1
|
Lingani M, Ouoba S, Rouamba T, Valea I, Sanou M, Samadoulougou S, Tinto H, Sorgho H. Sulfadoxine-pyrimethamine alone or with azithromycin for the intermittent preventive treatment of malaria in pregnancy: protocol for a systematic review and meta-analysis. BMJ Open 2025; 15:e093931. [PMID: 40398947 DOI: 10.1136/bmjopen-2024-093931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
INTRODUCTION Increasing Plasmodium resistance levels to sulfadoxine-pyrimethamine (SP) threaten the effectiveness of intermittent preventive treatment in pregnancy (IPTp) and have prompted the evaluation of alternative strategies. Azithromycin (AZ) could have add-on effects on malaria and treat sexually transmitted infections (STIs), both conditions described as major causes of adverse pregnancy outcomes (APO). Inconsistent findings on the utility of AZ for the prevention of APO were reported; however, thus far, no comprehensive meta-analytic synthesis of data has been published. This review aims to investigate the effects of SP+AZ administered in women as IPTp on the risk of low birth weight in malaria-endemic areas. METHODS AND ANALYSIS Eligible studies will be identified through a pre-established search strategy in several electronic databases (Medline, Cochrane Library, Web of Science, EMBASE, WHO International Clinical Trials Registry Platform, ClinicalTrials.gov and AJOL) and will comprise peer-reviewed papers reporting original data on the effects of SP+AZ on the risk of APO. Only randomised controlled trials published until 30 September 2024 in English or French will be included. IPTp with SP+AZ regimens (intervention) will be compared with IPTp with SP alone or with a placebo (control). As primary outcomes, data on the frequency of low birth weight will be collected. Secondary outcomes include the rates of stillbirth, preterm birth, miscarriage and neonatal death. Data will be extracted independently by two reviewers using a predefined extraction form. If the data quality allows for quantitative synthesis, a fixed-effects meta-analysis will be conducted if there is low inter-study heterogeneity. Otherwise, the random-effects meta-analysis will be conducted to take into account uncertainty in pooled estimates that could be due to inter-study heterogeneity. The review protocol was designed according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses Protocols (PRISMA-P) guidelines. ETHICS AND DISSEMINATION Ethical clearance is not needed as the data will be from already published studies in which informed consent and ethical approval were obtained by primary investigators. Our dissemination plan includes the publication in a peer-reviewed journal as well as conference presentations. PROSPERO REGISTRATION NUMBER CRD42020149592.
Collapse
Affiliation(s)
- Moussa Lingani
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| | - Serge Ouoba
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| | - Innocent Valea
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| | | | - Sékou Samadoulougou
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Laval University, Quebec city, Québec, Canada
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| | - Hermann Sorgho
- Institut de Recherche en Sciences de la Santé, Ouagadougou, Burkina Faso
- Unité de Recherche Clinique de Nanoro (URCN), Ouagadougou, Burkina Faso
| |
Collapse
|
2
|
Home JL, McFadden GI, Goodman CD. Resistance to apicoplast translational inhibitors in Plasmodium. Int J Parasitol Drugs Drug Resist 2025; 28:100597. [PMID: 40381412 DOI: 10.1016/j.ijpddr.2025.100597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025]
Abstract
The spread of drug-resistant Plasmodium threatens malaria control efforts. Thus, understanding the mechanisms of resistance is crucial for implementing effective treatments and prevention strategies. The prokaryote-like translational machinery encoded by the apicoplast is the apparent target of several antibiotics with antimalarial activity. Among them, doxycycline and clindamycin are widely used for malaria treatment and/or chemoprophylaxis. However, the mechanisms underlying Plasmodium resistance to apicoplast-targeting antibiotics, and the evolution of such resistance mechanisms, remain largely unknown. In this review, we summarise reported cases of resistance to apicoplast translational inhibitors uncovered in either laboratory or clinical settings. We highlight the potential evolutionary pathway of doxycycline resistance, explore why resistance to these antibiotics remains rare in the field, and assess whether expanding their use in malaria treatment and prevention is a viable strategy.
Collapse
Affiliation(s)
- Jessica L Home
- School of BioSciences University of Melbourne, VIC, 3010, Australia.
| | | | | |
Collapse
|
3
|
Kwansa BK, Atobrah D, Anaba EA, Kyere A, Kretchy IA. Sex of household head and trends in uptake of sulfadoxine-pyrimethamine intermittent preventive treatment for malaria during pregnancy: insights from secondary data in sub-Saharan Africa. DISCOVER SOCIAL SCIENCE AND HEALTH 2025; 5:43. [PMID: 40162126 PMCID: PMC11953170 DOI: 10.1007/s44155-025-00184-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
Background Malaria in pregnancy remains a serious public health concern in sub-Saharan Africa. The household head as a primary decision-maker plays a major role in women's utilization of maternal health services. This study aimed to examine the trends, and the association between the sex of household head and the uptake of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP) in six sub-Saharan African countries. The findings provide insight into the progress, status and gender-specific barriers to IPTp-SP uptake. Methods Secondary data from the most recent Malaria Indicator Surveys for the six countries were analysed. A total of 15,452 (weighted) women aged 15-49 years from the six countries were included in this study. Both descriptive and inferential statistics were computed, including a chi-square test and binary logistic regression. Results The pooled data showed that 77% of the participants took at least one dose of IPTp-SP and 37% took ≥ 3 doses. The trend analysis showed that the uptake of IPTp-SP has increased over time. Women with a female household head (AOR = 1.21, 95% CI 1.08-1.38) had higher odds of taking ≥ 3 doses of IPTp-SP compared to those with a male household head. Conclusion The findings suggest that promoting women's participation in decision-making and leadership at the household level may help increase the uptake of IPTp-SP in sub-Saharan Africa.
