and pharmacodynamic models. We modelled SMC drug attributes for several possible modes of action, linked to their potential public health impact. Global sensitivity analyses identified trade-offs between drug elimination half-life, maximum parasite killing effect, and SMC coverage, and optimisation identified minimum requirements to maximise malaria burden reductions. FINDINGS Model predictions show that preventing infection for the entire period between SMC cycles is more important than drug curative efficacy for clinical disease effectiveness outcomes, but similarly important for impact on prevalence. When children younger than 5 years receive four SMC cycles with high levels of coverage (ie, 69% of children receiving all cycles), drug candidates require a duration of protection half-life higher than 23 days (elimination half-life >10 days) to achieve reductions higher than 75% in clinical incidence and severe disease (measured over the intervention period in the target population, compared with no intervention across a range of modelled scenarios). High coverage is crucial to achieve these targets, requiring more than 60% of children to receive all SMC cycles and more than 90% of children to receive at least one cycle regardless of the protection duration of the drug. INTERPRETATION Although efficacy is crucial for malaria prevalence reductions, chemoprevention development should select drug candidates for their duration of protection to maximise burden reductions, with the duration half-life determining cycle timing. Explicitly designing or selecting drug properties to increase community uptake is paramount. FUNDING Bill & Melinda Gates Foundation and the Swiss National Science Foundation.
Collapse
Affiliation(s)
- Lydia Braunack-Mayer
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Josephine Malinga
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Thiery Masserey
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Narimane Nekkab
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Swapnoleena Sen
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - David Schellenberg
- Department of Disease Control, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Sherrie L Kelly
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Melissa A Penny
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland; Telethon Kids Institute, Nedlands, WA, Australia; Centre for Child Health Research, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
8
|
Littmann J, Achu D, Laufer MK, Karema C, Schellenberg D. Making the most of malaria chemoprevention. Malar J 2024; 23:51. [PMID: 38369497 PMCID: PMC10875741 DOI: 10.1186/s12936-024-04867-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Against a backdrop of stalled progress in malaria control, it is surprising that the various forms of malaria chemoprevention are not more widely used. The World Health Organization (WHO) has recommended several malaria chemoprevention strategies, some of them for over a decade, and each with documented efficacy and cost effectiveness. In 2022, the WHO updated and augmented its malaria chemoprevention guidelines to facilitate their wider use. This paper considers new insights into the empirical evidence that supports the broader application of chemoprevention and encourages its application as a default strategy for young children living in moderate to high transmission settings given their high risk of severe disease and death. Chemoprevention is an effective medium-term strategy with potential benefits far outweighing costs. There is a strong argument for urgently increasing malaria chemoprevention in endemic countries.
Collapse
Affiliation(s)
- Jasper Littmann
- Bergen Centre for Ethics and Priority Setting-BCEPS, Department of Global Public Health and Primary Care, Faculty of Medicine, University of Bergen, Bergen, Norway.
- Division for Infection Control, The Norwegian Institute for Public Health, Oslo, Norway.
| | - Dorothy Achu
- World Health Organization, Regional Office for Africa, Brazzaville, Republic of Congo
| | - Miriam K Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - David Schellenberg
- Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
9
|
Gatiba P, Laury J, Steinhardt L, Hwang J, Thwing JI, Zulliger R, Emerson C, Gutman JR. Contextual Factors to Improve Implementation of Malaria Chemoprevention in Children: A Systematic Review. Am J Trop Med Hyg 2024; 110:69-78. [PMID: 38081055 PMCID: PMC10793032 DOI: 10.4269/ajtmh.23-0478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/15/2023] [Indexed: 01/05/2024] Open
Abstract
Malaria remains a leading cause of childhood morbidity and mortality in sub-Saharan Africa, particularly among children under 5 years of age. To help address this challenge, the WHO recommends chemoprevention for certain populations. For children and infants, the WHO recommends seasonal malaria chemoprevention (SMC), perennial malaria chemoprevention (PMC; formerly intermittent preventive treatment in infants [IPTi]), and, more recently, intermittent preventive treatment in school children (IPTsc). This review describes the contextual factors, including feasibility, acceptability, health equity, financial considerations, and values and preferences, that impact implementation of these strategies. A systematic search was conducted on July 5, 2022, and repeated April 13, 2023, to identify relevant literature. Two reviewers independently screened titles for eligibility, extracted data from eligible articles, and identified and summarized themes. Of 6,295 unique titles identified, 65 were included. The most frequently evaluated strategy was SMC (n = 40), followed by IPTi (n = 18) and then IPTsc (n = 6). Overall, these strategies were highly acceptable, although with IPTsc, there were community concerns with providing drugs to girls of reproductive age and the use of nonmedical staff for drug distribution. For SMC, door-to-door delivery resulted in higher coverage, improved caregiver acceptance, and reduced cost. Lower adherence was noted when caregivers were charged with giving doses 2 and 3 unsupervised. For SMC and IPTi, travel distances and inclement weather limited accessibility. Sensitization and caregiver education efforts, retention of high-quality drug distributors, and improved transportation were key to improving coverage. Additional research is needed to understand the role of community values and preferences in chemoprevention implementation.
Collapse
Affiliation(s)
- Peris Gatiba
- Public Health Institute, Oakland, California
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica Laury
- Public Health Institute, Oakland, California
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Laura Steinhardt
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jimee Hwang
- U.S. President’s Malaria Initiative, Malaria Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Julie I. Thwing
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Rose Zulliger
- U.S. President’s Malaria Initiative, United States Agency for International Development, Washington, District of Columbia
| | - Courtney Emerson
- U.S. President’s Malaria Initiative, Malaria Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Julie R. Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
10
|
Runge M, Stahlfeld A, Ambrose M, Toh KB, Rahman S, Omoniwa OF, Bever CA, Oresanya O, Uhomoibhi P, Galatas B, Tibenderana JK, Gerardin J. Perennial malaria chemoprevention with and without malaria vaccination to reduce malaria burden in young children: a modelling analysis. Malar J 2023; 22:133. [PMID: 37095480 PMCID: PMC10124689 DOI: 10.1186/s12936-023-04564-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND A recent WHO recommendation for perennial malaria chemoprevention (PMC) encourages countries to adapt dose timing and number to local conditions. However, knowledge gaps on the epidemiological impact of PMC and possible combination with the malaria vaccine RTS,S hinder informed policy decisions in countries where malaria burden in young children remains high. METHODS The EMOD malaria model was used to predict the impact of PMC with and without RTS,S on clinical and severe malaria cases in children under the age of two years (U2). PMC and RTS,S effect sizes were fit to trial data. PMC was simulated with three to seven doses (PMC-3-7) before the age of eighteen months and RTS,S with three doses, shown to be effective at nine months. Simulations were run for transmission intensities of one to 128 infectious bites per person per year, corresponding to incidences of < 1 to 5500 cases per 1000 population U2. Intervention coverage was either set to 80% or based on 2018 household survey data for Southern Nigeria as a sample use case. The protective efficacy (PE) for clinical and severe cases in children U2 was calculated in comparison to no PMC and no RTS,S. RESULTS The projected impact of PMC or RTS,S was greater at moderate to high transmission than at low or very high transmission. Across the simulated transmission levels, PE estimates of PMC-3 at 80% coverage ranged from 5.7 to 8.8% for clinical, and from 6.1 to 13.6% for severe malaria (PE of RTS,S 10-32% and 24.6-27.5% for clinical and severe malaria, respectively. In children U2, PMC with seven doses nearly averted as many cases as RTS,S, while the combination of both was more impactful than either intervention alone. When operational coverage, as seen in Southern Nigeria, increased to a hypothetical target of 80%, cases were reduced beyond the relative increase in coverage. CONCLUSIONS PMC can substantially reduce clinical and severe cases in the first two years of life in areas with high malaria burden and perennial transmission. A better understanding of the malaria risk profile by age in early childhood and on feasible coverage by age, is needed for selecting an appropriate PMC schedule in a given setting.
Collapse
Affiliation(s)
- Manuela Runge
- Department of Preventive Medicine, Institute for Global Health, Northwestern University, Chicago, IL USA
| | - Anne Stahlfeld
- Department of Preventive Medicine, Institute for Global Health, Northwestern University, Chicago, IL USA
| | - Monique Ambrose
- Institute for Disease Modeling, Bill and Melinda Gates Foundation, Seattle, USA
| | - Kok Ben Toh
- Department of Preventive Medicine, Institute for Global Health, Northwestern University, Chicago, IL USA
| | - Semiu Rahman
- Malaria Consortium Nigeria, 33 Pope John Paul Street, Off Gana Street, Maitama, Abuja-FCT Nigeria
| | - Omowunmi F. Omoniwa
- Malaria Consortium Nigeria, 33 Pope John Paul Street, Off Gana Street, Maitama, Abuja-FCT Nigeria
| | - Caitlin A. Bever
- Institute for Disease Modeling, Bill and Melinda Gates Foundation, Seattle, USA
| | - Olusola Oresanya
- Malaria Consortium Nigeria, 33 Pope John Paul Street, Off Gana Street, Maitama, Abuja-FCT Nigeria
| | - Perpetua Uhomoibhi
- National Malaria Elimination Programme, Federal Ministry of Health, Abuja, Nigeria
| | - Beatriz Galatas
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | | | - Jaline Gerardin
- Department of Preventive Medicine, Institute for Global Health, Northwestern University, Chicago, IL USA
| |
Collapse
|
11
|
Cheung YB, Ma X, Lam KF, Yung CF, Milligan P. Modelling non-linear patterns of time-varying intervention effects on recurrent events in infectious disease prevention studies. J Biopharm Stat 2023; 33:220-233. [PMID: 35946934 DOI: 10.1080/10543406.2022.2108826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Protective efficacy of vaccines and pharmaceutical products for prevention of infectious diseases usually vary over time. Information on the trajectory of the level of protection is valuable. We consider a parsimonious, non-linear and non-monotonic function for modelling time-varying intervention effects and compare it with several alternatives. The cumulative effects of multiple doses of intervention over time can be captured by an additive series of the function. We apply it to the Andersen-Gill model for analysis of recurrent time-to-event data. We re-analyze data from a trial of intermittent preventive treatment for malaria to illustrate and evaluate the method by simulation.
