1
|
Idro R, Nkosi-Gondwe T, Opoka R, Ssenkusu JM, Dennis K, Tsirizani L, Akun P, Rujumba J, Nambatya W, Kamya C, Phiri N, Joanita K, Komata R, Innussa M, Tenywa E, John CC, Tarning J, Denti P, Wasmann RE, Ter Kuile FO, Robberstad B, Phiri KS. Weekly dihydroartemisinin-piperaquine versus monthly sulfadoxine-pyrimethamine for malaria chemoprevention in children with sickle cell anaemia in Uganda and Malawi (CHEMCHA): a randomised, double-blind, placebo-controlled trial. THE LANCET. INFECTIOUS DISEASES 2025; 25:643-655. [PMID: 39718172 PMCID: PMC12095115 DOI: 10.1016/s1473-3099(24)00737-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/23/2024] [Accepted: 10/29/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND In many sub-Saharan African countries, it is recommended that children with sickle cell anaemia receive malaria chemoprevention with monthly sulfadoxine-pyrimethamine or daily proguanil as the standard of care. However, the efficacy of these interventions is compromised by high-grade antifolate resistance of Plasmodium falciparum and poor adherence. We aimed to compare the efficacy of weekly dihydroartemisinin-piperaquine and monthly sulfadoxine-pyrimethamine for the prevention of clinical malaria in children with sickle cell anaemia in areas with high-grade sulfadoxine-pyrimethamine resistance of P falciparum in Uganda and Malawi. METHODS We did an individually randomised, parallel group, double-blind, placebo-controlled trial at two hospitals in Uganda and two hospitals in Malawi. Children (aged 6 months to 15 years) with sickle cell anaemia with a bodyweight of at least 5kg were randomly assigned (1:1) by computer-generated block randomisation, stratified by site and weight category, to receive either weekly dihydroartemisinin-piperaquine (approximately 2·5 mg per kg bodyweight dihydroartemisinin and 20 mg per kg bodyweight per day piperaquine) or monthly sulfadoxine-pyrimethamine (approximately 25 mg per kg bodyweight sulfadoxine and 1·25 mg per kg bodyweight). Placebos matching the alternative treatment were used in each treatment group to maintain masking of the different dosing schedules from the participants and caregivers, study staff, investigators, and data analysts. All children younger than 5 years received penicillin twice daily as standard of care. The primary endpoint was the incidence of clinical malaria, defined as a history of fever in the preceding 48 h or documented axillary temperature of 37·5°C or higher plus the detection of P falciparum parasites on microscopy (any parasite density). Secondary efficacy outcomes were any malaria parasitaemia (on either microscopy or malaria rapid diagnostic test), all-cause unscheduled clinic visits, all-cause and malaria-specific hospitalisation, sickle cell anaemia-related events (including vaso-occlusive crises, acute chest syndrome, stroke), need for blood transfusion, and death. All primary and secondary outcomes were assessed in the modified intention-to-treat population, which included all participants who were randomly assigned for whom endpoint data were available. Safety was assessed in in all children who received at least one dose of the study drug. Complete case analysis was conducted using negative-binomial regression. This study was registered with Clinicaltrials.gov, NCT04844099. FINDINGS Between April 17, 2021, and May 30, 2022, 725 participants were randomly assigned; of whom 724 were included in the primary analysis (367 participants in the dihydroartemisinin-piperaquine group and 357 participants in the sulfadoxine-pyrimethamine group). The median follow-up time was 14·7 months (IQR 11·2-18·2). The incidence of clinical malaria was 8·8 cases per 100 person-years in the dihydroartemisinin-piperaquine group and 43.7 events per 100 person-years in the sulfadoxine-pyrimethamine group (incidence rate ratio [IRR] 0·20 [95% CI 0·14-0·30], p<0·0001). The incidence of hospitalisation with any malaria was lower in the dihydroartemisinin-piperaquine group than the sulfadoxine-pyrimethamine group (10·4 vs 37·0 events per 100 person-years; IRR 0·29 [0·20-0·42], p<0·0001) and the number of blood transfusions was also lower in the dihydroartemisinin-piperaquine group than the sulfadoxine-pyrimethamine group (52·1 vs 72·5 events per 100 person-years; IRR 0·70 [0·54-0·90], p=0·006). The incidence of all-cause unscheduled clinic visits and all-cause hospitalisations were similar between the two groups, however, participants in the dihydroartemisinin-piperaquine group had more clinic visits unrelated to malaria (IRR 1·12 [1·00-1·24], p=0·042) and more hospitalisations with lower respiratory tract events (16·5 vs 8·5 events per 100 person-years; IRR 1·99 [1·25-3·16], p=0·0036) than participants in the sulfadoxine-pyrimethamine group. The number of serious adverse events in the dihydroartemisinin-piperaquine group was similar to that in the sulfadoxine-pyrimethamine group (vaso-occlusive crisis [154 of 367 participants dihydroartemisinin-piperaquine group vs 132 of 357 participants in the sulfadoxine-pyrimethamine group] and suspected sepsis [115 participants vs 92 participants]), with the exception of acute chest syndrome or pneumonia (51 participants vs 32 participants). The number of deaths were similar between groups (six [2%] of 367 participants in the dihydroartemisinin-piperaquine group and eight (2%) of 357 participants in the sulfadoxine-pyrimethamine group). INTERPRETATION Malaria chemoprophylaxis with weekly dihydroartemisinin-piperaquine in children with sickle cell anaemia is safe and considerably more efficacious than monthly sulfadoxine-pyrimethamine. However, monthly sulfadoxine-pyrimethamine was associated with fewer episodes of non-malaria-related illnesses, especially in children 5 years or older not receiving penicillin prophylaxis, which might reflect its antimicrobial effects. In areas with high P falciparum antifolate resistance, dihydroartemisinin-piperaquine should be considered as an alternative to sulfadoxine-pyrimethamine for malaria chemoprevention in children younger than 5 years with sickle cell anaemia receiving penicillin-V prophylaxis. However, there is need for further studies in children older than 5 years. FUNDING Research Council of Norway and UK Medical Research Council. TRANSLATIONS For the Chichewa, Acholi, Lusoga and Luganda translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Richard Idro
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda.
| | | | | | - John M Ssenkusu
- Department of Epidemiology and Biostatistics, Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Lufina Tsirizani
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Pamela Akun
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Joseph Rujumba
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Winnie Nambatya
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Carol Kamya
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Nomsa Phiri
- Training and Research Unit of Excellence, Blantyre, Malawi
| | - Kirikumwino Joanita
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | - Ronald Komata
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Emmanuel Tenywa
- Department of Paediatrics, Jinja Regional Referral Hospital, Jinja, Uganda
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel Tarning
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Paolo Denti
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Roeland E Wasmann
- Division of Clinical Pharmacology, University of Cape Town, Cape Town, South Africa
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Bjarne Robberstad
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Kamija S Phiri
- Training and Research Unit of Excellence, Blantyre, Malawi; School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| |
Collapse
|
2
|
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
|
3
|
Raman J, Mabona M, Nyawo Q, Mangena B, Kok G, Mathaba L, Malatje G, Lauterbach SB, Makhanthisa TI, Gwarinda H, Letinić BD, Ndadza A, Raswiswi E, Shandukani M, Baloyi E, Fekema Z, Ismail A, Featherston J, Greenhouse B, Smith JL, Aranda-Diaz A. Very low prevalence of validated kelch13 mutations and absence of hrp2/3 double gene deletions in South African malaria-eliminating districts (2022-2024). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.31.25324948. [PMID: 40236387 PMCID: PMC11998825 DOI: 10.1101/2025.03.31.25324948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
South Africa is one of 25 countries identified by the World Health Organization as having the potential to eliminate malaria in the near future. In response to the emerging threat of antimalarial resistance, South Africa enhanced its surveillance programs to enable the mapping of resistance markers prevalence down to the facility level. A total of 4 471 samples, collected between January 2022 and August 2024 from healthcare facilities, and during active surveillance in malaria-geliminating districts in KwaZulu-Natal and Mpumalanga provinces, were assessed for drug (mutations in the kelch13 , crt , mdr1 , dhfr, and dhps genes) and diagnostic ( hrp 2/3 gene deletions) resistance markers using PCR and sequencing (Sanger and/or targeted amplicon) protocols. Validated markers of artemisinin partial resistance were rare, with the P574L mutation detected as a minor allele in two samples and the P553L mutation present in one sample. Of the 60 additional non-synonymous mutations detected, the A578S (30/999) and the I494V (13/951) mutations were most prevalent. Almost all parasites assessed carried the crtK76 (99.8%) and mdr1N 86 (99.0%) alleles, suggesting susceptibility to chloroquine. The dhfr triple (99.9%) and dhps double (98.2%) mutations associated with pyrimethamine and sulfadoxine resistance, respectively, were close to fixation. No double hrp 2/3 gene deletions were detected. These findings suggest that the recommended treatments and diagnostics in South Africa are effective. However, the strong selection for antimalarial drug resistance markers across southern Africa combined with high regional interconnectedness, emphasizes the need for sustained malaria molecular surveillance to support South and southern Africa achieve their elimination goals.