Collapse
Affiliation(s)
- Benjamin Kobina Kwansa
- Institute of African Studies, University of Ghana, Accra, Ghana
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
| | - Deborah Atobrah
- Institute of African Studies, University of Ghana, Accra, Ghana
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
| | - Emmanuel Anongeba Anaba
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
- School of Public Health, University of Ghana, Accra, Ghana
| | - Abena Kyere
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
| | - Irene Akwo Kretchy
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
- School of Pharmacy, University of Ghana, Accra, Ghana
| |
Collapse
|
4
|
Tshiongo JK, Khote FL, Kabena M, Mavoko HM, Kalonji-Mukendi T, Luzolo L, Schallig HDFH, Kayentao K, Mens PF, Lutumba P, Tinto H, Maketa V. Intermittent screening using ultra-sensitive malaria rapid diagnostic test and treatment with pyronaridine-artesunate compared to standard preventive treatment with sulfadoxine-pyrimethamine for malaria prevention in pregnant women in Kinshasa, DRC. Malar J 2025; 24:58. [PMID: 39985024 PMCID: PMC11846385 DOI: 10.1186/s12936-025-05260-6] [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: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND The declining effectiveness of Intermittent Preventive Treatment in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP) due to the emergence of Plasmodium falciparum resistance highlights the need for alternative malaria prevention strategies in pregnant women. A novel approach was proposed: screening with an ultra-sensitive rapid diagnostic test and treating positive with pyronaridine-artesunate (ISTp-uRDT-PA). This trial compared the impact of both strategies on maternal malaria and anaemia, abortion, intrauterine death, birth weight, preterm delivery. METHODS This non-inferiority trial, conducted in Kinshasa, enrolled pregnant women in their second and third trimesters. Participants in the IPTp-SP arm (n = 124) received SP at monthly antenatal visit as per guidelines, while those in the ISTp-uRDT-PA arm (n = 126) were screened monthly with an uRDT and treated with PA if positive. Primary outcomes included asymptomatic parasitaemia (uRDT positive without fever) or symptomatic parasitaemia (uRDT positive with fever or history of fever, and parasite density by microscopy during pregnancy. RESULTS Asymptomatic parasitaemia by uRDT during pregnancy was similar in both arms (20.8% in IPTp-SP vs 21.0% in ISTp-uRDT-PA). At delivery, asymptomatic parasitaemia was 51% higher in ISTp-uRDT-PA arm compared to IPTp-SP (cRR = 1.51 [95% CI 0.76-3.00], p = 0.24). Symptomatic parasitaemia by uRDT at delivery showed no significant difference. Malaria by microscopy at enrolment was detected in 34.4% of women. Malaria by microscopy during pregnancy was 9.6% in IPTp-SP and 10.1%. ISTp-uRDT-PA (p = 0.19), decreasing to 3.2% and 0.9%, respectively, at delivery (p = 0.24). Mean haemoglobin concentration at enrolment was 10.1 g/dl in the IPTp-SP and 9.8 g/dl in the ISTp-uRDT-PA with no significant difference in maternal anaemia at delivery (7%; cRR = 1.07 [95% CI 0.87-1.31], p = 0.52). No significant differences were found for spontaneous abortions and in utero death in both arms. The risk of a premature newborn declined by 14% in ISTp-uRDT-PA compared to the IPTp-SP arm (cRR = 0.86 [95% CI 0.29-2.85], p = 0.79) while low-birth-weight was not significantly higher (cRR = 1.74 [95% CI 0.86-3.53], p = 0.12). CONCLUSION ISTp-uRDT-PA was non inferior to IPTp-SP and can be considered as a future alternative for IPTp-SP in case this intervention can no longer be used due to high SP resistance. CLINICAL TRIALS REGISTRATION NCT04783051.
Collapse
Affiliation(s)
- Japhet Kabalu Tshiongo
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo.
- Laboratory for Experimental Parasitology, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centre, Amsterdam, The Netherlands.
- Amsterdam Institute for Immunology and Infectious Diseases, Infectious Diseases Programme, Amsterdam, The Netherlands.