Collapse
Affiliation(s)
- Yin Bun Cheung
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore.,Programme in Health Services & Systems Research, Duke-NUS Medical School, Singapore.,Tampere Center for Child, Adolescent and Maternal Health Research, Tampere University, Tampere, Finland
| | - Xiangmei Ma
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - K F Lam
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore.,Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong, Pok Fu Lam, China
| | - Chee Fu Yung
- Infectious Disease Service, KK Women's and Children's Hospital, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Academic Medicine Department, Duke-NUS Medical School, Singapore
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
12
|
Cairns M, Barry A, Zongo I, Sagara I, Yerbanga SR, Diarra M, Zoungrana C, Issiaka D, Sienou AA, Tapily A, Sanogo K, Kaya M, Traore S, Diarra K, Yalcouye H, Sidibe Y, Haro A, Thera I, Snell P, Grant J, Tinto H, Milligan P, Chandramohan D, Greenwood B, Dicko A, Ouedraogo JB. The duration of protection against clinical malaria provided by the combination of seasonal RTS,S/AS01 E vaccination and seasonal malaria chemoprevention versus either intervention given alone. BMC Med 2022; 20:352. [PMID: 36203149 PMCID: PMC9540742 DOI: 10.1186/s12916-022-02536-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A recent trial of 5920 children in Burkina Faso and Mali showed that the combination of seasonal vaccination with the RTS,S/AS01E malaria vaccine (primary series and two seasonal boosters) and seasonal malaria chemoprevention (four monthly cycles per year) was markedly more effective than either intervention given alone in preventing clinical malaria, severe malaria, and deaths from malaria. METHODS In order to help optimise the timing of these two interventions, trial data were reanalysed to estimate the duration of protection against clinical malaria provided by RTS,S/AS01E when deployed seasonally, by comparing the group who received the combination of SMC and RTS,S/AS01E with the group who received SMC alone. The duration of protection from SMC was also estimated comparing the combined intervention group with the group who received RTS,S/AS01E alone. Three methods were used: Piecewise Cox regression, Flexible parametric survival models and Smoothed Schoenfeld residuals from Cox models, stratifying on the study area and using robust standard errors to control for within-child clustering of multiple episodes. RESULTS The overall protective efficacy from RTS,S/AS01E over 6 months was at least 60% following the primary series and the two seasonal booster doses and remained at a high level over the full malaria transmission season. Beyond 6 months, protective efficacy appeared to wane more rapidly, but the uncertainty around the estimates increases due to the lower number of cases during this period (coinciding with the onset of the dry season). Protection from SMC exceeded 90% in the first 2-3 weeks post-administration after several cycles, but was not 100%, even immediately post-administration. Efficacy begins to decline from approximately day 21 and then declines more sharply after day 28, indicating the importance of preserving the delivery interval for SMC cycles at a maximum of four weeks. CONCLUSIONS The efficacy of both interventions was highest immediately post-administration. Understanding differences between these interventions in their peak efficacy and how rapidly efficacy declines over time will help to optimise the scheduling of SMC, malaria vaccination and the combination in areas of seasonal transmission with differing epidemiology, and using different vaccine delivery systems. TRIAL REGISTRATION The RTS,S-SMC trial in which these data were collected was registered at clinicaltrials.gov: NCT03143218.
Collapse
Affiliation(s)
- Matthew Cairns
- International Statistics and Epidemiology Group, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Amadou Barry
- Malaria Research and Training Centre, Bamako, Mali
| | - Issaka Zongo
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | | | - Serge R Yerbanga
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | | | - Charles Zoungrana
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | | | - Abdoul Aziz Sienou
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | | | | | | | | | | | | | | | - Alassane Haro
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | | | - Paul Snell
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Jane Grant
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Bobo Dioulasso, Burkina Faso
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Daniel Chandramohan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | | | | |
Collapse
|
13
|
Phiri MD, Cairns M, Zongo I, Nikiema F, Diarra M, Yerbanga RS, Barry A, Tapily A, Coumare S, Thera I, Kuepfer I, Milligan P, Tinto H, Dicko A, Ouédraogo JB, Greenwood B, Chandramohan D, Sagara I. The Duration of Protection from Azithromycin Against Malaria, Acute Respiratory, Gastrointestinal, and Skin Infections When Given Alongside Seasonal Malaria Chemoprevention: Secondary Analyses of Data from a Clinical Trial in Houndé, Burkina Faso, and Bougouni, Mali. Clin Infect Dis 2021; 73:e2379-e2386. [PMID: 33417683 PMCID: PMC8492219 DOI: 10.1093/cid/ciaa1905] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Mass drug administration (MDA) with azithromycin (AZ) is being considered as a strategy to promote child survival in sub-Saharan Africa, but the mechanism by which AZ reduces mortality is unclear. To better understand the nature and extent of protection provided by AZ, we explored the profile of protection by time since administration, using data from a household-randomized, placebo-controlled trial in Burkina Faso and Mali. METHODS Between 2014 and 2016, 30 977 children aged 3-59 months received seasonal malaria chemoprevention (SMC) with sulfadoxine-pyrimethamine plus amodiaquine and either AZ or placebo monthly, on 4 occasions each year. Poisson regression with gamma-distributed random effects, accounting for the household randomization and within-individual clustering of illness episodes, was used to compare incidence of prespecified outcomes between SMC+AZ versus SMC+placebo groups in fixed time strata post-treatment. The likelihood ratio test was used to assess evidence for a time-treatment group interaction. RESULTS Relative to SMC+placebo, there was no evidence of protection from SMC+AZ against hospital admissions and deaths. Additional protection from SMC+AZ against malaria was confined to the first 2 weeks post-administration (protective efficacy (PE): 24.2% [95% CI: 17.8%, 30.1%]). Gastroenteritis and pneumonia were reduced by 29.9% [21.7; 37.3%], and 34.3% [14.9; 49.3%], respectively, in the first 2 weeks postadministration. Protection against nonmalaria fevers with a skin condition persisted up to 28 days: PE: 46.3% [35.1; 55.6%]. CONCLUSIONS The benefits of AZ-MDA are broad-ranging but short-lived. To maximize impact, timing of AZ-MDA must address the challenge of targeting asynchronous morbidity and mortality peaks from different causes.
Collapse
Affiliation(s)
- Mphatso Dennis Phiri
- Malaria Epidemiology Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Matthew Cairns
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Issaka Zongo
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Frederic Nikiema
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Modibo Diarra
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Rakiswendé Serge Yerbanga
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Amadou Barry
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Amadou Tapily
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Samba Coumare
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Ismaila Thera
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Irene Kuepfer
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Paul Milligan
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Halidou Tinto
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Alassane Dicko
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Jean Bosco Ouédraogo
- Le Département Biomédical et de Santé Publique, Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | - Brian Greenwood
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Daniel Chandramohan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Issaka Sagara
- Malaria Research and Training Center, University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| |
Collapse
|
14
|
Impact of Three-Year Intermittent Preventive Treatment Using Artemisinin-Based Combination Therapies on Malaria Morbidity in Malian Schoolchildren. Trop Med Infect Dis 2020; 5:tropicalmed5030148. [PMID: 32957604 PMCID: PMC7560034 DOI: 10.3390/tropicalmed5030148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 11/28/2022] Open
Abstract
Previous studies have shown that a single season of intermittent preventive treatment in schoolchildren (IPTsc) targeting the transmission season has reduced the rates of clinical malaria, all-cause clinic visits, asymptomatic parasitemia, and anemia. Efficacy over the course of multiple years of IPTsc has been scantly investigated. Methods: An open, randomized-controlled trial among schoolchildren aged 6–13 years was conducted from September 2007 to January 2010 in Kolle, Mali. Students were included in three arms: sulphadoxine-pyrimethamine+artesunate (SP+AS), amodiaquine+artesunate (AQ+AS), and control (C). All students received two full doses, given 2 months apart, and were compared with respect to the incidence of clinical malaria, all-cause clinic visits, asymptomatic parasitemia, and anemia. Results: A total of 296 students were randomized. All-cause clinic visits were in the SP+AS versus control (29 (20.1%) vs. 68 (47.2%); 20 (21.7%) vs. 41 (44.6%); and 14 (21.2%) vs. 30 (44.6%); p < 0.02) in 2007, 2008, and 2009, respectively. The prevalence of asymptomatic parasitemia was lower in the SP+AS compared to control (38 (7.5%) vs. 143 (28.7%); and 47 (12.7%) vs. 75 (21.2%); p < 0.002) in 2007 and 2008, respectively. Hemoglobin concentration was significantly higher in children receiving SP+AS (11.96, 12.06, and 12.62 g/dL) than in control children (11.60, 11.64, and 12.15 g/dL; p < 0.001) in 2007, 2008, and 2009, respectively. No impact on clinical malaria was observed. Conclusion: IPTsc with SP+AS reduced the rates of all-cause clinic visits and anemia during a three-year implementation.
Collapse
|
15
|
Wangdi K, Furuya-Kanamori L, Clark J, Barendregt JJ, Gatton ML, Banwell C, Kelly GC, Doi SAR, Clements ACA. Comparative effectiveness of malaria prevention measures: a systematic review and network meta-analysis. Parasit Vectors 2018; 11:210. [PMID: 29587882 PMCID: PMC5869791 DOI: 10.1186/s13071-018-2783-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/06/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Malaria causes significant morbidity and mortality worldwide. There are several preventive measures that are currently employed, including insecticide-treated nets (ITNs, including long-lasting insecticidal nets and insecticidal-treated bed nets), indoor residual spraying (IRS), prophylactic drugs (PD), and untreated nets (UN). However, it is unclear which measure is the most effective for malaria prevention. We therefore undertook a network meta-analysis to compare the efficacy of different preventive measures on incidence of malaria infection. METHODS A systematic literature review was undertaken across four medical and life sciences databases (PubMed, Cochrane Central, Embase, and Web of Science) from their inception to July 2016 to compare the effectiveness of different preventive measures on malaria incidence. Data from the included studies were analysed for the effectiveness of several measures against no intervention (NI). This was carried out using an automated generalized pairwise modeling (GPM) framework for network meta-analysis to generate mixed treatment effects against a common comparator of no intervention (NI). RESULTS There were 30 studies that met the inclusion criteria from 1998-2016. The GPM framework led to a final ranking of effectiveness of measures in the following order from best to worst: PD, ITN, IRS and UN, in comparison with NI. However, only ITN (RR: 0.49, 95% CI: 0.32-0.74) showed precision while other methods [PD (RR: 0.24, 95% CI: 0.004-15.43), IRS (RR: 0.55, 95% CI: 0.20-1.56) and UN (RR: 0.73, 95% CI: 0.28-1.90)] demonstrating considerable uncertainty associated with their point estimates. CONCLUSION Current evidence is strong for the protective effect of ITN interventions in malaria prevention. Even though ITNs were found to be the only preventive measure with statistical support for their effectiveness, the role of other malaria control measures may be important adjuncts in the global drive to eliminate malaria.