Collapse
|
4
|
Millogo KS, Zabré A, Sondo P, Kaboré B, Kouevi AFC, Compaoré EW, Bayala IMC, Ismaïla B, Hien SVF, Rouamba T, Kazienga A, Derra K, Rouamba E, Tahita MC, Ouédraogo F, Ilboudo H, Bamba S, Tinto H. Seasonal malaria chemoprevention and mutations in Pfdhfr and Pfdhps genes in children in the health district of Nanoro, Burkina Faso. MALARIAWORLD JOURNAL 2025; 16:5. [PMID: 40124705 PMCID: PMC11926660 DOI: 10.5281/zenodo.15039792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Introduction Seasonal malaria chemoprevention (SMC) is an effective malaria preventive intervention in sub-Saharan Africa. As with other drug-based interventions, large-scale deployment increases drug pressure, which may result in drug-resistant parasite strains thereby jeopardising the impact of the intervention. Mutations in Pfdhps and Pfdhfr genes are known to be associated with resistance to sulfadoxine and pyrimethamine, respectively, making the surveillance of molecular markers crucial in settings where SMC is widely applied. This study aimed at assessing the distribution of Pfdhfr and Pfdhps alleles before and after the 2021 annual campaign of SMC in the health district of Nanoro in Burkina Faso. Materials and Methods Randomly selected dried blood spots collected prior (n=100) and after (n=100) the 2021 SMC campaign were used for the detection of mutation in codons 51, 59 and 108 of the Pfdhfr gene and in codons 437 and 540 of Pfdhps gene using a nested PCR with restriction fragment length polymorphism approach. Results The prevalence of Pfdhfr and Pfdhps mutant alleles were very high before and after SMC, ranging from 88.42% to 97.98%. However, no significant change in the prevalence of Pfdhfr and Pfdhps mutant alleles was observed in the period before and after SMC campaign (p>0.05). No mutation was observed in Pfdhps codon 540. In addition, the prevalence of the Pfdhfr triple mutant and Pfhfr-dhps quadruple mutant was higher in the study area but with no significant variation before and after SMC campaign (p>0.05). . Conclusions The prevalence of Pfdhfr and Pfdhps mutant alleles were higher either in pre or post SMC. However, no significant variation in the prevalence of these alleles was observed following the SMC campaign suggesting that these high mutation frequencies may be the result of continuous use of SMC in Burkina Faso since 2014.
Collapse
Affiliation(s)
- Kié Solange Millogo
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Adjaratou Zabré
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Paul Sondo
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Bérenger Kaboré
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Amélé Fifi Chantal Kouevi
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Eulalie W. Compaoré
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Ipéné Mylène Carenne Bayala
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Bouda Ismaïla
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - So-vii Franck Hien
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Adama Kazienga
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Karim Derra
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Eli Rouamba
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Marc Christian Tahita
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Florence Ouédraogo
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Hamidou Ilboudo
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Sanata Bamba
- Institut Supérieur des Sciences de la Santé (INSSA)/Université Nazi Boni, Bobo Dioulasso, Burkina Faso
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé (IRSS)/ Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| |
Collapse
|
5
|
Ngwana-Joseph GC, Phelan JE, Manko E, Dombrowski JG, da Silva Santos S, Suarez-Mutis M, Vélez-Tobón G, Tobón Castaño A, Machado RLD, Marinho CRF, Nolder D, Nosten F, Sutherland CJ, Campino S, Clark TG. Genomic analysis of global Plasmodium vivax populations reveals insights into the evolution of drug resistance. Nat Commun 2024; 15:10771. [PMID: 39738010 PMCID: PMC11685768 DOI: 10.1038/s41467-024-54964-x] [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: 04/08/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Increasing reports of chloroquine resistance (CQR) in Plasmodium vivax endemic regions have led to several countries, including Indonesia, to adopt dihydroarteminsin-piperaquine instead. However, the molecular drivers of CQR remain unclear. Using a genome-wide approach, we perform a genomic analysis of 1534 P. vivax isolates across 29 endemic countries, detailing population structure, patterns of relatedness, selection, and resistance profiling, providing insights into potential drivers of CQR. Selective sweeps in a locus proximal to pvmdr1, a putative marker for CQR, along with transcriptional regulation genes, distinguish isolates from Indonesia from those in regions where chloroquine remains highly effective. In 106 isolates from Indonesian Papua, the epicentre of CQR, we observe an increasing prevalence of novel SNPs in the candidate resistance gene pvmrp1 since the introduction of dihydroartemisinin-piperaquine. Overall, we provide novel markers for resistance surveillance, supported by evidence of regions under recent directional selection and temporal analysis in this continually evolving parasite.
Collapse
Affiliation(s)
- Gabrielle C Ngwana-Joseph
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Jody E Phelan
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Emilia Manko
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Jamille G Dombrowski
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - Gabriel Vélez-Tobón
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Antioquia, Colombia
| | | | - Ricardo Luiz Dantas Machado
- Centro de Investigação de Microrganismos - CIM, Departamento de Microbiologia e Parasitologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Claudio R F Marinho
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Debbie Nolder
- UK Health Security Agency, Malaria Reference Laboratory, London School of Hygiene and Tropical Medicine, London, UK
| | - François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Colin J Sutherland
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- UK Health Security Agency, Malaria Reference Laboratory, London School of Hygiene and Tropical Medicine, London, UK
| | - Susana Campino
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| | - Taane G Clark
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
6
|
Zhou R, Li S, Ji P, Ruan S, Liu Y, Yang C, Qian D, He Z, Wang D, Lu D, Zhang H, Deng Y. Prevalence of molecular markers of sulfadoxine-pyrimethamine resistance in Plasmodium falciparum isolates from West Africa during 2012-2022. Sci Rep 2024; 14:26567. [PMID: 39496624 PMCID: PMC11535326 DOI: 10.1038/s41598-024-75828-w] [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: 05/21/2024] [Accepted: 10/08/2024] [Indexed: 11/06/2024] Open
Abstract
Sulfadoxine-pyrimethamine (SP) is a key drug recommended by the World Health Organization for the chemoprevention of malaria. However, the strategy is affected by the parasite resistance to SP. This study evaluated Plasmodium falciparum dihydrofolate reductase (Pfdhfr) and dihydropteroate synthase (Pfdhps) genes, associated with SP resistance, from 508 P. falciparum isolates imported from West African countries to Henan Province, China, during 2012-2022. High mutant prevalence of the genes Pfdhfr (94.7%) and Pfdhps (96.8%) was observed. The mutants Pfdhfr N51I, C59R, S108N, and Pfdhps A437G were at high frequency in all countries analyzed. The overall prevalence of the mutant Pfdhps K540E was low (3.4%), but with a high frequency in Liberia (24.3%). The frequency of mutants Pfdhps I431V, A581G, and A613S was 11.7%, 9.8%, and 16.2%, respectively, all of which had the highest mutant prevalence in Nigeria. The mutant Pfdhps A581G and A613S were identified in the absence of K540E. The partially resistant haplotype (I51R59N108 - G437) was the most common (72.6%), and the fully resistant haplotype (I51R59N108 - G437E540) had a low prevalence of 3.4% and mainly occurred in Liberia. No super resistant haplotype was identified. The mutant Pfdhps I431V and the octuple mutant haplotype I51R59N108 - V431A436G437G581S613 deserve more attention. In areas of high SP resistance, the intervention still reduces low birthweight and maternal anaemia. SP should continue to be used in areas of high SP resistance until more effective alternatives for malaria chemoprevention are found. It is important to continuously monitor the molecular markers associated with SP resistance to better implement intermittent preventive treatment policies in pregnancy (IPTp) and infants (IPTi).