| | - Flory Luzolo Khote
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Melissa Kabena
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Hypolite Muhindo Mavoko
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Thierry Kalonji-Mukendi
- Programme National de Lutte Contre le Monkeypox et les Fièvres Hémorragiques Virales, Ministère de la Santé (PNLMPX-FHV), Kinshasa, Democratic Republic of the Congo
| | - Landrine Luzolo
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Henk D F H Schallig
- Laboratory for Experimental Parasitology, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Infectious Diseases Programme, Amsterdam, The Netherlands
- Amsterdam Institute for Global Health and Development, Amsterdam, The Netherlands
| | - Kassoum Kayentao
- Malaria Research and Training Center (MRTC), University of Sciences of Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Petra F Mens
- Laboratory for Experimental Parasitology, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Infectious Diseases Programme, Amsterdam, The Netherlands
| | - Pascal Lutumba
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
- Institut Supérieur des Techniques Médicales de Kinshasa (ISTM-Kinshasa), Kinshasa, Democratic Republic of the Congo
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé-Clinical Research Unit of Nanoro (IRSS-CRUN), Nanoro, Burkina Faso
| | - Vivi Maketa
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
5
|
Kretchy IA, Atobrah D, Adumbire DA, Ankamah S, Adanu T, Badasu DM, Kwansa BK. Enhancing the uptake of intermittent preventive treatment for malaria in pregnancy: a scoping review of interventions and gender-informed approaches. Malar J 2025; 24:49. [PMID: 39966899 PMCID: PMC11837586 DOI: 10.1186/s12936-025-05275-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: 09/20/2024] [Accepted: 01/29/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Malaria infection in pregnancy is a critical determinant of maternal and neonatal health outcomes in endemic regions. Intermittent preventive treatment of malaria in pregnancy (IPTp) using sulfadoxine-pyrimethamine has been recommended by the World Health Organization (WHO), but its uptake remains low because of factors such as gender norms and expectations. However, interventions to optimize IPTp uptake, especially in malaria-endemic regions, have resulted in a decline in malaria during pregnancy, maternal and neonatal mortality, low birth weight, and placental parasitaemia. This scoping review aimed to synthesize evidence on IPTp uptake, particularly emphasizing gender-related strategies. METHODS The modified version of Arksey and O'Malley's framework and the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews (PRISMA-ScR) were adopted for this review. Documents were retrieved from the following electronic databases and search engines: scopus, Web of Science, CINAHL Complete (EBSCO), PubMed, WHO, Global Index Medicus, and Google Scholar. The titles and abstracts of the publications were independently screened via Rayyan review management software, and the data were organized using the reach, effectiveness, adoption, implementation, and maintenance (RE-AIM) framework and gender analysis matrix. RESULTS A total of 32 studies met the inclusion criteria. The most reported criterion was the effectiveness of the interventions, which demonstrated an increase in IPTp uptake after the intervention. The gender analysis framework revealed that involving both men and women in decision-making processes, empowering women, and promoting shared roles could improve the success of IPTp interventions. CONCLUSIONS Interventions to increase IPTp uptake should be targeted at empowering women through education, increasing financial independence, and making decisions about their health.
Collapse
Affiliation(s)
- Irene A Kretchy
- Department of Pharmacy Practice and Clinical Pharmacy, University of Ghana School of Pharmacy, Accra, Ghana
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
| | - Deborah Atobrah
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
- Institute of African Studies, University of Ghana, Accra, Ghana
| | - David A Adumbire
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
- Regional Institute for Population Studies, University of Ghana, Accra, Ghana
| | - Samuel Ankamah
- University of Ghana Library System, University of Ghana, Accra, Ghana
| | - Theodosia Adanu
- University of Ghana Library System, University of Ghana, Accra, Ghana
| | - Delali M Badasu
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana
- Regional Institute for Population Studies, University of Ghana, Accra, Ghana
| | - Benjamin K Kwansa
- Centre for Gender Studies and Advocacy, University of Ghana, Accra, Ghana.
- Institute of African Studies, University of Ghana, Accra, Ghana.
| |
Collapse
|
6
|
Ter Kuile FO. Expanding seasonal malaria chemoprevention beyond the Sahel region. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00766-7. [PMID: 39826560 DOI: 10.1016/s1473-3099(24)00766-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK.
| |
Collapse
|
7
|
Lufele E, Pascoe S, Mengi A, Auwun A, Neuendorf N, Bolnga JW, Laman M, Rogerson SJ, Thriemer K, Unger HW. Acceptability of intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine plus dihydroartemisinin-piperaquine in Papua New Guinea: a qualitative study. Malar J 2025; 24:13. [PMID: 39806485 PMCID: PMC11731547 DOI: 10.1186/s12936-024-05233-1] [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: 07/24/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND In moderate-to-high malaria transmission regions, the World Health Organization recommends intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) alongside insecticide-treated bed nets to reduce the adverse consequences of pregnancy-associated malaria. Due to high-grade Plasmodium falciparum resistance to SP, novel treatment regimens need to be evaluated for IPTp, but these increase pill burden and treatment days. The present qualitative study assessed the acceptability of IPTp-SP plus dihydroartemisinin-piperaquine (DP) in Papua New Guinea, where IPTp-SP was implemented in 2009. METHODS Individual in-depth interviews (IDIs) and focus group discussions were conducted at health facilities where a clinical trial evaluated IPTp-SP plus DP (three-day regimen) versus IPTp-SP plus DP-placebo. IDIs were conducted with: (1) trial participants at different stages of engagement with ANC and IPTp, e.g. first antenatal clinic visit, subsequent antenatal clinic visits and postpartum; (2) local health workers (nurses, community health workers, midwives, health extension officers, doctors); and (3) representatives of district, provincial and national health authorities involved in programming ANC and IPTp. Focus group discussions comprised pregnant women only, including those engaged in the clinical trial and those receiving routine ANC outside of the trial. All interviews were audio recorded and transcribed. Transcripts were analysed using inductive and deductive thematic analysis applying a framework assessing: affective attitude, burden, ethicality, intervention coherence, opportunity costs, perceived effectiveness, and self-efficacy. RESULTS Women expressed positive feelings and attitudes towards SP plus DP/DP-placebo; reported limited side effects; and found the size, number, colour, and taste of study medicines acceptable. Health workers and policymakers were concerned that, compared to SP alone, additional tablets, frequency (three-day regimen), and tablet size might be barriers to acceptability for users outside a non-trial setting. There was a high perceived effectiveness of SP plus DP; most women reported that they did not get malaria or felt sick during pregnancy. Broader healthcare benefits received through trial participation and the involvement of health workers, relatives and community members in the clinical trial enabled antenatal clinic attendance and perceived acceptability of this IPTp regimen. CONCLUSIONS In the trial context, IPTp-SP plus DP was acceptable to both users and providers. Healthcare providers were concerned about the realities of acceptability and adherence to SP plus DP outside a clinical trial setting.