Collapse
Affiliation(s)
- Kinley Wangdi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Luis Furuya-Kanamori
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Justin Clark
- Centre for Research in Evidence-Based Practice (CREBP), Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Queensland Australia
| | - Jan J. Barendregt
- School of Public Health, The University of Queensland, Brisbane, Queensland Australia
- Epigear International Pty Ltd, Sunrise Beach, Queensland Australia
| | - Michelle L. Gatton
- School of Public Health & Social Work, Queensland University of Technology, Brisbane, Queensland Australia
| | - Cathy Banwell
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Gerard C. Kelly
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| | - Suhail A. R. Doi
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
- Department of Population Medicine, College of Medicine, Qatar University, Doha, Qatar
| | - Archie C. A. Clements
- Research School of Population Health, College of Health and Medicine, The Australian National University, ACT, Canberra, Australia
| |
Collapse
|
16
|
Estimated impact on birth weight of scaling up intermittent preventive treatment of malaria in pregnancy given sulphadoxine-pyrimethamine resistance in Africa: A mathematical model. PLoS Med 2017; 14:e1002243. [PMID: 28245259 PMCID: PMC5330448 DOI: 10.1371/journal.pmed.1002243] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/23/2017] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Malaria transmission has declined substantially in the 21st century, but pregnant women in areas of sustained transmission still require protection to prevent the adverse pregnancy and birth outcomes associated with malaria in pregnancy (MiP). A recent call to action has been issued to address the continuing low coverage of intermittent preventive treatment of malaria in pregnancy (IPTp). This call has, however, been questioned by some, in part due to concerns about resistance to sulphadoxine-pyrimethamine (SP), the only drug currently recommended for IPTp. METHODS AND FINDINGS Using an existing mathematical model of MiP, we combined estimates of the changing endemicity of malaria across Africa with maps of SP resistance mutations and current coverage of antenatal access and IPTp with SP (IPTp-SP) across Africa. Using estimates of the relationship between SP resistance mutations and the parasitological efficacy of SP during pregnancy, we estimated the varying impact of IPTp-SP across Africa and the incremental value of enhancing IPTp-SP uptake to match current antenatal care (ANC) coverage. The risks of MiP and malaria-attributable low birthweight (mLBW) in unprotected pregnancies (i.e., those not using insecticide-treated nets [ITNs]) leading to live births fell by 37% (33%-41% 95% credible interval [crI]) and 31% (27%-34% 95% crI), respectively, from 2000 to 2015 across endemic areas in sub-Saharan Africa. However, these gains are fragile, and coverage is far from optimal. In 2015, 9.5 million (8.3 million-10.4 million 95% crI) of 30.6 million pregnancies in these areas would still have been infected with Plasmodium falciparum without intervention, leading to 750,000 (390,000-1.1 million 95% crI) mLBW deliveries. In all, 6.6 million (5.6 million-7.3 million 95% crI) of these 9.5 million (69.3%) pregnancies at risk of infection (and 53.4% [16.3 million/30.6 million] of all pregnancies) occurred in settings with near-perfect SP curative efficacy (>99%) based on the most recent estimates of resistance. Forty-four percent of these pregnancies (23% of all pregnancies) were not receiving any IPTp-SP despite making ≥3 ANC visits, representing 160,000 (94,000-236,000 95% crI) preventable low birthweight (LBW) deliveries. Only 4% (1.4 million) of pregnancies occurred in settings with >10% prevalence of the sextuple haplotype associated with compromised SP effectiveness. Forty-two percent of all pregnancies occurred in settings where the quintuple dhfr/dhps haplotype had become established but where in vivo efficacy data suggest SP maintains the majority of its effectiveness in clearing infections. Not accounting for protection from the use of ITNs during pregnancy, expanding IPTp-SP to all women with ≥3 ANC visits in Africa could prevent an additional 215,000 (128,000-318,000 95% crI) LBW deliveries. In 26 countries with sufficient recent data to estimate ITN impact (population-based ITN usage data that can be stratified by gravidity), we estimate that, due primarily to low ITN use by primigravidae, only 16.5% of the potential LBW births prevented by scaling up IPTp-SP would in fact have already have been prevented through ITN use. Our analysis also highlights the difficulties associated with estimating the relationship between the effectiveness of interventions against parasitological endpoints such as placental infection at delivery and health outcomes including birthweight, which is also determined by a wide range of unrelated factors. We also did not capture other aspects of malaria burden such as clinical malaria, maternal and neonatal anaemia, and miscarriage, all of which increase the overall importance of effective preventative strategies but have their own relationship with transmission intensity, parity, and SP resistance. CONCLUSIONS Despite recent declines in malaria transmission in Africa, the burden of MiP in the absence of adequate prevention remains substantial. Even accounting for SP resistance, extending IPTp-SP to all women attending ANC, as well as long-lasting insecticidal net distribution targeted towards first-time mothers, would have a sizeable impact upon maternal and infant health in almost all malaria-endemic settings in sub-Saharan Africa.
Collapse
|
17
|
Walker PGT, Griffin JT, Ferguson NM, Ghani AC. Estimating the most efficient allocation of interventions to achieve reductions in Plasmodium falciparum malaria burden and transmission in Africa: a modelling study. LANCET GLOBAL HEALTH 2016; 4:e474-84. [PMID: 27269393 DOI: 10.1016/s2214-109x(16)30073-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/04/2016] [Accepted: 04/21/2016] [Indexed: 11/26/2022]
Abstract
BACKGROUND Reducing the burden of malaria is a global priority, but financial constraints mean that available resources must be allocated rationally to maximise their effect. We aimed to develop a model to estimate the most efficient (ie, minimum cost) ordering of interventions to reduce malaria burden and transmission. We also aimed to estimate the efficiency of different spatial scales of implementation. METHODS We combined a dynamic model capturing heterogeneity in malaria transmission across Africa with financial unit cost data for key malaria interventions. We combined estimates of patterns of malaria endemicity, seasonality in rainfall, and mosquito composition to map optimum packages of these interventions across Africa. Using non-linear optimisation methods, we examined how these optimum packages vary when control measures are deployed and assessed at national, subnational first administrative (provincial), or fine-scale (5 km(2) pixel) spatial scales. FINDINGS The most efficient package in a given setting varies depending on whether disease reduction or elimination is the target. Long-lasting insecticide-treated nets are generally the most cost-effective first intervention to achieve either goal, with seasonal malaria chemoprevention or indoor residual spraying added second depending on seasonality and vector species. These interventions are estimated to reduce malaria transmission to less than one case per 1000 people per year in 43·4% (95% CI 40·0-49·0) of the population at risk in Africa. Adding three rounds of mass drug administration per year is estimated to increase this proportion to 90·9% (95% CI 86·9-94·6). Further optimisation can be achieved by targeting policies at the provincial level, achieving an estimated 32·1% (95% CI 29·6-34·5) cost saving relative to adopting country-wide policies. Nevertheless, we predict that only 26 (95% CI 22-29) of 41 countries could reduce transmission to these levels with these approaches. INTERPRETATION These results highlight the cost-benefits of carefully tailoring malaria interventions to the ecological landscape of different areas. However, novel interventions are necessary if malaria eradication is to be achieved. FUNDING Bill & Melinda Gates Foundation, UK Medical Research Council.
Collapse
Affiliation(s)
- Patrick G T Walker
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| | - Jamie T Griffin
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Neil M Ferguson
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Azra C Ghani
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| |
Collapse
|
18
|
Hamma M. Impact of seasonal malaria chemoprevention of sulphadoxinepyrimethamine plus amodiaquine on molecular markers resistance of Plasmodium falciparum malaria: A review in West Africa. ACTA ACUST UNITED AC 2016. [DOI: 10.5897/cro15.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
19
|
Xu Y, Lam KF, Cheung YB. Estimation of intervention effects using recurrent event time data in the presence of event dependence and a cured fraction. Stat Med 2014; 33:2263-74. [DOI: 10.1002/sim.6093] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 12/17/2013] [Accepted: 12/26/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Ying Xu
- Centre for Quantitative Medicine, Office of Clinical Sciences; Duke-NUS Graduate Medical School; Singapore
- Scientific Development Division; Singapore Clinical Research Institute; Singapore
| | - K. F. Lam
- Department of Statistics and Actuarial Science; The University of Hong Kong; Hong Kong
| | - Yin Bun Cheung
- Centre for Quantitative Medicine, Office of Clinical Sciences; Duke-NUS Graduate Medical School; Singapore
- Scientific Development Division; Singapore Clinical Research Institute; Singapore
- Department of International Health; University of Tampere; Tampere Finland
| |
Collapse
|
20
|
Gosling RD, Cairns ME, Chico RM, Chandramohan D. Intermittent preventive treatment against malaria: an update. Expert Rev Anti Infect Ther 2014; 8:589-606. [DOI: 10.1586/eri.10.36] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Abstract
Childhood anemia has major adverse consequences for health and development. It's prevalence in India continues to range from 70 to 90%. Although anemia is multifactorial in etiology, preventative efforts have predominantly focused on increasing iron intake, primarily through supplementation in pregnant and lactating women. Policy thrust for childhood anemia is only recent. However, program implementation is dismal; only 3.8-4.7% of preschoolers receive iron-folate supplements. There is an urgent need for effective governance and implementation. Policy makers must distinguish anemia from iron deficiency, and introduce additional area-specific interventions as an integrated package.Increased iron intake may yield maximum benefit but will only address up to half the burden. In 6-59 months old children, instead of 100 days' continuous dosing with iron-folate syrup in a year, a directly supervised intermittent supplementation (biweekly; ~100 days per year) merits consideration. Multiple micronutrient powders for home fortification of foods in 6-23 months old infants do not appear viable. Additional interventions include delayed cord clamping, earlier supplementation in low birth weight infants, appropriate infant and young child feeding guidelines, and intermittent supervised supplementation in children and adolescents through school health programs. Use of double (iron-folate)-fortified salt in mid-day meal programs deserves piloting.Important area-specific, non-iron interventions include targeted deworming, and prevention and treatment of hemoglobinopathies, malaria and other common infections. Routine addition of multi-micronutrients to iron-folate supplementation appears unjustified currently. There is a pressing need to conduct relevant research, especially to inform etiology, additional interventions and implementation issues.