Collapse
Affiliation(s)
- Ruimin Zhou
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Suhua Li
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Penghui Ji
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Shucheng Ruan
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ying Liu
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Chengyun Yang
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Dan Qian
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Zhiquan He
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Dan Wang
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Deling Lu
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China
| | - Hongwei Zhang
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China.
| | - Yan Deng
- Department of Parasite Disease Control and Prevention, Henan Provincial Key Laboratory of Pathogenic Microbiology, Henan Provincial Medical Key Laboratory of Parasitic Diseases and Vector, Henan Province Center for Disease Control and Prevention, Zhengzhou, People's Republic of China.
| |
Collapse
|
7
|
El Gaaloul M, Tchouatieu AM, Kayentao K, Campo B, Buffet B, Ramachandruni H, Ndiaye JL, Wells TNC, Audibert C, Achan J, Donini C, Barsosio HC, Tinto H. Chemoprevention of malaria with long-acting oral and injectable drugs: an updated target product profile. Malar J 2024; 23:315. [PMID: 39425110 PMCID: PMC11490162 DOI: 10.1186/s12936-024-05128-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
Malaria is preventable, but the burden of disease remains high with over 249 million cases and 608,000 deaths reported in 2022. Historically, the most important protective interventions have been vector control and chemopreventive medicines with over 50 million children receiving seasonal malaria chemoprevention in the year 2023. Two vaccines are approved and starting to be deployed, bringing additional protection for children up to 36 months. However, the impact of these currently available tools is somewhat limited on various fronts. Vaccines exhibit partial efficacy, are relatively costly, and not accessible in all settings. The challenges encountered with chemoprevention are barriers to acceptability and feasibility, including frequency of dosing, and the lack of options in the first trimester of pregnancy and for women living with HIV. Also, the emergence of resistance against chemopreventive medicines is concerning. To address these limitations, a target product profile (TPP) is proposed as a road map to guide innovation and to boost the quest for novel chemopreventive alternatives. This TPP describes the ideal product attributes, while acknowledging potential trade-offs that may be needed. Critically, it considers the target populations most at risk; primarily infants, children, and pregnant women. Malaria control and elimination requires appropriate chemoprevention, not only in areas of high endemicity and transmission, but also in lower transmission areas where immunity is declining, as well as for travellers from areas where malaria has been eliminated. New medicines should show acceptable safety and tolerability, with high and long protective efficacy. Formulations and costs need to support operational adherence, access, and effectiveness. Next generation long-acting oral and injectable drugs are likely to constitute the backbone of malaria prevention. Therefore, the perspectives of front-line experts in malaria prevention, researchers, and those involved in drug development are captured in the TPP. This inclusive approach aims at concentrating efforts and aligning responses across the community to develop new and transformative medicines.
Collapse
Affiliation(s)
- Myriam El Gaaloul
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland.
| | - Andre Marie Tchouatieu
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland.
| | - Kassoum Kayentao
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Brice Campo
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Benedicte Buffet
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Hanu Ramachandruni
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | | | - Timothy N C Wells
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Celine Audibert
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | | | - Cristina Donini
- MMV Medicines for Malaria Venture, Route de Pré-Bois 20, Post Box 1826, 1215, Geneva 15, Switzerland
| | - Hellen C Barsosio
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| |
Collapse
|
8
|
Maiga M, Dembele L, Courlet P, Khandelwal A, Dara A, Sogore F, Diakité O, Maiga FO, Dao F, Sissoko S, Barre Y, Goita S, Diakite M, Diakite SAS, Djimde AA, Oeuvray C, Spangenberg T, Wicha SG, Demarta-Gatsi C. Towards clinically relevant dose ratios for Cabamiquine and Pyronaridine combination using P. falciparum field isolate data. Nat Commun 2024; 15:7659. [PMID: 39227370 PMCID: PMC11372057 DOI: 10.1038/s41467-024-51994-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024] Open
Abstract
The selection and combination of dose regimens for antimalarials involve complex considerations including pharmacokinetic and pharmacodynamic interactions. In this study, we use immediate ex vivo P. falciparum field isolates to evaluate the effect of cabamiquine and pyronaridine as standalone treatments and in combination therapy. We feed the data into a pharmacometrics model to generate an interaction map and simulate meaningful clinical dose ratios. We demonstrate that the pharmacometrics model of parasite growth and killing provides a detailed description of parasite kinetics against cabamiquine-susceptible and resistant parasites. Pyronaridine monotherapy provides suboptimal killing rates at doses as high as 720 mg. In contrast, the combination of a single dose of 330 mg cabamiquine and 360 mg pyronaridine provides over 90% parasite killing in most of the simulated patients. The described methodology that combines a rapid, 3R-compliant in vitro method and modelling to set meaningful doses for new antimalarials could contribute to clinical drug development.
Collapse
Affiliation(s)
- Mohamed Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Laurent Dembele
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali.
| | - Perrine Courlet
- Merck Institute of Pharmacometrics (an affiliate of Merck KGaA), Lausanne, Switzerland
| | - Akash Khandelwal
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
- UCB Biosciences GmbH, Rolf-Schwarz-Schütte-Platz 1, Monheim am Rhein, Germany
| | - Antoine Dara
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fanta Sogore
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Ousmaila Diakité
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Fatoumata O Maiga
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - François Dao
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Sekou Sissoko
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Yacouba Barre
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Siaka Goita
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Mahamadou Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Seidina A S Diakite
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
| | - Abdoulaye A Djimde
- Université des Sciences, des Techniques et des Technologies de Bamako (USTTB), Malaria Research and Training Centre (MRTC), Faculty of Pharmacy, Bamako, Mali
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali
| | - Claude Oeuvray
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Thomas Spangenberg
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland
| | - Sebastian G Wicha
- Department of Clinical Pharmacy, Institute of Pharmacy, University of Hamburg, Bundesstr. 45, Hamburg, Germany.
| | - Claudia Demarta-Gatsi
- Pathogens genomic Diversity Network Africa, Sotuba, Bamako, Mali.