Collapse
Affiliation(s)
- Elvin Lufele
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia.
- Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea.
| | - Sophie Pascoe
- Wellbeing and Preventable Chronic Disease Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Alice Mengi
- Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Alma Auwun
- Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Nalisa Neuendorf
- Population Health Unit, Papua New Guinea Institute of Medical Research, Goroka, Eastern Highlands Province, Papua New Guinea
| | - John W Bolnga
- Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
- Department of Obstetrics and Gynaecology, Madang General Hospital, Madang, Madang Province, Papua New Guinea
| | - Moses Laman
- Vector-Borne Diseases Unit, Papua New Guinea Institute of Medical Research, Madang, Madang Province, Papua New Guinea
| | - Stephen J Rogerson
- Department of Infectious Diseases, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia
| | - Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Holger W Unger
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Department of Infectious Diseases, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia
| |
Collapse
|
8
|
Jafari-Guemouri S, Dégbègni R, Courtois L, Accrombessi M, Massougbodji A, Ding XC, Ndam NT, Mama A, Fievet N, Sarrasin-Hubert V, Cotrell G, Briand V. Dynamics of Persistent Submicroscopic and Microscopic Plasmodium falciparum in Pregnant Women Under Intermittent Preventive Treatment: A Study Cohort in Benin. Open Forum Infect Dis 2025; 12:ofae762. [PMID: 39829636 PMCID: PMC11739809 DOI: 10.1093/ofid/ofae762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Background Malaria infections in pregnancy are a major cause of maternal morbidity and neonatal mortality in sub-Saharan Africa. A high proportion of these infections are submicroscopic, which are usually asymptomatic and therefore untreated during pregnancy. Intermittent preventive treatment with sulfadoxine-pyrimethamine (IPTp-SP) aims to prevent and treat all potential infections whether submicroscopic or not. However, the resistance of parasites to SP is steadily increasing. The dynamic of microscopic and submicroscopic infections in a cohort of Beninese women throughout their pregnancy and its relation to IPTp-SP has been assessed. Methods As a subsample of the RECIPAL project, 130 women with at least 2 infections detected by polymerase chain reaction during their pregnancy were included. Infections were categorized as new (isolated) or persistent based on msp-2 genotyping, where persistent infections had identical genotypes in all studied time points. Submicroscopic infections were defined as polymerase chain reaction-positive and thick blood smear-negative. The persistence of infections according to IPTp-SP uptake was assessed. Results A total of 73.1% of women (95 women of 130) had exclusively persistent infections throughout their pregnancy, whereas only 7.7% (10 of 130) had exclusively new infections. During pregnancy, the median time spent with 1 persistent infection was 7.2 weeks. A considerable proportion of these persistent infections 64.3% (72 of 113) was only submicroscopic. Approximately 20% of these persistent infections occurred despite the use of IPTp-SP. Conclusions Using new antimalarial combinations could contribute to limit the persistence of submicroscopic infections and their probable negative effects on the mother and the fetus.
Collapse
Affiliation(s)
| | | | - Laura Courtois
- Genetics Department, Institut Curie, PSL Research University, Paris, France
| | - Manfred Accrombessi
- Clinical Research Institute of Benin (IRCB), Abomey-Calavi, Benin
- Faculty of Infectious and Tropical Diseases, Disease Control Department, London School of Hygiene and Tropical Medicine, London, Royaume-Uni
| | | | | | | | - Atika Mama
- Clinical Research Institute of Benin (IRCB), Abomey-Calavi, Benin
| | - Nadine Fievet
- UMR261 MERIT, Université Paris Cité, IRD, Paris, France
| | | | | | - Valérie Briand
- UMR261 MERIT, Université Paris Cité, IRD, Paris, France
- Epicentre, Paris, France
| |
Collapse
|
9
|
Gill J, Anvikar AR. New Strides in Prevention of Malaria during Pregnancy Present Multitudinous Opportunities. ACS Infect Dis 2024; 10:3721-3735. [PMID: 39405402 DOI: 10.1021/acsinfecdis.4c00566] [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] [Indexed: 11/09/2024]
Abstract
Pregnant women are at a higher risk of developing complications from malaria, a mosquito-borne disease caused by Plasmodium parasites, resulting in considerable maternal and infant morbidity and mortality. Malaria in pregnancy causes unfavorable and life-threatening outcomes for both the mother and fetus not limited to maternal anemia, hypoglycaemia, cerebral malaria, pulmonary edema, and puerperal sepsis. WHO recommends wide-ranging strategies for this detrimental but preventable disease; however, numerous challenges persist in ensuring high uptake of preventive therapies, effective usage of insecticide-treated bed nets, and early initiation and optimal antenatal care coverage for pregnant women. This work distils recent global advances in preventive strategies for malaria in pregnancy. We discuss three mainstay interventions by WHO, viz. intermittent preventive treatment of malaria in pregnancy (IPTp), utilization and outcomes of insecticide-treated bed nets (ITNs), and headways in malaria case management using therapeutic drugs. We cover multitudinous facets of antenatal care, WHO-advised community-based delivery of IPTp (c-IPTp), intermittent screening and treatment for malaria in pregnancy (ISTp), a malaria vaccine for pregnant women, and auxiliary factors that are crucial for improving prevention outcomes. Despite the reduction in malaria globally, malaria in pregnancy remains a prevalent issue in endemic areas, which warrants strengthening of preventative strategies. This work attempts to consolidate pivotal observations of the prevention of malaria during pregnancy by highlighting key advances, priority areas, new opportunities, research gaps, and challenges that need to be addressed to ensure improved outcomes in pregnant women infected with malaria.