Collapse
|
22
|
Bardají A, Bassat Q, Alonso PL, Menéndez C. Intermittent preventive treatment of malaria in pregnant women and infants: making best use of the available evidence. Expert Opin Pharmacother 2012; 13:1719-36. [PMID: 22775553 DOI: 10.1517/14656566.2012.703651] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Malaria continues to represent a huge global health burden on the most vulnerable populations. The Intermittent Preventive Treatment (IPT) strategy has been shown to be an efficacious intervention in preventing most of the deleterious effects of malaria in pregnant women and infants. Yet, the effectiveness of the IPT strategy may be impaired by the increasing resistance to sulfadoxine-pyrimethamine (SP), and the scarcity of alternative antimalarial drugs. AREAS COVERED This review examines all the available information on IPT, in an aim to provide the scientific community with a framework to understand the benefits and limitations of this malaria control strategy. It includes the understanding of the historical background of the IPT strategy, the drug's mechanisms of actions, updated information on current available evidence, the implications of drug resistance and choice of alternative drugs, and a comprehensive discussion on the perspectives of IPT for malaria control in pregnant women and infants. EXPERT OPINION IPT in pregnancy and infants is a cost-effective strategy that can contribute significantly to the control of malaria in endemic areas. Monitoring its effectiveness will allow tracking of progress, evaluation of the adequacy of currently used drugs and will highlight the eventual need for new therapies or alternative interventions.
Collapse
Affiliation(s)
- Azucena Bardají
- University of Barcelona, Hospital Clínic, Barcelona Centre for International Health Research, Roselló, 132, 08036, Barcelona, Spain.
| | | | | | | |
Collapse
|
23
|
Bijker EM, Sauerwein RW. Enhancement of naturally acquired immunity against malaria by drug use. J Med Microbiol 2012; 61:904-910. [PMID: 22282458 DOI: 10.1099/jmm.0.041277-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Combination of chemoprophylaxis with chloroquine and so-called 'controlled human malaria infections' has been shown to induce sustained and fully protective immunity against malaria in experimental settings. This opens possibilities of translating this approach into an effective and applicable strategy for the field. We review the different ways in which antimalarial drugs have been used for prevention of malaria in endemic settings and discuss the possibilities and challenges of applying a strategy of drug use and naturally acquired infection in the field.
Collapse
Affiliation(s)
- Else M Bijker
- Radboud University Nijmegen Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| | - Robert W Sauerwein
- Radboud University Nijmegen Medical Center, Department of Medical Microbiology, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Gosling R, Okell L, Mosha J, Chandramohan D. The role of antimalarial treatment in the elimination of malaria. Clin Microbiol Infect 2011; 17:1617-23. [DOI: 10.1111/j.1469-0691.2011.03660.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Borrmann S, Matuschewski K. Targeting Plasmodium liver stages: better late than never. Trends Mol Med 2011; 17:527-36. [DOI: 10.1016/j.molmed.2011.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2011] [Revised: 05/08/2011] [Accepted: 05/20/2011] [Indexed: 10/18/2022]
|
26
|
Anaemia in Ugandan preschool-aged children: the relative contribution of intestinal parasites and malaria. Parasitology 2011; 138:1534-45. [PMID: 21819635 DOI: 10.1017/s0031182011001016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Anaemia is a severe public health issue among African preschool-aged children, yet little effective progress has been made towards its amelioration, in part due to difficulties in unravelling its complex, multifactorial aetiology. To determine the current anaemia situation and assess the relative contribution of malaria, intestinal schistosomiasis and infection with soil-transmitted helminths, two separate cross-sectional epidemiological surveys were carried out in Uganda including 573 and 455 preschool-aged children (≤6 years) living along the shores of Lake Albert and on the islands in Lake Victoria, respectively. Anaemia was found to be a severe public health problem in Lake Albert, affecting 68·9% of children (ninety-five percent confidence intervals (95% CI) 64·9-72·7%), a statistically significant higher prevalence relative to the 27·3% detected in Lake Victoria (95% CI: 23·3-31·7%). After multivariate analysis (controlling for sex and age of the child), the only factor found to be significantly associated with increased odds of anaemia in both lake systems was malaria (Lake Albert, odds ratio (OR)=2·1, 95% CI: 1·4-3·2; Lake Victoria, OR=1·9, 95% CI: 1·2-2·9). Thus intervention strategies primarily focusing on very young children and combating malaria appear to represent the most appropriate use of human and financial resources for the prevention of anaemia in this age group and area. Looking to the future, these activities could be further emphasised within the National Child Health Days(PLUS) agenda.
Collapse
|
27
|
Okell LC, Griffin JT, Kleinschmidt I, Hollingsworth TD, Churcher TS, White MJ, Bousema T, Drakeley CJ, Ghani AC. The potential contribution of mass treatment to the control of Plasmodium falciparum malaria. PLoS One 2011; 6:e20179. [PMID: 21629651 PMCID: PMC3101232 DOI: 10.1371/journal.pone.0020179] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 04/27/2011] [Indexed: 11/19/2022] Open
Abstract
Mass treatment as a means to reducing P. falciparum malaria transmission was used during the first global malaria eradication campaign and is increasingly being considered for current control programmes. We used a previously developed mathematical transmission model to explore both the short and long-term impact of possible mass treatment strategies in different scenarios of endemic transmission. Mass treatment is predicted to provide a longer-term benefit in areas with lower malaria transmission, with reduced transmission levels for at least 2 years after mass treatment is ended in a scenario where the baseline slide-prevalence is 5%, compared to less than one year in a scenario with baseline slide-prevalence at 50%. However, repeated annual mass treatment at 80% coverage could achieve around 25% reduction in infectious bites in moderate-to-high transmission settings if sustained. Using vector control could reduce transmission to levels at which mass treatment has a longer-term impact. In a limited number of settings (which have isolated transmission in small populations of 1000-10,000 with low-to-medium levels of baseline transmission) we find that five closely spaced rounds of mass treatment combined with vector control could make at least temporary elimination a feasible goal. We also estimate the effects of using gametocytocidal treatments such as primaquine and of restricting treatment to parasite-positive individuals. In conclusion, mass treatment needs to be repeated or combined with other interventions for long-term impact in many endemic settings. The benefits of mass treatment need to be carefully weighed against the risks of increasing drug selection pressure.
Collapse
Affiliation(s)
- Lucy C Okell
- Department of Infectious Disease Epidemiology, MRC Centre for Outbreak Analysis and Modeling, Imperial College London, London, United Kingdom.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cairns M, Ghani A, Okell L, Gosling R, Carneiro I, Anto F, Asoala V, Owusu-Agyei S, Greenwood B, Chandramohan D, Milligan P. Modelling the protective efficacy of alternative delivery schedules for intermittent preventive treatment of malaria in infants and children. PLoS One 2011; 6:e18947. [PMID: 21533088 PMCID: PMC3080380 DOI: 10.1371/journal.pone.0018947] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2010] [Accepted: 03/24/2011] [Indexed: 11/18/2022] Open
Abstract
Background Intermittent preventive treatment in infants (IPTi) with sulfadoxine-pyrimethamine (SP) is recommended by WHO where malaria incidence in infancy is high and SP resistance is low. The current delivery strategy is via routine Expanded Program on Immunisation contacts during infancy (EPI-IPTi). However, improvements to this approach may be possible where malaria transmission is seasonal, or where the malaria burden lies mainly outside infancy. Methods and Findings A mathematical model was developed to estimate the protective efficacy (PE) of IPT against clinical malaria in children aged 2-24 months, using entomological and epidemiological data from an EPI-IPTi trial in Navrongo, Ghana to parameterise the model. The protection achieved by seasonally-targeted IPT in infants (sIPTi), seasonal IPT in children (sIPTc), and by case-management with long-acting artemisinin combination therapies (LA-ACTs) was predicted for Navrongo and for sites with different transmission intensity and seasonality. In Navrongo, the predicted PE of sIPTi was 26% by 24 months of age, compared to 16% with EPI-IPTi. sIPTc given to all children under 2 years would provide PE of 52% by 24 months of age. Seasonally-targeted IPT retained its advantages in a range of transmission patterns. Under certain circumstances, LA-ACTs for case-management may provide similar protection to EPI-IPTi. However, EPI-IPTi or sIPT combined with LA-ACTs would be substantially more protective than either strategy used alone. Conclusion Delivery of IPT to infants via the EPI is sub-optimal because individuals are not protected by IPT at the time of highest malaria risk, and because older children are not protected. Alternative delivery strategies to the EPI are needed where transmission varies seasonally or the malaria burden extends beyond infancy. Long-acting ACTs may also make important reductions in malaria incidence. However, delivery systems must be developed to ensure that both forms of chemoprevention reach the individuals who are most exposed to malaria.
Collapse
Affiliation(s)
- Matthew Cairns
- London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wilson AL, on behalf of the IPTc Taskforce. A systematic review and meta-analysis of the efficacy and safety of intermittent preventive treatment of malaria in children (IPTc). PLoS One 2011; 6:e16976. [PMID: 21340029 PMCID: PMC3038871 DOI: 10.1371/journal.pone.0016976] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 01/12/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment of malaria in children less than five years of age (IPTc) has been investigated as a measure to control the burden of malaria in the Sahel and sub-Sahelian areas of Africa where malaria transmission is markedly seasonal. METHODS AND FINDINGS IPTc studies were identified using a systematic literature search. Meta-analysis was used to assess the protective efficacy of IPTc against clinical episodes of falciparum malaria. The impact of IPTc on all-cause mortality, hospital admissions, severe malaria and the prevalence of parasitaemia and anaemia was investigated. Three aspects of safety were also assessed: adverse reactions to study drugs, development of drug resistance and loss of immunity to malaria. Twelve IPTc studies were identified: seven controlled and five non-controlled trials. Controlled studies demonstrated protective efficacies against clinical malaria of between 31% and 93% and meta-analysis gave an overall protective efficacy of monthly administered IPTc of 82% (95%CI 75%-87%) during the malaria transmission season. Pooling results from twelve studies demonstrated a protective effect of IPTc against all-cause mortality of 57% (95%CI 24%-76%) during the malaria transmission season. No serious adverse events attributable to the drugs used for IPTc were observed in any of the studies. Data from three studies that followed children during the malaria transmission season in the year following IPTc administration showed evidence of a slight increase in the incidence of clinical malaria compared to children who had not received IPTc. CONCLUSIONS IPTc is a safe method of malaria control that has the potential to avert a significant proportion of clinical malaria episodes in areas with markedly seasonal malaria transmission and also appears to have a substantial protective effect against all-cause mortality. These findings indicate that IPTc is a potentially valuable tool that can contribute to the control of malaria in areas with markedly seasonal transmission.