- Global Health R&D of the healthcare business of Merck KGaA, Darmstadt, Germany, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Eysins, Switzerland.
| |
Collapse
|
9
|
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
|
10
|
Mousa A, Cuomo-Dannenburg G, Thompson HA, Chico RM, Beshir KB, Sutherland CJ, Schellenberg D, Gosling R, Alifrangis M, Hocke EF, Hansson H, Chopo-Pizarro A, Mbacham WF, Ali IM, Chaponda M, Roper C, Okell LC. Measuring protective efficacy and quantifying the impact of drug resistance: A novel malaria chemoprevention trial design and methodology. PLoS Med 2024; 21:e1004376. [PMID: 38723040 PMCID: PMC11081503 DOI: 10.1371/journal.pmed.1004376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/14/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND Recently revised WHO guidelines on malaria chemoprevention have opened the door to more tailored implementation. Countries face choices on whether to replace old drugs, target additional age groups, and adapt delivery schedules according to local drug resistance levels and malaria transmission patterns. Regular routine assessment of protective efficacy of chemoprevention is key. Here, we apply a novel modelling approach to aid the design and analysis of chemoprevention trials and generate measures of protection that can be applied across a range of transmission settings. METHODS AND FINDINGS We developed a model of genotype-specific drug protection, which accounts for underlying risk of infection and circulating genotypes. Using a Bayesian framework, we fitted the model to multiple simulated scenarios to explore variations in study design, setting, and participant characteristics. We find that a placebo or control group with no drug protection is valuable but not always feasible. An alternative approach is a single-arm trial with an extended follow-up (>42 days), which allows measurement of the underlying infection risk after drug protection wanes, as long as transmission is relatively constant. We show that the currently recommended 28-day follow-up in a single-arm trial results in low precision of estimated 30-day chemoprevention efficacy and low power in determining genotype differences of 12 days in the duration of protection (power = 1.4%). Extending follow-up to 42 days increased precision and power (71.5%) in settings with constant transmission over this time period. However, in settings of unstable transmission, protective efficacy in a single-arm trial was overestimated by 24.3% if recruitment occurred during increasing transmission and underestimated by 15.8% when recruitment occurred during declining transmission. Protective efficacy was estimated with greater precision in high transmission settings, and power to detect differences by resistance genotype was lower in scenarios where the resistant genotype was either rare or too common. CONCLUSIONS These findings have important implications for the current guidelines on chemoprevention efficacy studies and will be valuable for informing where these studies should be optimally placed. The results underscore the need for a comparator group in seasonal settings and provide evidence that the extension of follow-up in single-arm trials improves the accuracy of measures of protective efficacy in settings with more stable transmission. Extension of follow-up may pose logistical challenges to trial feasibility and associated costs. However, these studies may not need to be repeated multiple times, as the estimates of drug protection against different genotypes can be applied to different settings by adjusting for transmission intensity and frequency of resistance.
Collapse
Affiliation(s)
- Andria Mousa
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gina Cuomo-Dannenburg
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Hayley A. Thompson
- Malaria and Neglected Tropical Diseases, PATH, Seattle, Washington, United States of America
| | - R. Matthew Chico
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Khalid B. Beshir
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Colin J. Sutherland
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - David Schellenberg
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Roly Gosling
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Malaria Elimination Initiative, Institute of Global Health, University of California, San Francisco, California, United States of America
| | - Michael Alifrangis
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emma Filtenborg Hocke
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helle Hansson
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Ana Chopo-Pizarro
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Wilfred F. Mbacham
- The Biotechnology Centre, University of Yaoundé, Yaoundé, Cameroon
- The Fobang Institutes for Innovation in Science and Technology, Yaoundé, Cameroon
- The Faculty of Northwest University, Faculty of Natural and Agricultural Sciences, Potchefstroom, South Africa
| | - Innocent M. Ali
- The Biotechnology Centre, University of Yaoundé, Yaoundé, Cameroon
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Mike Chaponda
- Department of Clinical Sciences, Tropical Diseases Research Centre, Ndola, Zambia
| | - Cally Roper
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lucy C. Okell
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Nain M, Dhorda M, Flegg JA, Gupta A, Harrison LE, Singh-Phulgenda S, Otienoburu SD, Harriss E, Bharti PK, Behera B, Rahi M, Guerin PJ, Sharma A. Systematic Review and Geospatial Modeling of Molecular Markers of Resistance to Artemisinins and Sulfadoxine-Pyrimethamine in Plasmodium falciparum in India. Am J Trop Med Hyg 2024; 110:910-920. [PMID: 38574550 PMCID: PMC11066343 DOI: 10.4269/ajtmh.23-0631] [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: 09/12/2023] [Accepted: 12/17/2023] [Indexed: 04/06/2024] Open
Abstract
Surveillance for genetic markers of resistance can provide valuable information on the likely efficacy of antimalarials but needs to be targeted to ensure optimal use of resources. We conducted a systematic search and review of publications in seven databases to compile resistance marker data from studies in India. The sample collection from the studies identified from this search was conducted between 1994 and 2020, and these studies were published between 1994 and 2022. In all, Plasmodium falciparum Kelch13 (PfK13), P. falciparum dihydropteroate synthase, and P. falciparum dihydrofolate reductase (PfDHPS) genotype data from 2,953, 4,148, and 4,222 blood samples from patients with laboratory-confirmed malaria, respectively, were extracted from these publications and uploaded onto the WorldWide Antimalarial Resistance Network molecular surveyors. These data were fed into hierarchical geostatistical models to produce maps with a predicted prevalence of the PfK13 and PfDHPS markers, and of the associated uncertainty. Zones with a predicted PfDHPS 540E prevalence of >15% were identified in central, eastern, and northeastern India. The predicted prevalence of PfK13 mutants was nonzero at only a few locations, but were within or adjacent to the zones with >15% prevalence of PfDHPS 540E. There may be a greater probability of artesunate-sulfadoxine-pyrimethamine failures in these regions, but these predictions need confirmation. This work can be applied in India and elsewhere to help identify the treatments most likely to be effective for malaria elimination.
Collapse
Affiliation(s)
- Minu Nain
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Mehul Dhorda
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Infectious Diseases Data Observatory, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jennifer A. Flegg
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Apoorv Gupta
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Lucinda E. Harrison
- School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria, Australia
| | - Sauman Singh-Phulgenda
- Infectious Diseases Data Observatory, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sabina D. Otienoburu
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Infectious Diseases Data Observatory, Oxford, United Kingdom
- College of STEM, Johnson C. Smith University, Charlotte, North Carolina
| | - Eli Harriss
- The Knowledge Centre, Bodleian Health Care Libraries, University of Oxford, Oxford, United Kingdom
| | | | - Beauty Behera
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Manju Rahi
- ICMR-National Institute of Malaria Research, New Delhi, India
- Indian Council of Medical Research, New Delhi, India
- Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh
| | - Philippe J. Guerin
- WorldWide Antimalarial Resistance Network, Oxford, United Kingdom
- Infectious Diseases Data Observatory, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Amit Sharma
- ICMR-National Institute of Malaria Research, New Delhi, India
- Molecular Medicine, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
12
|
Flegg JA, Kandanaarachchi S, Guerin PJ, Dondorp AM, Nosten FH, Otienoburu SD, Golding N. Spatio-temporal spread of artemisinin resistance in Southeast Asia. PLoS Comput Biol 2024; 20:e1012017. [PMID: 38626207 PMCID: PMC11051648 DOI: 10.1371/journal.pcbi.1012017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 04/26/2024] [Accepted: 03/22/2024] [Indexed: 04/18/2024] Open
Abstract
Current malaria elimination targets must withstand a colossal challenge-resistance to the current gold standard antimalarial drug, namely artemisinin derivatives. If artemisinin resistance significantly expands to Africa or India, cases and malaria-related deaths are set to increase substantially. Spatial information on the changing levels of artemisinin resistance in Southeast Asia is therefore critical for health organisations to prioritise malaria control measures, but available data on artemisinin resistance are sparse. We use a comprehensive database from the WorldWide Antimalarial Resistance Network on the prevalence of non-synonymous mutations in the Kelch 13 (K13) gene, which are known to be associated with artemisinin resistance, and a Bayesian geostatistical model to produce spatio-temporal predictions of artemisinin resistance. Our maps of estimated prevalence show an expansion of the K13 mutation across the Greater Mekong Subregion from 2000 to 2022. Moreover, the period between 2010 and 2015 demonstrated the most spatial change across the region. Our model and maps provide important insights into the spatial and temporal trends of artemisinin resistance in a way that is not possible using data alone, thereby enabling improved spatial decision support systems on an unprecedented fine-scale spatial resolution. By predicting for the first time spatio-temporal patterns and extents of artemisinin resistance at the subcontinent level, this study provides critical information for supporting malaria elimination goals in Southeast Asia.