Collapse
Affiliation(s)
- Jasmita Gill
- ICMR─National Institute of Malaria Research, Sector 8 Dwarka, New Delhi 110077, India
| | - Anupkumar R Anvikar
- ICMR─National Institute of Malaria Research, Sector 8 Dwarka, New Delhi 110077, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| |
Collapse
|
10
|
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
|
11
|
Tong Y, Ratnasiri K, Hanif S, Nguyen AT, Roh ME, Dorsey G, Kakuru A, Jagannathan P, Benjamin-Chung J. Intermittent preventive treatment for malaria in pregnancy and infant growth: a mediation analysis of a randomised trial. EBioMedicine 2024; 109:105397. [PMID: 39418986 PMCID: PMC11530852 DOI: 10.1016/j.ebiom.2024.105397] [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: 06/21/2024] [Revised: 09/18/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Intermittent preventive treatment for malaria in pregnancy (IPTp) can improve birth outcomes, but whether it confers benefits to postnatal growth is unclear. We investigated the effect of IPTp on infant growth in Uganda and its pathways of effects using causal mediation analyses. METHODS We analysed data from 633 infants born to mothers enrolled in a randomised trial of monthly IPTp with dihydroartemisinin-piperaquine (DP) vs. sulfadoxine-pyrimethamine (SP) (NCT02793622). Weight and length were measured from 0 to 12 months of age. Using generalised linear models, we estimated effects of DP vs. SP on gravidity-stratified mean length-for-age (LAZ) and weight-for-length Z-scores (WLZ). We investigated mediation by placental malaria, gestational weight change, maternal anaemia, maternal inflammation-related proteins, preterm birth, birth length, and birth weight. Mediation models adjusted for infant sex, gravidity, gestational age at enrolment, maternal age, maternal parasitaemia at enrolment, education, and wealth. FINDINGS SP increased mean LAZ by 0.18-0.28 Z from birth through age 4 months compared to DP, while DP increased mean WLZ by 0.11-0.28 Z from 2 to 8 months compared to SP among infants of multigravidae; at these ages, confidence intervals for mean differences excluded 0. We did not observe differences among primigravida. Mediators of SP included birth weight, birth length, maternal stem cell factor, and DNER. Mediators of DP included placental malaria and birth length, maternal IL-18, CDCP1, and CD6 at delivery. INTERPRETATION In high malaria transmission settings, this exploratory study suggests different IPTp regimens may influence infant growth among multigravidae, potentially through distinct pathways, in the exclusive breastfeeding period, when few other interventions are available. FUNDING Stanford Center for Innovation in Global Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Yanwei Tong
- Department of Statistics, Stanford University, Stanford, United States
| | - Kalani Ratnasiri
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States; Department of Microbiology and Immunology, Stanford University, Stanford, United States
| | - Suhi Hanif
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
| | - Anna T Nguyen
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
| | - Michelle E Roh
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, United States; Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, United States
| | - Grant Dorsey
- Department of Medicine, Division of HIV, ID, and Global Medicine, University of California San Francisco, San Francisco, United States
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Jade Benjamin-Chung
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States; Chan Zuckerberg Biohub, San Francisco, United States.
| |
Collapse
|
12
|
Tzitiridou-Chatzopoulou M, Zournatzidou G, Orovou E, Lavasidis L, Tsiotsias A, Eskitzis P, Papoutsis D. Intermittent Preventive Treatment of Malaria in Pregnancy and the Impact on Neonates in African Countries as Assessed by Entropy Weight and TOPSIS Methods. J Clin Med 2024; 13:6231. [PMID: 39458181 PMCID: PMC11508258 DOI: 10.3390/jcm13206231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: In regions of Africa with a high prevalence of malaria, pregnant women in their first or second trimester should be administered intermittent preventive treatment in pregnancy (IPTp). However, infants may contract malaria despite the IPTp therapy that their mothers have received. The objective of the present investigation was to assess the symptoms and various treatments for neonatal malaria. Methods: Entropy weight and TOPSIS were used to achieve the study goal. The TOPSIS multi-attribute decision-making system was used to assess newborn malaria symptoms and select the optimal treatment, even for mothers receiving IPTp medication during pregnancy. The entropy weight approach calculated TOPSIS attribute weights. The present research used UNICEF data for 14 African nations in 2023. Results: The results indicated that neonates whose mothers received IPTp therapy ultimately contracted malaria, with diarrhea being the primary symptom. It is important to note that health providers administer a combination of zinc and oral rehydration solution (ORS) to infants as the most effective treatment for malaria symptoms, thereby abandoning the first-line treatment for malaria, artemisinin-based combination therapy (ACT). Conclusions: The most effective treatment for neonatal malaria is a combination of zinc and ORS, although less than half of children in Africa have access to ORS. Therefore, the findings of this study may encourage African countries to prioritize co-pack therapy in their procurement and supply, healthcare provider training, and expenditures. This therapy will also help alleviate the symptoms of malaria in neonates.