Collapse
Affiliation(s)
- Anne L. Wilson
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | |
Collapse
|
30
|
Intermittent preventive treatment of malaria provides substantial protection against malaria in children already protected by an insecticide-treated bednet in Mali: a randomised, double-blind, placebo-controlled trial. PLoS Med 2011; 8:e1000407. [PMID: 21304923 PMCID: PMC3032550 DOI: 10.1371/journal.pmed.1000407] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 12/16/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Previous studies have shown that in areas of seasonal malaria transmission, intermittent preventive treatment of malaria in children (IPTc), targeting the transmission season, reduces the incidence of clinical malaria. However, these studies were conducted in communities with low coverage with insecticide-treated nets (ITNs). Whether IPTc provides additional protection to children sleeping under an ITN has not been established. METHODS AND FINDINGS To assess whether IPTc provides additional protection to children sleeping under an ITN, we conducted a randomised, double-blind, placebo-controlled trial of IPTc with sulphadoxine pyrimethamine (SP) plus amodiaquine (AQ) in three localities in Kati, Mali. After screening, eligible children aged 3-59 mo were given a long-lasting insecticide-treated net (LLIN) and randomised to receive three rounds of active drugs or placebos. Treatments were administered under observation at monthly intervals during the high malaria transmission season in August, September, and October 2008. Adverse events were monitored immediately after the administration of each course of IPTc and throughout the follow-up period. The primary endpoint was clinical episodes of malaria recorded through passive surveillance by study clinicians available at all times during the follow-up. Cross-sectional surveys were conducted in 150 randomly selected children weekly and in all children at the end of the malaria transmission season to assess usage of ITNs and the impact of IPTc on the prevalence of malaria, anaemia, and malnutrition. Cox regression was used to compare incidence rates between intervention and control arms. The effects of IPTc on the prevalence of malaria infection and anaemia were estimated using logistic regression. 3,065 children were screened and 3,017 (1,508 in the control and 1,509 in the intervention arm) were enrolled in the study. 1,485 children (98.5%) in the control arm and 1,481 (98.1%) in the intervention arm completed follow-up. During the intervention period, the proportion of children reported to have slept under an ITN was 99.7% in the control and 99.3% in intervention arm (p = 0.45). A total of 672 episodes of clinical malaria defined as fever or a history of fever and the presence of at least 5,000 asexual forms of Plasmodium falciparum per microlitre (incidence rate of 1.90; 95% confidence interval [CI] 1.76-2.05 episodes per person year) were observed in the control arm versus 126 (incidence rate of 0.34; 95% CI 0.29-0.41 episodes per person year) in the intervention arm, indicating a protective effect (PE) of 82% (95% CI 78%-85%) (p<0.001) on the primary endpoint. There were 15 episodes of severe malaria in children in the control arm compared to two in children in the intervention group giving a PE of 87% (95% CI 42%-99%) (p = 0.001). IPTc reduced the prevalence of malaria infection by 85% (95% CI 73%-92%) (p<0.001) during the intervention period and by 46% (95% CI 31%-68%) (p<0.001) at the end of the intervention period. The prevalence of moderate anaemia (haemoglobin [Hb] <8 g/dl) was reduced by 47% (95% CI 15%-67%) (p<0.007) at the end of intervention period. The frequencies of adverse events were similar between the two arms. There was no drug-related serious adverse event. CONCLUSIONS IPTc given during the malaria transmission season provided substantial protection against clinical episodes of malaria, malaria infection, and anaemia in children using an LLIN. SP+AQ was safe and well tolerated. These findings indicate that IPTc could make a valuable contribution to malaria control in areas of seasonal malaria transmission alongside other interventions. TRIAL REGISTRATION ClinicalTrials.gov NCT00738946. Please see later in the article for the Editors' Summary.
Collapse
|
31
|
Pharmacokinetic properties of conventional and double-dose sulfadoxine-pyrimethamine given as intermittent preventive treatment in infancy. Antimicrob Agents Chemother 2011; 55:1693-700. [PMID: 21282434 DOI: 10.1128/aac.01075-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Intermittent preventive treatment in infancy (IPTi) entails routine administration of antimalarial treatment doses at specified times in at-risk infants. Sulfadoxine-pyrimethamine (SDX/PYR) is a combination that has been used as first-line IPTi. Because of limited pharmacokinetic data and suggestions that higher milligram/kilogram pediatric doses than recommended should be considered, we assessed SDX/PYR disposition, randomized to conventional (25/1.25 mg/kg of body weight) or double (50/2.5 mg/kg) dose, in 70 Papua New Guinean children aged 2 to 13 months. Blood samples were drawn at baseline, 28 days, and three time points randomly selected for each infant at 4 to 8 h or 2, 5, 7, 14, or 21 days. Plasma SDX, PYR, and N(4)-acetylsulfadoxine (NSX, the principal metabolite of SDX) were assayed by high-performance liquid chromatography (HPLC). Using population modeling incorporating hepatic maturation and cystatin C-based renal function, two-compartment models provided best fits for PYR and SDX/NSX plasma concentration profiles. The area under the plasma concentration-time curve from 0 h to infinity (AUC(0-∞)) was greater with the double dose versus the conventional dose of PYR (4,915 versus 2,844 μg/day/liter) and SDX (2,434 versus 1,460 mg/day/liter). There was a 32% reduction in SDX relative bioavailability with the double dose but no evidence of dose-dependent metabolism. Terminal elimination half-lives (15.6 days for PYR, 9.1 days for SDX) were longer than previously reported. Both doses were well tolerated without changes in hemoglobin or hepatorenal function. Five children in the conventional and three in the double-dose group developed malaria during follow-up. These data support the potential use of double-dose SDX/PYR in infancy, but further studies should examine the influence of hepatorenal maturation in very young infants.
Collapse
|
32
|
Boulanger D, Sarr JB, Fillol F, Sokhna C, Cisse B, Schacht AM, Trape JF, Riveau G, Simondon F, Greenwood B, Remoué F. Immunological consequences of intermittent preventive treatment against malaria in Senegalese preschool children. Malar J 2010; 9:363. [PMID: 21167018 PMCID: PMC3012051 DOI: 10.1186/1475-2875-9-363] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 12/17/2010] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Intermittent preventive treatment in children (IPTc) is a promising strategy to control malaria morbidity. A significant concern is whether IPTc increases children's susceptibility to subsequent malaria infection by altering their anti-Plasmodium acquired immunity. METHODS To investigate this concern, IgG antibody (Ab) responses to Plasmodium falciparum schizont extract were measured in Senegalese children (6 months-5 years old) who had received three rounds of IPTc with artesunate + sulphadoxine-pyrimethamine (or placebo) at monthly intervals eight months earlier. Potential confounding factors, such as asexual malaria parasitaemia and nutritional status were also evaluated. RESULTS Firstly, a bivariate analysis showed that children who had received IPTc had lower anti-Plasmodium IgG Ab levels than the non-treated controls. When epidemiological parameters were incorporated into a multivariate regression, gender, nutritional status and haemoglobin concentration did not have any significant influence. In contrast, parasitaemia, past malaria morbidity and increasing age were strongly associated with a higher specific IgG response. CONCLUSIONS The intensity of the contacts with P. falciparum seems to represent the main factor influencing anti-schizont IgG responses. Previous IPTc does not seem to interfere with this parasite-dependent acquired humoral response eight months after the last drug administration.
Collapse
Affiliation(s)
- Denis Boulanger
- Unité Mixte de Recherche 145, Institut de Recherche pour le Développement and Université Montpellier 1, 911 avenue Agropolis, BP64501, 34394 Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Tagbor H, Cairns M, Nakwa E, Browne E, Sarkodie B, Counihan H, Meek S, Chandramohan D. The clinical impact of combining intermittent preventive treatment with home management of malaria in children aged below 5 years: cluster randomised trial. Trop Med Int Health 2010; 16:280-9. [PMID: 21159081 DOI: 10.1111/j.1365-3156.2010.02699.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the impact of seasonal intermittent preventive treatment (IPTc) on malaria-related morbidity in children <5 years of age who already had access to home-based management of malaria (HMM) for presumptive treatment of fevers. METHOD Thirty community-based drug distributors (CDDs) from all 13 communities of a rural subdistrict in Ghana were trained to provide prompt treatment for presumptive malaria using artesunate-amodiaquine (AS+AQ) to all children under 5 years of age. Six communities were randomised to also receive bimonthly courses of seasonal IPTc with AS+AQ in May, July and September of 2007. The primary outcome was the incidence rate of febrile episodes diagnosed presumptively as malaria by the CDDs in the communities in each intervention group. Cross-sectional surveys were conducted to determine the prevalence of parasitaemia and anaemia among the study children. RESULTS During the 6 months in which IPTc was delivered, incidence of fevers in communities given HMM+IPTc was lower than in communities given HMM alone, but this difference was not statistically significant (protective efficacy: 37.0%(95% CI: -9.7 to 63.8; P = 0.14). However, incidence of presumptive malaria was significantly lower in IPTc communities when only children who received all three courses of IPTc were included in the analysis: protective efficacy 61.5% (95% CI:31.2-78.5; P = 0.018). Protection with IPTc was not followed by rebound morbidity in the following year. At the end of the intervention period, prevalence of asymptomatic parasitaemia was lower in communities that had received IPTc, but there were no differences in anaemia or haemoglobin concentration. CONCLUSION In this study area, incidence of fevers was lower in communities given three courses of IPTc during the time of peak transmission than in communities that received only HMM. However, high levels of coverage for IPTc will be necessary for maximum impact.
Collapse
Affiliation(s)
- Harry Tagbor
- Department of Community Health, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Steketee RW, Campbell CC. Impact of national malaria control scale-up programmes in Africa: magnitude and attribution of effects. Malar J 2010; 9:299. [PMID: 20979634 PMCID: PMC2988827 DOI: 10.1186/1475-2875-9-299] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 10/27/2010] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Since 2005, malaria control scale-up has progressed in many African countries. Controlled studies of insecticide-treated mosquito nets (ITNs), indoor residual spraying (IRS), intermittent preventive treatment during pregnancy (IPTp) and malaria case management suggested that when incorporated into national programmes a dramatic health impact, likely more than a 20% decrease in all-cause childhood mortality, was possible. To assess the extent to which national malaria programmes are achieving impact the authors reviewed African country programme data available through 2009. METHODS National survey data, published literature, and organization or country reports produced during 2000-2009 were reviewed to assess available malaria financing, intervention delivery, household or target population coverage, and reported health benefits including infection, illness, severe anaemia, and death. RESULTS By the end of 2009, reports were available for ITN household ownership (n = 34) and IPTp use (n = 27) in malaria-endemic countries in Africa, with at least two estimates (pre-2005 and post-2005 intervals). Information linking IRS and case management coverage to impact were more limited. There was generally at least a three-fold increase in household ITN ownership across these countries between pre-2005 (median of 2.4% of households with at least one ITN) and post-2005 (median of 32.5% of households with at least one ITN). Ten countries had temporal data to assess programme impact, and all reported progress on at least one impact indicator (typically on mortality); in under-five year mortality rates most observed a decline of more than 20%. The causal relationship between malaria programme scale-up and reduced child illness and mortality rates is supported by biologic plausibility including mortality declines consistent with experience from intervention efficacy trials, consistency of findings across multiple countries and different epidemiologic settings, and temporal congruity where morbidity and mortality declines have been documented in the 18 to 36 months following intervention scale-up. CONCLUSIONS Several factors potentially have contributed to recent health improvement in African countries, but there is substantial evidence that achieving high malaria control intervention coverage, especially with ITNs and targeted IRS, has been the leading contributor to reduced child mortality. The documented impact provides the evidence required to support a global commitment to the expansion and long-term investment in malaria control to sustain and increase the health impact that malaria control is producing in Africa.