Collapse
Affiliation(s)
- Jennifer A. Flegg
- School of Mathematics and Statistics, University of Melbourne, Melbourne, Australia
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, United Kingdom
| | | | - Philippe J. Guerin
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, United Kingdom
- Infectious Diseases Data Observatory (IDDO), Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Arjen M. Dondorp
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Francois H. Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Sabina Dahlström Otienoburu
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, United Kingdom
- Infectious Diseases Data Observatory (IDDO), Oxford, United Kingdom
- College of Science, Technology, Engineering and Mathematics, Johnson C. Smith University, Charlotte, North Carolina, United States of America
| | - Nick Golding
- Telethon Kids Institute and Curtin University, Perth, Australia
| |
Collapse
|
13
|
Kamya MR, Kakuru A, Dorsey G. Dihydroartemisinin-piperaquine for prevention of malaria in pregnant women living with HIV. Lancet 2024; 403:327-330. [PMID: 38224711 DOI: 10.1016/s0140-6736(24)00048-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Affiliation(s)
- Moses R Kamya
- Department of Medicine, Makerere University, PO Box 7072, Kampala, Uganda; Infectious Diseases Research Collaboration, Kampala, Uganda.
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda; Department of Community and Public Health, Busitema University, Tororo, Uganda
| | - Grant Dorsey
- and Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
14
|
Barsosio HC, Madanitsa M, Ondieki ED, Dodd J, Onyango ED, Otieno K, Wang D, Hill J, Mwapasa V, Phiri KS, Maleta K, Taegtmeyer M, Kariuki S, Schmiegelow C, Gutman JR, Ter Kuile FO. Chemoprevention for malaria with monthly intermittent preventive treatment with dihydroartemisinin-piperaquine in pregnant women living with HIV on daily co-trimoxazole in Kenya and Malawi: a randomised, double-blind, placebo-controlled trial. Lancet 2024; 403:365-378. [PMID: 38224710 PMCID: PMC10865779 DOI: 10.1016/s0140-6736(23)02631-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND The efficacy of daily co-trimoxazole, an antifolate used for malaria chemoprevention in pregnant women living with HIV, is threatened by cross-resistance of Plasmodium falciparum to the antifolate sulfadoxine-pyrimethamine. We assessed whether addition of monthly dihydroartemisinin-piperaquine to daily co-trimoxazole is more effective at preventing malaria infection than monthly placebo plus daily co-trimoxazole in pregnant women living with HIV. METHODS We did an individually randomised, two-arm, placebo-controlled trial in areas with high-grade sulfadoxine-pyrimethamine resistance in Kenya and Malawi. Pregnant women living with HIV on dolutegravir-based combination antiretroviral therapy (cART) who had singleton pregnancies between 16 weeks' and 28 weeks' gestation were randomly assigned (1:1) by computer-generated block randomisation, stratified by site and HIV status (known positive vs newly diagnosed), to daily co-trimoxazole plus monthly dihydroartemisinin-piperaquine (three tablets of 40 mg dihydroartemisinin and 320 mg piperaquine given daily for 3 days) or daily co-trimoxazole plus monthly placebo. Daily co-trimoxazole consisted of one tablet of 160 mg sulfamethoxazole and 800 mg trimethoprim. The primary endpoint was the incidence of Plasmodium infection detected in the peripheral (maternal) or placental (maternal) blood or tissue by PCR, microscopy, rapid diagnostic test, or placental histology (active infection) from 2 weeks after the first dose of dihydroartemisinin-piperaquine or placebo to delivery. Log-binomial regression was used for binary outcomes, and Poisson regression for count outcomes. The primary analysis was by modified intention to treat, consisting of all randomised eligible participants with primary endpoint data. The safety analysis included all women who received at least one dose of study drug. All investigators, laboratory staff, data analysts, and participants were masked to treatment assignment. This trial is registered with ClinicalTrials.gov, NCT04158713. FINDINGS From Nov 11, 2019, to Aug 3, 2021, 904 women were enrolled and randomly assigned to co-trimoxazole plus dihydroartemisinin-piperaquine (n=448) or co-trimoxazole plus placebo (n=456), of whom 895 (99%) contributed to the primary analysis (co-trimoxazole plus dihydroartemisinin-piperaquine, n=443; co-trimoxazole plus placebo, n=452). The cumulative risk of any malaria infection during pregnancy or delivery was lower in the co-trimoxazole plus dihydroartemisinin-piperaquine group than in the co-trimoxazole plus placebo group (31 [7%] of 443 women vs 70 [15%] of 452 women, risk ratio 0·45, 95% CI 0·30-0·67; p=0·0001). The incidence of any malaria infection during pregnancy or delivery was 25·4 per 100 person-years in the co-trimoxazole plus dihydroartemisinin-piperaquine group versus 77·3 per 100 person-years in the co-trimoxazole plus placebo group (incidence rate ratio 0·32, 95% CI 0·22-0·47, p<0·0001). The number needed to treat to avert one malaria infection per pregnancy was 7 (95% CI 5-10). The incidence of serious adverse events was similar between groups in mothers (17·7 per 100 person-years in the co-trimoxazole plus dihydroartemisinin-piperaquine group [23 events] vs 17·8 per 100 person-years in the co-trimoxazole group [25 events]) and infants (45·4 per 100 person-years [23 events] vs 40·2 per 100 person-years [21 events]). Nausea within the first 4 days after the start of treatment was reported by 29 (7%) of 446 women in the co-trimoxazole plus dihydroartemisinin-piperaquine group versus 12 (3%) of 445 women in the co-trimoxazole plus placebo group. The risk of adverse pregnancy outcomes did not differ between groups. INTERPRETATION Addition of monthly intermittent preventive treatment with dihydroartemisinin-piperaquine to the standard of care with daily unsupervised co-trimoxazole in areas of high antifolate resistance substantially improves malaria chemoprevention in pregnant women living with HIV on dolutegravir-based cART and should be considered for policy. FUNDING European and Developing Countries Clinical Trials Partnership 2; UK Joint Global Health Trials Scheme (UK Foreign, Commonwealth and Development Office; Medical Research Council; National Institute for Health Research; Wellcome); and Swedish International Development Cooperation Agency.
Collapse
Affiliation(s)
- Hellen C Barsosio
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Mwayiwawo Madanitsa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi; Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | - Everlyne D Ondieki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Eric D Onyango
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Victor Mwapasa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Kamija S Phiri
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Kenneth Maleta
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Miriam Taegtmeyer
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Christentze Schmiegelow
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Gynaecology and Obstetrics, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Feiko O Ter Kuile
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
15
|
Thwing J, Williamson J, Cavros I, Gutman JR. Systematic Review and Meta-Analysis of Seasonal Malaria Chemoprevention. Am J Trop Med Hyg 2024; 110:20-31. [PMID: 38081050 PMCID: PMC10793029 DOI: 10.4269/ajtmh.23-0481] [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: 07/23/2023] [Accepted: 10/23/2023] [Indexed: 01/05/2024] Open
Abstract
Seasonal malaria chemoprevention (SMC) for children under 5 years of age for up to four monthly cycles during malaria transmission season was recommended by the WHO in 2012 and has been implemented in 13 countries in the Sahel, reaching more than 30 million children annually. Malaria control programs implementing SMC have asked the WHO to consider expanding the age range or number of monthly cycles. We conducted a systematic review and meta-analysis of SMC among children up to 15 years of age and up to six monthly cycles. Twelve randomized studies were included, with outcomes stratified by age (< 5/≥ 5 years), by three or four versus five or six cycles, and by drug where possible. Drug regimens included sulfadoxine-pyrimethamine + amodiaquine, amodiaquine-artesunate, and sulfadoxine-pyrimethamine + artesunate. Included studies were all conducted in Sahelian countries in which high-grade resistance to sulfadoxine-pyrimethamine was rare and in zones with parasite prevalence ranging from 1% to 79%. Seasonal malaria chemoprevention resulted in substantial reductions in uncomplicated malaria incidence measured during that transmission season (rate ratio: 0.27, 95% CI: 0.25-0.29 among children < 5 years; rate ratio: 0.27, 95% CI: 0.25-0.30 among children ≥ 5 years) and in the prevalence of malaria parasitemia measured within 4-6 weeks from the final SMC cycle (risk ratio: 0.38, 95% CI: 0.34-0.43 among children < 5 years; risk ratio: 0.23, 95% CI: 0.11-0.48 among children ≥ 5 years). In high-transmission zones, SMC resulted in a moderately reduced risk of any anemia (risk ratio: 0.77, 95% CI: 0.72-0.83 among children < 5 years; risk ratio: 0.70, 95% CI: 0.52-0.95 among children ≥ 5 years [one study]). Children < 10 years of age had a moderate reduction in severe malaria (risk ratio: 0.53, 95% CI: 0.37-0.76) but no evidence of a mortality reduction. The evidence suggests that in areas in which sulfadoxine-pyrimethamine and amodiaquine remained efficacious, SMC effectively reduced malaria disease burden among children both < 5 and ≥ 5 years old and that the number of cycles should be commensurate with the length of the transmission season, up to six cycles.