Collapse
Affiliation(s)
- Maria Tzitiridou-Chatzopoulou
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| | - Georgia Zournatzidou
- Department of Business Administration, University of Western Macedonia, 51100 Grevena, Greece
| | - Eirini Orovou
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| | - Lazaros Lavasidis
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| | - Arsenios Tsiotsias
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| | - Panagiotis Eskitzis
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| | - Dimitrios Papoutsis
- Department of Midwifery, School of Healthcare Sciences, University of Western Macedonia, Keptse, 50200 Ptolemaida, Greece; (M.T.-C.); (E.O.); (L.L.); (A.T.); (P.E.); (D.P.)
| |
Collapse
|
13
|
Bohissou FET, Sondo P, Inoue J, Rouamba T, Kaboré B, Nassa GJW, Kambou AES, Traoré TE, Asua V, Borrmann S, Tinto H, Held J. Evolution of Pfdhps and Pfdhfr mutations before and after adopting seasonal malaria chemoprevention in Nanoro, Burkina Faso. Sci Rep 2024; 14:24224. [PMID: 39414909 PMCID: PMC11484836 DOI: 10.1038/s41598-024-75369-2] [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: 06/04/2024] [Accepted: 10/04/2024] [Indexed: 10/18/2024] Open
Abstract
Seasonal Malaria Chemoprevention consisting of monthly administration of amodiaquine/sulfadoxine-pyrimethamine to children aged 3-59 months during the transmission season could promote SP-resistance. Mutations in dihydrofolate reductase (Pfdhfr) and dihydropteroate synthase (Pfdhps) genes were assessed before and after SMC adoption in Burkina Faso. A total of 769 dried blood spots were selected from studies conducted in Nanoro, Burkina Faso, between 2010 and 2020. Of those, 299 were pre-SMC (2010-2012) and 470 were post-SMC-samples. Pfdhps and Pfdhfr genes were PCR-amplified and sequenced. A systematic review/meta-analysis of published studies conducted in Burkina Faso (2009-2023) was additionally performed. In Nanoro, the prevalence of Pfdhfr triple mutations (CIRNI) rose from 43.6% pre-SMC to 89.4% post-SMC (p < 0.0001). There was no mutation in Pfdhfr 164 and Pfdhps 540; Pfdhps A437G mutation increased from 63.9% (2010-2012) to 84.7% (2020) (p < 0.0001). The VAGKGS haplotype was 2.8% (2020). Pfdhfr/Pfdhps quintuple mutant IRN-436A437G rose from 18.6% (2010-2012) to 58.3% (2020) (p < 0.0001). Meta-analysis results from Burkina Faso showed an increase in mutations at Pfdhfr N51I, C59R, S108N, and Pfdhps A437G after SMC adoption. Post-SMC, the pyrimethamine-resistance marker prevalence increased, while the sulfadoxine-resistance marker prevalence remained stable. Detection of emerging PfdhpsVAGKGS haplotypes in 2020 underscores the importance of continuous SP-resistance monitoring.
Collapse
Affiliation(s)
- Francis Emmanuel Towanou Bohissou
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
- Centre de Recherche Entomologique de Cotonou (CREC), Cotonou, Benin
| | - Paul Sondo
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Juliana Inoue
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Toussaint Rouamba
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Berenger Kaboré
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | | | - A Elisée Sié Kambou
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Tiampan Edwig Traoré
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Victor Asua
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Steffen Borrmann
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, (CERMEL), Lambaréné, Gabon
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé (IRSS)/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso.
| | - Jana Held
- Institute of Tropical Medicine, University Hospital Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
- Centre de Recherches Médicales de Lambaréné, (CERMEL), Lambaréné, Gabon.