Collapse
Affiliation(s)
- Richard W Steketee
- Malaria Control and Evaluation Partnership in Africa (MACEPA), PATH 2201 Westlake Avenue, Suite 200, Seattle, WA 98121 USA
| | - Carlos C Campbell
- Malaria Control and Evaluation Partnership in Africa (MACEPA), PATH 2201 Westlake Avenue, Suite 200, Seattle, WA 98121 USA
| |
Collapse
|
35
|
Protective efficacy of intermittent preventive treatment of malaria in infants (IPTi) using sulfadoxine-pyrimethamine and parasite resistance. PLoS One 2010; 5:e12618. [PMID: 20838642 PMCID: PMC2935388 DOI: 10.1371/journal.pone.0012618] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 08/06/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intermittent Preventive Treatment of malaria in infants using sulfadoxine-pyrimethamine (SP-IPTi) is recommended by WHO for implementation in settings where resistance to SP is not high. Here we examine the relationship between the protective efficacy of SP-IPTi and measures of SP resistance. METHODS AND RESULTS We analysed the relationship between protective efficacy reported in the 7 SP-IPTi trials and contemporaneous data from 6 in vivo efficacy studies using SP and 7 molecular studies reporting frequency of dhfr triple and dhps double mutations within 50 km of the trial sites. We found a borderline significant association between frequency of the dhfr triple mutation and protective efficacy to 12 months of age of SP-IPTi. This association is significantly biased due to differences between studies, namely number of doses of SP given and follow up times. However, fitting a simple probabilistic model to determine the relationship between the frequency of the dhfr triple, dhps double and dhfr/dhps quintuple mutations associated with resistance to SP and protective efficacy, we found a significant inverse relationship between the dhfr triple mutation frequency alone and the dhfr/dhps quintuple mutations and efficacy at 35 days post the 9 month dose and up to 12 months of age respectively. CONCLUSIONS A significant relationship was found between the frequency of the dhfr triple mutation and SP-IPTi protective efficacy at 35 days post the 9 month dose. An association between the protective efficacy to 12 months of age and dhfr triple and dhfr/dhps quintuple mutations was found but should be viewed with caution due to bias. It was not possible to define a more definite relationship based on the data available from these trials.
Collapse
|
36
|
Buchholz U, Kobbe R, Danquah I, Zanger P, Reither K, Abruquah HH, Grobusch MP, Ziniel P, May J, Mockenhaupt FP. Multiplicity of Plasmodium falciparum infection following intermittent preventive treatment in infants. Malar J 2010; 9:244. [PMID: 20796302 PMCID: PMC2939622 DOI: 10.1186/1475-2875-9-244] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 08/26/2010] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment in infants with sulphadoxine-pyrimethamine (IPTi-SP) reduces malaria morbidity by 20% to 33%. Potentially, however, this intervention may compromise the acquisition of immunity, including the tolerance towards multiple infections with Plasmodium falciparum. METHODS Plasmodium falciparum isolates were obtained from children participating in two Ghanaian IPTi-SP trials (Tamale, Afigya Sekyere) at 15 months of age, i.e., six months after they had received the second dose of IPTi-SP or placebo. By typing the polymorphic merozoite surface protein 1 (msp1) and msp2 genes, multiplicity of infection (MOI) was assessed in 389 isolates. A total of additional 133 samples were collected in Tamale at 3, 6, 9, and 12 months of age. Comparisons of MOI between groups were done by non-parametric statistical tests. RESULTS The number of distinguishable P. falciparum clones (MOI) ranged between one and six. Mean MOI in Tamale was stable at 2.13 - 2.17 during the first year of life, and increased to 2.57 at age 15 months (P = 0.01). At no age did MOI differ between the IPTi-SP and placebo groups (each, P ≥ 0.5). At 15 months of age, i.e., six months after the second dose, MOI was very similar for children who had received IPTi or placebo (means, 2.25 vs. 2.33; P = 0.55) as was the proportion of polyclonal infections (69.6% vs. 69.7%; P = 0.99). Adjusting for study site, current and prior malaria, parasite density, and season did not change this finding. CONCLUSIONS IPTi-SP appears to have no impact on the multiplicity of infection during infancy and thereafter. This suggests that tolerance of multiple infections, a component of protective immunity in highly endemic areas, is not affected by this intervention.
Collapse
Affiliation(s)
- Ulrike Buchholz
- Institute of Tropical Medicine and International Health, Charité - University Medicine, Berlin, Germany
| | - Robin Kobbe
- Infectious Disease Epidemiology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ina Danquah
- Institute of Tropical Medicine and International Health, Charité - University Medicine, Berlin, Germany
| | - Philipp Zanger
- Institute of Tropical Medicine and International Health, Charité - University Medicine, Berlin, Germany
- Institute of Tropical Medicine, University of Tuebingen, Tuebingen, Germany
| | - Klaus Reither
- Institute of Tropical Medicine and International Health, Charité - University Medicine, Berlin, Germany
- Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Harry H Abruquah
- Division of Microbiology and Infectious Diseases, University Hospital, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Martin P Grobusch
- Institute of Tropical Medicine, University of Tuebingen, Tuebingen, Germany
- Medical Research Unit, Hôpital Albert Schweitzer, Lambaréné, Gabon
- Department of Infectious Diseases, Tropical Medicine, HIV/AIDS, Amsterdam Medical Center, University of Amsterdam, The Netherlands
| | - Peter Ziniel
- Northern Region Malaria Project, Tamale, Ghana
- Dept. of Immunology & Microbiology, Rush University, Chicago, USA
| | - Jürgen May
- Infectious Disease Epidemiology Group, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Frank P Mockenhaupt
- Institute of Tropical Medicine and International Health, Charité - University Medicine, Berlin, Germany
| |
Collapse
|
37
|
O'Meara WP, Mangeni JN, Steketee R, Greenwood B. Changes in the burden of malaria in sub-Saharan Africa. THE LANCET. INFECTIOUS DISEASES 2010; 10:545-55. [PMID: 20637696 DOI: 10.1016/s1473-3099(10)70096-7] [Citation(s) in RCA: 360] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The burden of malaria in countries in sub-Saharan Africa has declined with scaling up of prevention, diagnosis, and treatment. To assess the contribution of specific malaria interventions and other general factors in bringing about these changes, we reviewed studies that have reported recent changes in the incidence or prevalence of malaria in sub-Saharan Africa. Malaria control in southern Africa (South Africa, Mozambique, and Swaziland) began in the 1980s and has shown substantial, lasting declines linked to scale-up of specific interventions. In The Horn of Africa, Ethiopia and Eritrea have also experienced substantial decreases in the burden of malaria linked to the introduction of malaria control measures. Substantial increases in funding for malaria control and the procurement and distribution of effective means for prevention and treatment are associated with falls in malaria burden. In central Africa, little progress has been documented, possibly because of publication bias. In some countries a decline in malaria incidence began several years before scale-up of malaria control. In other countries, the change from a failing drug (chloroquine) to a more effective drug (sulphadoxine plus pyrimethamine or an artemisinin combination) led to immediate improvements; in others malaria reduction seemed to be associated with the scale-up of insecticide-treated bednets and indoor residual spraying.
Collapse
|
38
|
Carneiro I, Smith L, Ross A, Roca-Feltrer A, Greenwood B, Schellenberg JA, Smith T, Schellenberg D. Intermittent preventive treatment for malaria in infants: a decision-support tool for sub-Saharan Africa. Bull World Health Organ 2010; 88:807-14. [PMID: 21076561 DOI: 10.2471/blt.09.072397] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 03/05/2010] [Accepted: 03/10/2010] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To develop a decision-support tool to help policy-makers in sub-Saharan Africa assess whether intermittent preventive treatment in infants (IPTi) would be effective for local malaria control. METHODS An algorithm for predicting the effect of IPTi was developed using two approaches. First, study data on the age patterns of clinical cases of Plasmodium falciparum malaria, hospital admissions for infection with malaria parasites and malaria-associated death for different levels of malaria transmission intensity and seasonality were used to estimate the percentage of cases of these outcomes that would occur in children aged <10 years targeted by IPTi. Second, a previously developed stochastic mathematical model of IPTi was used to predict the number of cases likely to be averted by implementing IPTi under different epidemiological conditions. The decision-support tool uses the data from these two approaches that are most relevant to the context specified by the user. FINDINGS Findings from the two approaches indicated that the percentage of cases targeted by IPTi increases with the severity of the malaria outcome and with transmission intensity. The decision-support tool, available on the Internet, provides estimates of the percentage of malaria-associated deaths, hospitalizations and clinical cases that will be targeted by IPTi in a specified context and of the number of these outcomes that could be averted. CONCLUSION The effectiveness of IPTi varies with malaria transmission intensity and seasonality. Deciding where to implement IPTi must take into account the local epidemiology of malaria. The Internet-based decision-support tool described here predicts the likely effectiveness of IPTi under a wide range of epidemiological conditions.