Collapse
Affiliation(s)
- Julie Thwing
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - John Williamson
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Irene Cavros
- U.S. President’s Malaria Initiative, Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Julie R. Gutman
- Malaria Branch, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
16
|
Foo YS, Flegg JA. A spatio-temporal model of multi-marker antimalarial resistance. J R Soc Interface 2024; 21:20230570. [PMID: 38228183 PMCID: PMC10791536 DOI: 10.1098/rsif.2023.0570] [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: 10/01/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024] Open
Abstract
The emergence and spread of drug-resistant Plasmodium falciparum parasites have hindered efforts to eliminate malaria. Monitoring the spread of drug resistance is vital, as drug resistance can lead to widespread treatment failure. We develop a Bayesian model to produce spatio-temporal maps that depict the spread of drug resistance, and apply our methods for the antimalarial sulfadoxine-pyrimethamine. We infer from genetic count data the prevalences over space and time of various malaria parasite haplotypes associated with drug resistance. Previous work has focused on inferring the prevalence of individual molecular markers. In reality, combinations of mutations at multiple markers confer varying degrees of drug resistance to the parasite, indicating that multiple markers should be modelled together. However, the reporting of genetic count data is often inconsistent as some studies report haplotype counts, whereas some studies report mutation counts of individual markers separately. In response, we introduce a latent multinomial Gaussian process model to handle partially reported spatio-temporal count data. As drug-resistant mutations are often used as a proxy for treatment efficacy, point estimates from our spatio-temporal maps can help inform antimalarial drug policies, whereas the uncertainties from our maps can help with optimizing sampling strategies for future monitoring of drug resistance.
Collapse
Affiliation(s)
- Yong See Foo
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| | - Jennifer A. Flegg
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| |
Collapse
|
17
|
Wong S, Flegg JA, Golding N, Kandanaarachchi S. Comparison of new computational methods for spatial modelling of malaria. Malar J 2023; 22:356. [PMID: 37990242 PMCID: PMC10664662 DOI: 10.1186/s12936-023-04760-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 10/18/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Geostatistical analysis of health data is increasingly used to model spatial variation in malaria prevalence, burden, and other metrics. Traditional inference methods for geostatistical modelling are notoriously computationally intensive, motivating the development of newer, approximate methods for geostatistical analysis or, more broadly, computational modelling of spatial processes. The appeal of faster methods is particularly great as the size of the region and number of spatial locations being modelled increases. METHODS This work presents an applied comparison of four proposed 'fast' computational methods for spatial modelling and the software provided to implement them-Integrated Nested Laplace Approximation (INLA), tree boosting with Gaussian processes and mixed effect models (GPBoost), Fixed Rank Kriging (FRK) and Spatial Random Forests (SpRF). The four methods are illustrated by estimating malaria prevalence on two different spatial scales-country and continent. The performance of the four methods is compared on these data in terms of accuracy, computation time, and ease of implementation. RESULTS Two of these methods-SpRF and GPBoost-do not scale well as the data size increases, and so are likely to be infeasible for larger-scale analysis problems. The two remaining methods-INLA and FRK-do scale well computationally, however the resulting model fits are very sensitive to the user's modelling assumptions and parameter choices. The binomial observation distribution commonly used for disease prevalence mapping with INLA fails to account for small-scale overdispersion present in the malaria prevalence data, which can lead to poor predictions. Selection of an appropriate alternative such as the Beta-binomial distribution is required to produce a reliable model fit. The small-scale random effect term in FRK overcomes this pitfall, but FRK model estimates are very reliant on providing a sufficient number and appropriate configuration of basis functions. Unfortunately the computation time for FRK increases rapidly with increasing basis resolution. CONCLUSIONS INLA and FRK both enable scalable geostatistical modelling of malaria prevalence data. However care must be taken when using both methods to assess the fit of the model to data and plausibility of predictions, in order to select appropriate model assumptions and parameters.
Collapse
Affiliation(s)
- Spencer Wong
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jennifer A Flegg
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Nick Golding
- Telethon Kids Institute, Perth Children's Hospital, 15 Hospital Ave, Nedlands, WA, 6009, Australia
- Curtin University, Kent St, Bentley, WA, 6102, Australia
| | | |
Collapse
|
18
|
Alruwaili M, Uwimana A, Sethi R, Murindahabi M, Piercefield E, Umulisa N, Abram A, Eckert E, Munguti K, Mbituyumuremyi A, Gutman JR, Sullivan DJ. Peripheral and Placental Prevalence of Sulfadoxine-Pyrimethamine Resistance Markers in Plasmodium falciparum among Pregnant Women in Southern Province, Rwanda. Am J Trop Med Hyg 2023; 109:1057-1062. [PMID: 37783456 PMCID: PMC10622487 DOI: 10.4269/ajtmh.23-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/22/2023] [Indexed: 10/04/2023] Open
Abstract
Intermittent preventive therapy during pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is recommended in areas of moderate to high malaria transmission intensity. As a result of the increasing prevalence of SP resistance markers, IPTp-SP was withdrawn from Rwanda in 2008. Nonetheless, more recent findings suggest that SP may improve birthweight even in the face of parasite resistance, through alternative mechanisms that are independent of antimalarial effects. The prevalence of single nucleotide polymorphisms in Plasmodium falciparum dihydropteroate synthase (pfdhps) and dihydrofolate reductase (pfdhfr) genes associated with SP resistance among 148 pregnant women from 2016 to 2018 within Rwanda's Southern Province (Huye and Kamonyi districts) was measured using a ligase detection reaction-fluorescent microsphere assay. The frequency of pfdhps K540E, A581G, and the quintuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E) and sextuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E + A581G) mutant genotypes was 90%, 38%, 75%, and 28%, respectively. No significant genotype difference was seen between the two districts, which are approximately 50 km apart. Observed agreements for matched peripheral to placental blood were reported and found to be 207 of 208 (99%) for pfdhfr and 239 of 260 (92%) for pfdhps. The peripheral blood sample did not miss any pfdhfr drug-resistant mutants or pfdhps except at the S436 loci. At this level of the sextuple mutant, the antimalarial efficacy of SP for preventing low birthweight is reduced, although overall SP still exerts a nonmalarial benefit during pregnancy. This study further reveals the need to intensify preventive measures to sustain malaria control in Rwanda to keep the overall incidence of malaria during pregnancy low.