| |
Collapse
|
14
|
Barsosio HC, Webster J, Omiti F, K'Oloo A, Odero IA, Ojuok MA, Odiwa D, Omondi B, Okello E, Dodd J, Taegtmeyer M, Kuile FOT, Lesosky M, Kariuki S, Hill J. Delivery effectiveness of and adherence to intermittent preventive treatment for malaria in pregnancy with dihydroartemisinin-piperaquine with or without targeted information transfer or sulfadoxine-pyrimethamine in western Kenya: a three-armed, pragmatic, open-label, cluster-randomised trial. Lancet Glob Health 2024; 12:e1660-e1672. [PMID: 39304238 DOI: 10.1016/s2214-109x(24)00261-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND High-level resistance to sulfadoxine-pyrimethamine threatens the efficacy of WHO-recommended intermittent preventive treatment in pregnancy (IPTp) with single-dose sulfadoxine-pyrimethamine to prevent malaria. Monthly IPTp with dihydroartemisinin-piperaquine, a 3-day regimen, is an emerging alternative, but this regimen poses potential implementation and adherence challenges. We aimed to assess adherence to a multiday IPTp with dihydroartemisinin-piperaquine regimen and its delivery effectiveness in routine antenatal care settings in western Kenya. METHODS We conducted a pragmatic, three-armed, open-label, cluster-randomised trial in antenatal clinics in 18 health-care facilities (six facilities per group) in Kisumu County and Homa Bay County in western Kenya. Clusters were facilities offering routine antenatal care services provided by trained Ministry of Health staff with 100 or more antenatal clinic attendances per month between July, 2018, and June, 2019. Private or mission hospitals, dispensaries, referral hospitals, and trial sites were excluded. Individuals in their first trimester, living with HIV, or who were not attending a scheduled antenatal clinic visit were excluded. The 18 antenatal clinics were grouped into matched triplets stratified by location and clinics in each matched triplet were randomly assigned to one of the three study groups (1:1:1). Masking was not possible. Two groups were given IPTp with dihydroartemisinin-piperaquine (one group with a targeted information transfer intervention and one group without any additional interventions) and one group was given the standard of care (ie, IPTp with sulfadoxine-pyrimethamine). The primary endpoint, adherence, was defined as the proportion of participants completing their most recent 3-day IPTp with dihydroartemisinin-piperaquine regimen. This completion was verified by pill counts during home visits no more than 2 days after participants' 3-day regimens ended. The secondary endpoint, delivery effectiveness, was defined as the proportion of participants who received the correct number of IPTp tablets and correctly repeated dosing instructions (ie, correctly recalled the instructions they received about self-administered dihydroartemisinin-piperaquine doses and the number of sulfadoxine-pyrimethamine tablets they had received) at their exit from the antenatal clinic. Individuals receiving treatment for malaria, visiting a clinic for registration only, or interviewed during IPTp drug stock-outs were excluded from analyses. We used generalised linear mixed models to compare endpoints among the IPTp with dihydroartemisinin-piperaquine groups. This trial was registered with ClinicalTrials.gov, NCT04160026, and is complete. FINDINGS 15 facilities (five per group) completed the trial, with 1189 participants having exit interviews (377 in the IPTp with sulfadoxine-pyrimethamine group, 408 in the IPTp with dihydroartemisinin-piperaquine only group, and 404 in the IPTp with dihydroartemisinin-piperaquine plus targeted information transfer intervention group) and 586 participants having home visits (267 in the IPTp with dihydroartemisinin-piperaquine only group and 319 in the IPTp with dihydroartemisinin-piperaquine plus targeted information transfer intervention group) from Sept 8 to Dec 10, 2020. Relative to the IPTp with dihydroartemisinin-piperaquine only group, adherence was 16% higher in the IPTp with dihydroartemisinin-piperaquine plus targeted information transfer intervention group (266 [83%] of 319 participants vs 196 [73%] of 267 participants; adjusted relative risk [RR] 1·16, 95% CI 1·03-1·31; p=0·0140). Delivery effectiveness in the IPTp with dihydroartemisinin-piperaquine plus targeted information transfer intervention group was not significantly different from that in the IPTp with sulfadoxine-pyrimethamine group (352 [87%] of 403 participants vs 335 [89%] of 375 participants; adjusted RR 0·97, 95% CI 0·90-1·05; p=0·4810). However, delivery effectiveness in the IPTp with dihydroartemisinin-piperaquine only group was significantly lower than in the IPTp with sulfadoxine-pyrimethamine group (300 [74%] of 404 participants vs 335 [89%] of 375 participants; 0·84, 0·75-0·95; p=0·0030). INTERPRETATION Targeted information transfer interventions to health-care providers and pregnant individuals boost antenatal care delivery adherence to a multiday regimen with dihydroartemisinin-piperaquine. FUNDING European and Developing Countries Clinical Trials Partnership 2, UK Joint Global Health Trials Scheme of the Foreign, Commonwealth and Development Office, Medical Research Council, National Institute for Health and Care Research, and Wellcome Trust; and Swedish International Development Cooperation Agency.
Collapse
Affiliation(s)
- Hellen C Barsosio
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya.
| | - Jayne Webster
- Disease Control Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Frederick Omiti
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Alloys K'Oloo
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Isdorah A Odero
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Michael A Ojuok
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Dawn Odiwa
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Benson Omondi
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Elizabeth Okello
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Miriam Taegtmeyer
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK; Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Maia Lesosky
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
15
|
Asem L, Abdulia AG, Assuming PO, Abeka-Nkrumah G. Knowledge, and attitude of service user of intermittent preventive treatment of malaria in pregnancy using sulfadoxine pyrimethamine in the Volta Region of Ghana. PLoS One 2024; 19:e0309340. [PMID: 39240950 PMCID: PMC11379158 DOI: 10.1371/journal.pone.0309340] [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: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 09/08/2024] Open
Abstract
BACKGROUND Malaria in pregnancy (MiP) is a condition that can be prevented by using intermittent preventive treatment using Sulfadoxine-pyrimethamine. However, despite all the effort to reduce the consequences of MiP for the woman, the unborn child, and the neonate, the knowledge of Intermittent Preventive Treatment of Malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP) is low in most malaria-endemic countries, including Ghana. Thus, the need to examine knowledge, and attitude of service users of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine. METHODS The study was a cross-sectional survey of two selected districts in the Volta Region of Ghana. The study participants were randomly selected from communities within Nkwanta North and North Tongu District. In all a total of 438 mothers who have delivered in the past 24 months were selected for the study. The women were interviewed using a structured questionnaire and the bivariate and multivariable logistic regression results presented in tables. RESULTS The level of knowledge, and attitude were reported as 45.9% and 58.9% respectively. Knowledge of the service user is determined by the level of education of the women. The attitude of the service user is determined by making 4-7 visits during ANC, Gestational age at booking for ANC is 4-7 weeks, income level between 100 to 999, partner educational level above Middle/JHS/JSS, and age of a partner is above 40 years. CONCLUSION The findings from the present studies highlighted important factor such as number of antenatal visits that affect both knowledge of services and attitude to use IPTp-SP. Therefore, a community-based health promotion programmes to help to increase knowledges and improved attitude on timely and regular antenatal attendance to promote the benefit of IPTp-SP should be encouraged.