Collapse
Affiliation(s)
- Ilona Carneiro
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, England.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Odhiambo FO, Hamel MJ, Williamson J, Lindblade K, ter Kuile FO, Peterson E, Otieno P, Kariuki S, Vulule J, Slutsker L, Newman RD. Intermittent preventive treatment in infants for the prevention of malaria in rural Western kenya: a randomized, double-blind placebo-controlled trial. PLoS One 2010; 5:e10016. [PMID: 20368815 PMCID: PMC2848869 DOI: 10.1371/journal.pone.0010016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Accepted: 03/09/2010] [Indexed: 11/26/2022] Open
Abstract
Background Intermittent preventive treatment in infants (IPTi) with sulphadoxine-pyrimethamine (SP) for the prevention of malaria has shown promising results in six trials. However, resistance to SP is rising and alternative drug combinations need to be evaluated to better understand the role of treatment versus prophylactic effects. Methods Between March 2004 and March 2008, in an area of western Kenya with year round malaria transmission with high seasonal intensity and high usage of insecticide-treated nets, we conducted a randomized, double-blind placebo-controlled trial with SP plus 3 days of artesunate (SP-AS3), 3 days of amodiaquine-artesunate (AQ3-AS3), or 3 days of short-acting chlorproguanil-dapsone (CD3) administered at routine expanded programme of immunization visits (10 weeks, 14 weeks and 9 months). Principal Findings 1,365 subjects were included in the analysis. The incidence of first or only episode of clinical malaria during the first year of life (primary endpoint) was 0.98 episodes/person-year in the placebo group, 0.74 in the SP-AS3 group, 0.76 in the AQ3-AS3 group, and 0.82 in the CD3 group. The protective efficacy (PE) and 95% confidence intervals against the primary endpoint were: 25.7% (6.3, 41.1); 25.9% (6.8, 41.0); and 16.3% (−5.2, 33.5) in the SP-AS3, AQ3-AS3, and CD3 groups, respectively. The PEs for moderate-to-severe anaemia were: 27.5% (−6.9, 50.8); 23.1% (−11.9, 47.2); and 11.4% (−28.6, 39.0). The duration of the protective effect remained significant for up to 5 to 8 weeks for SP-AS3 and AQ3-AS3. There was no evidence for a sustained beneficial or rebound effect in the second year of life. All regimens were well tolerated. Conclusions These results support the view that IPTi with long-acting regimens provide protection against clinical malaria for up to 8 weeks even in the presence of high ITN coverage, and that the prophylactic rather than the treatment effect of IPTi appears central to its protective efficacy. Trial Registration ClinicalTrials.gov NCT00111163
Collapse
Affiliation(s)
- Frank O Odhiambo
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cairns M, Gosling R, Carneiro I, Gesase S, Mosha JF, Hashim R, Kaur H, Lemnge M, Mosha FW, Greenwood B, Chandramohan D. Duration of protection against clinical malaria provided by three regimens of intermittent preventive treatment in Tanzanian infants. PLoS One 2010; 5:e9467. [PMID: 20209126 PMCID: PMC2830887 DOI: 10.1371/journal.pone.0009467] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2009] [Accepted: 01/29/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment in infants (IPTi) is a new malaria control tool. However, it is uncertain whether IPTi works mainly through chemoprophylaxis or treatment of existing infections. Understanding the mechanism is essential for development of replacements for sulfadoxine-pyrimethamine (SP) where it is no longer effective. This study investigated how protection against malaria given by SP, chlorproguanil-dapsone (CD) and mefloquine (MQ), varied with time since administration of IPTi. METHODS AND FINDINGS A secondary analysis of data from a randomised, placebo-controlled trial in an area of high antifolate resistance in Tanzania was conducted. IPTi using SP, CD, MQ or placebo was given to 1280 infants at 2, 3 and 9 months of age. Poisson regression with random effects to adjust for potential clustering of malaria episodes within children was used to calculate incidence rate ratios for clinical malaria in defined time strata following IPTi. The short-acting antimalarial CD gave no protection against clinical malaria, whereas long-acting MQ gave two months of substantial protection (protective efficacy (PE) 73.1% (95% CI: 23.9, 90.5) and 73.3% (95% CI: 0, 92.9) in the first and second month respectively). SP gave some protection in the first month after treatment (PE 64.5% (95% CI: 10.6, 85.9)) although it did not reduce the incidence of malaria up to 12 months of age. There was no evidence of either long-term protection or increased risk of malaria for any of the regimens. CONCLUSION Post-treatment chemoprophylaxis appears to be the main mechanism by which IPTi protects children against malaria. Long-acting antimalarials are therefore likely to be the most effective drugs for IPTi, but as monotherapies could be vulnerable to development of drug resistance. Due to concerns about tolerability, the mefloquine formulation used in this study is not suitable for IPTi. Further investigation of combinations of long-acting antimalarials for IPTi is needed. TRIAL REGISTRATION Clinicaltrials.gov NCT00158574.
Collapse
Affiliation(s)
- Matthew Cairns
- Department of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Gosling RD, Gesase S, Mosha JF, Carneiro I, Hashim R, Lemnge M, Mosha FW, Greenwood B, Chandramohan D. Protective efficacy and safety of three antimalarial regimens for intermittent preventive treatment for malaria in infants: a randomised, double-blind, placebo-controlled trial. Lancet 2009; 374:1521-32. [PMID: 19765815 DOI: 10.1016/s0140-6736(09)60997-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Administration of sulfadoxine-pyrimethamine at times of vaccination-intermittent preventive treatment in infants (IPTi)-is a promising strategy to prevent malaria. However, rising resistance to this combination is a concern. We investigated a shortacting and longacting antimalarial drug as alternative regimens for IPTi. METHODS We undertook a double-blind, placebo-controlled trial of IPTi in an area of high resistance to sulfadoxine-pyrimethamine at sites of moderate (n=1280 infants enrolled) and low (n=1139) intensity of malaria transmission in Tanzania. Infants aged 8-16 weeks were randomly assigned in blocks of 16 to sulfadoxine (250 mg) plus pyrimethamine (12.5 mg; n=319 in moderate-transmission and 283 in low-transmission sites), chlorproguanil (15 mg) plus dapsone (18.75 mg; n=317 and 285), mefloquine (125 mg; n=320 and 284), or placebo (n=320 and 284), given at the second and third immunisations for diphtheria, pertussis, and tetanus, and for measles. Research team and child were masked to treatment. Recruitment was stopped early at the low-transmission site because of low malaria incidence. The primary endpoint was protective efficacy against all episodes of clinical malaria at 2-11 months of age. Analysis was by intention to treat. This study is registered with ClinicalTrials.gov, number NCT00158574. FINDINGS All randomly assigned infants were analysed. At the moderate-transmission site, mefloquine had a protective efficacy of 38.1% (95% CI 11.8-56.5, p=0.008) against clinical malaria in infants aged 2-11 months, but neither sulfadoxine-pyrimethamine (-6.7%, -45.9 to 22.0) nor chlorproguanil-dapsone (10.8%, -24.6 to 36.1) had a protective effect. No regimen had any protective efficacy against anaemia or hospital admission. Mefloquine caused vomiting in 141 of 1731 (8%) doses given on day 1 (odds ratio vs placebo 5.50, 95% CI 3.56-8.46). More infants died in the chlorproguanil-dapsone and mefloquine groups (18 and 15, respectively) than in the sulfadoxine-pyrimethamine or placebo groups (eight deaths per group; p=0.05 for difference between chlorproguanil-dapsone and placebo). INTERPRETATION IPTi with a longacting, efficacious drug such as mefloquine can reduce episodes of malaria in infants in a moderate-transmission setting. IPTi with sulfadoxine-pyrimethamine has no benefit in areas of very high resistance to this combination. The appropriateness of IPTi should be measured by the expected incidence of malaria and the efficacy, tolerability, and safety of the drug. FUNDING IPTi Consortium and the Gates Malaria Partnership.
Collapse
Affiliation(s)
- Roly D Gosling
- Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aponte JJ, Schellenberg D, Egan A, Breckenridge A, Carneiro I, Critchley J, Danquah I, Dodoo A, Kobbe R, Lell B, May J, Premji Z, Sanz S, Sevene E, Soulaymani-Becheikh R, Winstanley P, Adjei S, Anemana S, Chandramohan D, Issifou S, Mockenhaupt F, Owusu-Agyei S, Greenwood B, Grobusch MP, Kremsner PG, Macete E, Mshinda H, Newman RD, Slutsker L, Tanner M, Alonso P, Menendez C. Efficacy and safety of intermittent preventive treatment with sulfadoxine-pyrimethamine for malaria in African infants: a pooled analysis of six randomised, placebo-controlled trials. Lancet 2009; 374:1533-42. [PMID: 19765816 DOI: 10.1016/s0140-6736(09)61258-7] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Intermittent preventive treatment (IPT) is a promising strategy for malaria control in infants. We undertook a pooled analysis of the safety and efficacy of IPT in infants (IPTi) with sulfadoxine-pyrimethamine in Africa. METHODS We pooled data from six double-blind, randomised, placebo-controlled trials (undertaken one each in Tanzania, Mozambique, and Gabon, and three in Ghana) that assessed the efficacy of IPTi with sulfadoxine-pyrimethamine. In all trials, IPTi or placebo was given to infants at the time of routine vaccinations delivered by WHO's Expanded Program on Immunization. Data from the trials for incidence of clinical malaria, risk of anaemia (packed-cell volume <25% or haemoglobin <80 g/L), and incidence of hospital admissions and adverse events in infants up to 12 months of age were reanalysed by use of standard outcome definitions and time periods. Analysis was by modified intention to treat, including all infants who received at least one dose of IPTi or placebo. FINDINGS The six trials provided data for 7930 infants (IPTi, n=3958; placebo, n=3972). IPTi had a protective efficacy of 30.3% (95% CI 19.8-39.4, p<0.0001) against clinical malaria, 21.3% (8.2-32.5, p=0.002) against the risk of anaemia, 38.1% (12.5-56.2, p=0.007) against hospital admissions associated with malaria parasitaemia, and 22.9% (10.0-34.0, p=0.001) against all-cause hospital admissions. There were 56 deaths in the IPTi group compared with 53 in the placebo group (rate ratio 1.05, 95% CI 0.72-1.54, p=0.79). One death, judged as possibly related to IPTi because it occurred 19 days after a treatment dose, was subsequently attributed to probable sepsis. Four of 676 non-fatal hospital admissions in the IPTi group were deemed related to study treatment compared with five of 860 in the placebo group. None of three serious dermatological adverse events in the IPTi group were judged related to study treatment compared with one of 13 in the placebo group. INTERPRETATION IPTi with sulfadoxine-pyrimethamine was safe and efficacious across a range of malaria transmission settings, suggesting that this intervention is a useful contribution to malaria control. FUNDING Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- John J Aponte
- Barcelona Centre for International Health Research, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cairns M, Gosling R, Gesase S, Mosha J, Greenwood B, Chandramohan D. Mode of action and choice of antimalarial drugs for intermittent preventive treatment in infants. Trans R Soc Trop Med Hyg 2009; 103:1199-201. [PMID: 19740503 DOI: 10.1016/j.trstmh.2009.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 06/04/2009] [Accepted: 06/04/2009] [Indexed: 11/26/2022] Open
Abstract
Intermittent preventive treatment in infants (IPTi) is an effective and safe malaria control strategy. However, it remains unclear what antimalarials should be used to replace sulfadoxine-pyrimethamine (SP) when and where SP is no longer an effective drug for IPTi. Work recently conducted in Tanzania, combined with the findings of previous studies, indicates that IPTi is essentially intermittent chemoprophylaxis; consequently, long-acting antimalarials that provide a long period of post-treatment prophylaxis will be the most effective alternative to SP. However, because of concerns about development of drug resistance, new combinations of long-acting drugs are urgently needed.