Collapse
Affiliation(s)
- Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Aline Uwimana
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
| | - Reena Sethi
- Maternal and Child Survival Program/Jhpiego, Washington, District of Columbia
| | - Monique Murindahabi
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
- Roll Back Malaria, Ouagadougou, Burkina Faso
| | - Emily Piercefield
- U.S. President’s Malaria Initiative, Malaria Branch, U.S. Centers for Disease Control and Prevention, Kigali, Rwanda
| | - Noella Umulisa
- Maternal and Child Survival Program/Jhpiego, Kigali, Rwanda
| | | | - Erin Eckert
- RTI International, Washington, District of Columbia
| | - Kaendi Munguti
- U.S. President’s Malaria Initiative, U.S. Agency for International Development, Kigali, Rwanda
| | | | - Julie R. Gutman
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
19
|
Xu M, Hu YX, Lu SN, Idris MA, Zhou SD, Yang J, Feng XN, Huang YM, Xu X, Chen Y, Wang DQ. Seasonal malaria chemoprevention in Africa and China's upgraded role as a contributor: a scoping review. Infect Dis Poverty 2023; 12:63. [PMID: 37403183 PMCID: PMC10320994 DOI: 10.1186/s40249-023-01115-x] [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: 02/13/2023] [Accepted: 06/14/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Children under five are the vulnerable population most at risk of being infected with Plasmodium parasites, especially in the Sahel region. Seasonal malaria chemoprevention (SMC) recommended by World Health Organization (WHO), has proven to be a highly effective intervention to prevent malaria. Given more deaths reported during the COVID-19 pandemic than in previous years due to the disruptions to essential medical services, it is, therefore, necessary to seek a more coordinated and integrated approach to increasing the pace, coverage and resilience of SMC. Towards this end, fully leverage the resources of major players in the global fight against malaria, such as China could accelerate the SMC process in Africa. METHODS We searched PubMed, MEDLINE, Web of Science, and Embase for research articles and the Institutional Repository for Information Sharing of WHO for reports on SMC. We used gap analysis to investigate the challenges and gaps of SMC since COVID-19. Through the above methods to explore China's prospective contribution to SMC. RESULTS A total of 68 research articles and reports were found. Through gap analysis, we found that despite the delays in the SMC campaign, 11.8 million children received SMC in 2020. However, there remained some challenges: (1) a shortage of fully covered monthly courses; (2) lack of adherence to the second and third doses of amodiaquine; (3) four courses of SMC are not sufficient to cover the entire malaria transmission season in areas where the peak transmission lasts longer; (4) additional interventions are needed to consolidate SMC efforts. China was certified malaria-free by WHO in 2021, and its experience and expertise in malaria elimination can be shared with high-burden countries. With the potential to join the multilateral cooperation in SMC, including the supply of quality-assured health commodities, know-how transfer and experience sharing, China is expected to contribute to the ongoing scale-up of SMC. CONCLUSIONS A combination of necessary preventive and curative activities may prove beneficial both for targeted populations and for health system strengthening in the long run. More actions are entailed to promote the partnership and China can be one of the main contributors with various roles.
Collapse
Affiliation(s)
- Ming Xu
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Yun-Xuan Hu
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Shen-Ning Lu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, China
| | | | - Shu-Duo Zhou
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Jian Yang
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Xiang-Ning Feng
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Yang-Mu Huang
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China
- Institute for Global Health and Development, Peking University, Beijing, China
| | - Xian Xu
- Anhui Provincial Center for Disease Control and Prevention, Hefei, China
| | - Ying Chen
- Department of Global Health, School of Public Health, Peking University, Haidian District, 38 Xue Yuan Road, Beijing, 100191, China.
- Institute for Global Health and Development, Peking University, Beijing, China.
| | - Duo-Quan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), NHC Key Laboratory of Parasite and Vector Biology, WHO Collaborating Centre for Tropical Diseases, National Center for International Research On Tropical Diseases, Shanghai, China.
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
20
|
van Eijk AM, Stepniewska K, Hill J, Taylor SM, Rogerson SJ, Cottrell G, Chico RM, Gutman JR, Tinto H, Unger HW, Yanow SK, Meshnick SR, Ter Kuile FO, Mayor A. Prevalence of and risk factors for microscopic and submicroscopic malaria infections in pregnancy: a systematic review and meta-analysis. Lancet Glob Health 2023; 11:e1061-e1074. [PMID: 37276878 PMCID: PMC10880462 DOI: 10.1016/s2214-109x(23)00194-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Malaria infections during pregnancy can cause adverse birth outcomes, yet many infections are undetected by microscopy. We aimed to describe the epidemiology of submicroscopic malaria infections in pregnant women in Asia, the Americas, and Africa using aggregated and individual participant data (IPD). METHODS For this systematic review and meta-analysis, studies (published Jan 1, 1997 to Nov 10, 2021) with information on both microscopic and submicroscopic infections during pregnancy from Asia, the Americas, or Africa, identified in the Malaria-in-Pregnancy Library, were eligible. Studies (or subgroups or study groups) that selected participants on the basis of the presence of fever or a positive blood smear were excluded to avoid selection bias. We obtained IPD (when available) and aggregated data. Estimates of malaria transmission intensity and sulfadoxine-pyrimethamine resistance, matched by study location and year, were obtained using publicly available data. One-stage multivariable logit and multinomial models with random intercepts for study site were used in meta-analysis to assess prevalence of and risk factors for submicroscopic infections during pregnancy and at delivery. This study is registered with PROSPERO, number CRD42015027342. FINDINGS The search identified 87 eligible studies, 68 (78%) of which contributed to the analyses. Of these 68 studies, 45 (66%) studies contributed IPD (48 869 participants) and 23 (34%) studies contributed aggregated data (11 863 participants). During pregnancy, median prevalence estimates were 13·5% (range 0·0-55·9, 66 substudies) for submicroscopic and 8·0% (0·0-50·6, 66 substudies) for microscopic malaria. Among women with positive Plasmodium nucleic acid amplification tests (NAATs), the median proportion of submicroscopic infections was 58·7% (range 0·0-100); this proportion was highest in the Americas (73·3%, 0·0-100), followed by Asia (67·2%, 36·4-100) and Africa (56·5%, 20·5-97·7). In individual patient data analysis, compared with women with no malaria infections, those with submicroscopic infections were more likely to present with fever in Africa (adjusted odds ratio 1·32, 95% CI 1·02-1·72; p=0·038) but not in other regions. Among women with NAAT-positive infections in Asia and the Americas, Plasmodium vivax infections were more likely to be submicroscopic than Plasmodium falciparum infections (3·69, 2·45-5·54; p<0·0001). Risk factors for submicroscopic infections among women with NAAT-positive infections in Africa included older age (age ≥30 years), multigravidity, and no HIV infection. INTERPRETATION During pregnancy, submicroscopic infections are more common than microscopic infections and are associated with fever in Africa. Malaria control in pregnancy should target both microscopic and submicroscopic infections. FUNDING Bill & Melinda Gates Foundation through the Worldwide Antimalarial Resistance Network.
Collapse
Affiliation(s)
- Anna Maria van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Kasia Stepniewska
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Steve M Taylor
- Division of Infectious Diseases and Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Stephen J Rogerson
- Department of Infectious Diseases, Doherty Institute, The University of Melbourne, Melbourne, VIC, Australia
| | | | - R Matthew Chico
- Department of Disease Control, London School of Hygiene & Tropical Medicine, London, UK
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Sant-Unité de Recherche Clinique de Nanoro, Ouagadougou, Burkina Faso
| | - Holger W Unger
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Stephanie K Yanow
- School of Public Health, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Steven R Meshnick
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Alfredo Mayor
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça, Maputo, Mozambique
| |
Collapse
|
21
|
L'Episcopia M, Doderer-Lang C, Perrotti E, Priuli GB, Cavallari S, Guidetti C, Bernieri F, Menard D, Severini C. Polymorphism analysis of drug resistance markers in Plasmodium falciparum isolates from Benin. Acta Trop 2023; 245:106975. [PMID: 37348801 DOI: 10.1016/j.actatropica.2023.106975] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 06/24/2023]
Abstract
Like most countries in sub-Saharan African countries, Benin continues to bear a heavy malaria burden. In 2014, the National Malaria Control Programme (NMCP) changed its treatment policy, and recommended the use of artemisinin-based combination therapy (ACT) as first-line treatment for uncomplicated Plasmodium falciparum cases. The study presented here was conducted to investigate the impact of current antimalarial drug resistance on the country. Molecular surveillance targeting the Pfcrt, Pfmdr1, Pfkelch13, dhfr, and dhps genes was carried out on samples from patients positive for P. falciparum malaria by microscopy, LAMP and PCR diagnostic test. Molecular analysis was performed using targeted amplicon deep sequencing (TADS). In addition, the frequency of parasites with dual deletion of the histidine-rich protein 2 and 3 genes (pfhrp2 and pfhrp3), known to be responsible of the performance of HRP-based malaria rapid diagnostic tests (HRP-RDT), was estimated. Fifty-three falciparum samples collected at the Saint Jean de Dieu hospital in Tanguiéta, Benin, were tested. No Pfkelch13 validated or candidate artemisinin partial resistant variants were identified. A marked prevalence of Asn51Ile (N51I), Cys59Arg (C59R), and Ser108Asn (S108N) mutant alleles was found in the dhfr gene, representing the most frequent genotype (64%). Five-point mutations were detected in dhps, Ile431Val (I431V), Ser436Ala (S436A), Ala437Gly (A437G), Ala581Gly (A581G), Ala613Ser (A613S) of which the third was the most common (92%). No mutation was identified in dhps Lys540Glu (K540E). The quintuple mutant genotype resulting from the combination of the dhfr triple mutant (51I/59R/108N) with the dhps double mutant 436A/437G was detected at a frequency of 30%. Low levels of mutations in Pfcrt and no mutation at codon 86 in the Pfmdr1 DNA fragment were observed, whereas a high level of Tyr184Phe (Y184F) polymorphism in the Pfmdr1 gene was found. These results could be indicative, over a decade after the implementation of ACT therapy, of the return of chloroquine-sensitive but artemether-lumefantrine resistant falciparum genotypes in Benin. There was no evidence of HRP2 and HRP3 deletions. Data from the present study support the need for routine monitoring of molecular markers of antimalarial drug resistance as part of surveillance activities aimed to make informed treatment policy decisions at the national level.
Collapse
Affiliation(s)
| | - Cécile Doderer-Lang
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, F-67000 Strasbourg, France
| | - Edvige Perrotti
- Istituto Superiore di Sanità, Department of Infectious Diseases, Rome, Italy
| | | | | | | | | | - Didier Menard
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, F-67000 Strasbourg, France; CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, F-67000 Strasbourg, France; Institut Pasteur, Université de Paris, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - Carlo Severini
- Istituto Superiore di Sanità, Department of Infectious Diseases, Rome, Italy
| |
Collapse
|
22
|
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
|
23
|
Kakuru A, Jagannathan P. Can we reduce malaria in pregnancy and improve birth outcomes? Lancet 2023; 401:973-975. [PMID: 36913960 DOI: 10.1016/s0140-6736(23)00101-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/13/2022] [Indexed: 03/12/2023]
Affiliation(s)
- Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda; Department of Community and Public Health, Busitema University, Tororo, Uganda.
| | | |
Collapse
|
24
|
Lingani M, Zango SH, Valéa I, Samadoulougou S, Somé G, Sanou M, Kaboré B, Rouamba T, Sorgho H, Tahita MC, Derra K, Dramaix M, Tinto H, Donnen P, Robert A. Effects of maternal antenatal treatment with two doses of azithromycin added to monthly sulfadoxine-pyrimethamine for the prevention of low birth weight in Burkina Faso: an open-label randomized controlled trial. Malar J 2023; 22:101. [PMID: 36932435 PMCID: PMC10024378 DOI: 10.1186/s12936-023-04530-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Exposure during pregnancy to malaria and sexually-transmitted infections is associated with adverse birth outcomes including low birth weight (LBW). This study aimed at assessing if the adjunction of two doses of azithromycin to sulfadoxine-pyrimethamine for the intermittent preventive treatment of malaria in pregnancy can reduce LBW. METHODS A two parallel-groups, open-label randomized controlled trial involving pregnant women (16 to 35 years of age and 12 to 24 weeks of gestation as confirmed by last menstrual period or fundal height) was conducted in rural Burkina Faso. Women were assigned in a 1:1 ratio either to use azithromycin (1 g daily for 2 days) during the second and third trimesters of pregnancy plus monthly sulfadoxine-pyrimethamine (1500/75 mg) (SPAZ) (intervention) or to continue using a monthly sulfadoxine-pyrimethamine (1500/75 mg) (SP) (control). Primary outcome was a LBW (birth weight measured within 24 h after birth < 2500 g). Secondary outcomes including stillbirth, preterm birth or miscarriage are reported together with safety data. RESULTS A total of 992 pregnant women underwent randomization (496 per group) and 898 (90.5%) valid birth weights were available (450 in SPAZ and 448 in SP). LBW incidence was 8.7% (39/450) in SPAZ and 9.4% (42/448) in controls (p-value = 0.79). Compared with controls, pregnant women with SPAZ showed a risk ratio (RR) of 1.16 (95% confidence interval (CI 0.64-2.08]) for preterm births, 0.75 (95% CI 0.17-3.35) for miscarriage and 0.64 (95% CI 0.25-1.64) for stillbirths. No treatment-related serious adverse events (SAEs) have been observed, and there was no significant difference in the number of SAEs (13.5% [67/496] in SPAZ, 16.7% [83/496] in SP, p-value = 0.18) or AEs (17.1% [85/496] in SPAZ, 18.8% [93/496] in SP, p-value = 0.56). CONCLUSION Adequate prevention regimen with monthly sulfadoxine-pyrimethamine given to all pregnant women has been proved to reduce the risk of LBW in malaria endemic areas. Adding azithromycin to the regimen does not offer further benefits, as far as women receive a malaria prevention regimen early enough during pregnancy. Trial registration Pan African Clinical Trial Registry ( https://pactr.samrc.ac.za/Search.aspx ): PACTR201808177464681. Registered 21 August 2018.
Collapse
Affiliation(s)
- Moussa Lingani
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso.
- École de Santé Publique, Université Libre de Bruxelles. CP594, Route de Lennik 808, 1070, Brussels, Belgique.
| | - Serge H Zango
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Innocent Valéa
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Sékou Samadoulougou
- Evaluation Platform On Obesity Prevention, Quebec Heart and Lung Institute Research Center, Quebec City, QC, G1V 4G5, Canada
| | - Georges Somé
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Maïmouna Sanou
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Berenger Kaboré
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Herman Sorgho
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Marc C Tahita
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Karim Derra
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Michèle Dramaix
- École de Santé Publique, Université Libre de Bruxelles. CP594, Route de Lennik 808, 1070, Brussels, Belgique
| | - Halidou Tinto
- Institut de Recherche en Sciences de La Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 18, Nanoro, Burkina Faso
| | - Philippe Donnen
- École de Santé Publique, Université Libre de Bruxelles. CP594, Route de Lennik 808, 1070, Brussels, Belgique
| | - Annie Robert
- Epidemiology and Biostatistics Research Division, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Brussels B1.30.13, Clos Chapelle-Aux-Champs 30, B-1200, Brussels, Belgique
| |
Collapse
|
25
|
Briggs J, Murray M, Nideffer J, Jagannathan P. Sex-Linked Differences in Malaria Risk Across the Lifespan. Curr Top Microbiol Immunol 2023; 441:185-208. [PMID: 37695429 DOI: 10.1007/978-3-031-35139-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Despite the high burden of malaria worldwide, there is surprisingly scarce research on sex-based differences in malaria outside of pregnancy. A more thorough understanding of sexual dimorphism in malaria, and what underlies these sex-based differences, could elucidate the underlying mechanisms driving malaria pathogenesis and has the potential to inform malaria control efforts, including new vaccines. This review summarizes our current understanding of sex-based differences in the epidemiology of malaria across the lifespan, potential sex- or gender-based mechanisms driving these differences, and the knowledge gaps that need to be addressed.
Collapse
Affiliation(s)
- Jessica Briggs
- Department of Medicine, University of California, San Francisco, California, United States
| | - Margaret Murray
- Department of Medicine, University of California, San Francisco, California, United States
| | - Jason Nideffer
- Department of Medicine, Stanford University, Stanford, California, United States
| | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, California, United States.
| |
Collapse
|