Collapse
Affiliation(s)
- Matthew Cairns
- London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | | | | | | | | | | |
Collapse
|
44
|
Barger B, Maiga H, Traore OB, Tekete M, Tembine I, Dara A, Traore ZI, Gantt S, Doumbo OK, Djimde AA. Intermittent preventive treatment using artemisinin-based combination therapy reduces malaria morbidity among school-aged children in Mali. Trop Med Int Health 2009; 14:784-91. [PMID: 19497079 PMCID: PMC3038653 DOI: 10.1111/j.1365-3156.2009.02294.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To assess the efficacy of intermittent preventive treatment (IPT) against malaria in school-aged children. METHODS This was an open randomized controlled trial of seasonal IPT among school children (IPTsc) aged 6-13 years in Kollé, Mali. The study began in September 2007 and completed follow-up in May 2008. Students were randomized to one of three study arms: Sulfadoxine-pyrimethamine plus artesunate (SP/AS), amodiaquine plus artesunate (AQ/AS) or vitamin C. All students received two full treatment doses, given 2 months apart during the season of high transmission from September to December. Groups were compared with respect to incidence of clinical malaria, asymptomatic parasitemia and haemoglobin concentration. RESULTS A total of 296 students were randomized, and retention in the study was 99.3%. Clinical malaria incidence in the SP/AS and AQ/AS arms was reduced by 66.6% and 46.5%, respectively, vs. vitamin C (P < 0.001). There were fewer clinic visits for any cause among the children receiving SP/AS or AQ/AS (P = 0.024). The prevalence of asymptomatic parasitemia was fivefold higher in the vitamin C arm than either SP/AS or AQ/AS at each post-treatment evaluation (P < 0.001). At the end of the transmission period, children treated with IPT had lower rates of anaemia (SP/AS, 17.7%; AQ/AS, 16.0%; vitamin C, 29.6%; P = 0.039). CONCLUSION IPT among school children reduced the rates of clinical malaria, all-cause acute clinic visits, asymptomatic parasitemia and anaemia among school-aged children.
Collapse
Affiliation(s)
- Breanna Barger
- School of Medicine, University of Washington, Seattle, WA, USA
| | - Hamma Maiga
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Oumar Bila Traore
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Mamadou Tekete
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Intimbeye Tembine
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Antoine Dara
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Zoumana Isaac Traore
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Soren Gantt
- Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Ogobara K. Doumbo
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| | - Abdoulaye A. Djimde
- Malaria Research Training Center, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy and Odonto-Stomatology, University of Bamako, Mali
| |
Collapse
|
45
|
Gosling RD, Carneiro I, Chandramohan D. Intermittent preventive treatment of malaria in infants: how does it work and where will it work? Trop Med Int Health 2009; 14:1003-10. [PMID: 19558374 DOI: 10.1111/j.1365-3156.2009.02303.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We discuss the potential public health impact of IPTi by estimating the cases of malaria, anaemia and hospital admissions likely to be averted in different transmission settings; and we review the mechanism of action, choice of drugs regimens, and the effect on immunity of IPTi. IPTi using an efficacious drug is likely to substantially reduce cases of clinical malaria in moderate to high transmission settings. However, geographical heterogeneity in malaria transmission could hamper rolling out IPTi as a national policy.
Collapse
Affiliation(s)
- R D Gosling
- London School of Hygiene & Tropical Medicine, London, UK.
| | | | | |
Collapse
|
46
|
Seasonal intermittent preventive treatment for the prevention of anaemia and malaria in Ghanaian children: a randomized, placebo controlled trial. PLoS One 2008; 3:e4000. [PMID: 19098989 PMCID: PMC2602973 DOI: 10.1371/journal.pone.0004000] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Accepted: 11/13/2008] [Indexed: 11/23/2022] Open
Abstract
Background Malaria and anaemia are the leading causes of morbidity and mortality in children in sub-Saharan Africa. We have investigated the effect of intermittent preventive treatment with sulphadoxine-pyrimethamine or artesunate plus amodiaquine on anaemia and malaria in children in an area of intense, prolonged, seasonal malaria transmission in Ghana. Methods 2451 children aged 3–59 months from 30 villages were individually randomised to receive placebo or artesunate plus amodiaquine (AS+AQ) monthly or bimonthly, or sulphadoxine-pyrimethamine (SP) bimonthly over a period of six months. The primary outcome measures were episodes of anaemia (Hb<8.0 g/dl) or malaria detected through passive surveillance. Findings Monthly artesunate plus amodiaquine reduced the incidence of malaria by 69% (95% CI: 63%, 74%) and anaemia by 45% (95% CI: 25%,60%), bimonthly sulphadoxine-pyrimethamine reduced the incidence of malaria by 24% (95% CI: 14%,33%) and anaemia by 30% (95% CI: 6%, 49%) and bimonthly artesunate plus amodiaquine reduced the incidence of malaria by 17% (95% CI: 6%, 27%) and anaemia by 32% (95% CI: 7%, 50%) compared to placebo. There were no statistically significant reductions in the episodes of all cause or malaria specific hospital admissions in any of the intervention groups compared to the placebo group. There was no significant increase in the incidence of clinical malaria in the post intervention period in children who were >1 year old when they received IPTc compared to the placebo group. However the incidence of malaria in the post intervention period was higher in children who were <1 year old when they received AS+AQ monthly compared to the placebo group. Interpretation IPTc is safe and efficacious in reducing the burden of malaria in an area of Ghana with a prolonged, intense malaria transmission season. Trial Registration ClinicalTrials.gov NCT00119132
Collapse
|
47
|
May J, Adjei S, Busch W, Gabor JJ, Issifou S, Kobbe R, Kreuels B, Lell B, Schwarz NG, Adjei O, Kremsner PG, Grobusch MP. Therapeutic and prophylactic effect of intermittent preventive anti-malarial treatment in infants (IPTi) from Ghana and Gabon. Malar J 2008; 7:198. [PMID: 18828899 PMCID: PMC2586636 DOI: 10.1186/1475-2875-7-198] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Accepted: 10/01/2008] [Indexed: 11/10/2022] Open
Abstract
Background Intermittent preventive treatment in infants (IPTi) with sulphadoxine-pyrimethamine (SP) reduces the incidence of malaria episodes in young children. The exact mechanism by which the protective effect is mediated needs to be defined. This study aimed to investigate therapeutic, prophylactic, and possible exceeding effects of SP-based IPTi in two clinical trials. Methods Protective efficacies from two IPTi trials performed in Kumasi, Ghana, and Lambaréné, Gabon, were assessed for overlapping time series of 61 days. For six-months periods after each of three IPTi doses a multivariate Poisson regression model with the respective cohort as co-variate was generated and effect modification of protective efficacy with time strata was evaluated by log-likelihood tests. Results Protective efficacies were not significantly different between the two study cohorts. Study-cohort corrected protective efficacy was highest for the first 61 days after each IPTi application and decreased continuously. For the first 61 days after IPTi-1, IPTi-2, and IPTi-3 the protective efficacy was 71%, 44%, and 43%, respectively. A reduction of the malaria incidence rate was detectable for the first 60, 30 and 40 days after IPTi-1, IPTi-2 and IPTi-3 drug application, respectively. After IPTi-3 a higher risk for malaria could be seen after day 60. This effect was mainly based on the overwhelming influence of the Kumasi cohort. Conclusion The results suggest that SP-based IPTi mainly works through a therapeutic and prophylactic effect over 30 to 60 days after drug application and that a sustained effect beyond post-treatment prophylaxis might be very low. Trial registration Data analysis from clinical trials NCT ID # 00206739 (Kumasi Trial) and NCT ID # 00167843 (Lambaréné Trial), .
Collapse
Affiliation(s)
- Jürgen May
- Research Group Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht Strasse 74, D-20359 Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ross A, Penny M, Maire N, Studer A, Carneiro I, Schellenberg D, Greenwood B, Tanner M, Smith T. Modelling the epidemiological impact of intermittent preventive treatment against malaria in infants. PLoS One 2008; 3:e2661. [PMID: 18628828 PMCID: PMC2441827 DOI: 10.1371/journal.pone.0002661] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Accepted: 06/07/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Trials of intermittent preventive treatment against malaria in infants (IPTi) using sulphadoxine-pyrimethamine (SP) have shown a positive, albeit variable, protective efficacy against clinical malaria episodes. The impact of IPTi in different epidemiological settings and over time is unknown and predictions are hampered by the lack of knowledge about how IPTi works. We investigated mechanisms proposed for the action of IPTi and made predictions of the likely impact on morbidity and mortality. METHODS/PRINCIPAL FINDINGS We used a comprehensive, individual-based, stochastic model of malaria epidemiology to simulate recently published trials of IPTi using SP with site-specific characteristics as inputs. This baseline model was then modified to represent hypotheses concerning the duration of action of SP, the temporal pattern of fevers caused by individual infections, potential benefits of avoiding fevers on immunity and the effect of sub-therapeutic levels of SP on parasite dynamics. The baseline model reproduced the pattern of results reasonably well. None of the models based on alternative hypotheses improved the fit between the model predictions and observed data. Predictions suggest that IPTi would have a beneficial effect across a range of transmission intensities. IPTi was predicted to avert a greater number of episodes where IPTi coverage was higher, the health system treatment coverage lower, and for drugs which were more efficacious and had longer prophylactic periods. The predicted cumulative benefits were proportionately slightly greater for severe malaria episodes and malaria-attributable mortality than for acute episodes in the settings modelled. Modest increased susceptibility was predicted between doses and following the last dose, but these were outweighed by the cumulative benefits. The impact on transmission intensity was negligible. CONCLUSIONS The pattern of trial results can be accounted for by differences between the trial sites together with known features of malaria epidemiology and the action of SP. Predictions suggest that IPTi would have a beneficial impact across a variety of epidemiological settings.
Collapse
Affiliation(s)
- Amanda Ross
- Department of Public Health and Epidemiology, Swiss Tropical Institute, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|