Collapse
Affiliation(s)
- Livingstone Asem
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Accra, Ghana
- Department of Health Policy Planning and Management, School of Public Health, University of Health Allied Sciences, Ho, Ghana
| | - Abdul-Gafaru Abdulia
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Accra, Ghana
| | - Patrick Opoku Assuming
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Accra, Ghana
| | - Gordon Abeka-Nkrumah
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Accra, Ghana
| |
Collapse
|
16
|
Asem L, Abdulia AG, Assuming PO, Abeka-Nkrumah G. The demand for intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine in the Volta Region of Ghana. PLoS One 2024; 19:e0308321. [PMID: 39236029 PMCID: PMC11376531 DOI: 10.1371/journal.pone.0308321] [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: 05/28/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Malaria in pregnancy (MiP) is a preventable condition leading to maternal and neonatal morbidity and mortality. Invariably, with all the knowledge about the serious consequences of MiP for the woman, the unborn child, and the neonate, the uptake of Intermittent Preventive Treatment of Malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP) is low in most malaria-endemic countries, including Ghana. This study sought to examine the uptake and service user predictors of the implementation of IPTp-SP after the policy upgrade in 2014. METHODS This cross-sectional survey was carried out in two selected districts in the Volta Region. The study participants were randomly selected from communities within Nkwanta North and North Tongu District. A total of 438 mothers who have delivered in the past 24 months were selected for the study. The women were interviewed on their background, knowledge, and attitude toward the use of IPTp-SP using a structured questionnaire. Multiple logistic regression was done to determine the factors that influence the demand for IPTp-SP. The results were presented in the form of tables. RESULTS The mean number of antenatal care (ANC) attendance was 5 (SD:2.6) visits per client, with 262 (59.82%) of them getting the 3+ doses of IPTp-SP. Also, a significant 44 (10.1%) of the mothers did not receive any dose of IPTp-SP. Respondents who attended antenatal clinics 4-7 times had 7 (CI:3.9-12.3) times higher uptake of 3+ doses of IPTp-SP as compared to others who attended less than 4 visits. Similarly, women who had 8 or more visits had a 16.1 (CI: 5.9-43.6) times higher chance of getting more than 2 doses of IPTp-SP compared with others who had fewer than 4 attendances. CONCLUSION The uptake of 3+ doses of IPTp-SP is still lower than the global target of 80%. Thus, the need for innovative interventions aimed at improving antenatal attendance and early booking for IPTp-SP are recommended.
Collapse
Affiliation(s)
- Livingstone Asem
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Legon-Accra, Ghana
- Deparment of Health Policy Planning and Management, Fred N. Binka School of Public Health, University of Health and Allied Sciences, Ho, Ghana
| | - Abdul-Gafaru Abdulia
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Legon-Accra, Ghana
| | - Patrick Opoku Assuming
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Legon-Accra, Ghana
| | - Gordon Abeka-Nkrumah
- Department of Public Administration and Health Services Management, Business School, University of Ghana, Legon-Accra, Ghana
| |
Collapse
|
17
|
Berchie GO, Doe PF, Azu TD, Agyeiwaa J, Owusu G, Boso CM, Yeboa NK, Agyare DF, Aboh IK, Nabe B, Ofori GO, Anumel B, Kagbo JE, Alhassan A, Offei FO, Opoku-Danso R, Abraham SA, Amoadu M, Hagan JE. Uptake and Effectiveness of Intermittent Preventive Treatment with Sulfadoxine-Pyrimethamine during Pregnancy in Africa: A Scoping Review. Diseases 2024; 12:203. [PMID: 39329872 PMCID: PMC11431087 DOI: 10.3390/diseases12090203] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/16/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Malaria poses a significant threat to pregnant women in sub-Saharan Africa, necessitating effective interventions like the intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP). However, challenges persist in the uptake and effectiveness of this intervention. This scoping review aims to explore IPTp-SP uptake in African countries, identify influencing factors, and assess its effectiveness in preventing malaria and adverse outcomes in pregnancy. This scoping review follows Arksey and O'Malley's framework, employing the PRISMA-ScR guidelines for reporting. Searches were conducted in PubMed, Embase, Scopus, JSTOR, Web of Science, Google Scholar, and ProQuest, focusing on studies post-2000 published in the English language. The search produced 15,153 records, of which 104 full-text records were eligible and 101 papers were included in this review. The findings suggest varying IPTp-SP uptake rates, spanning from 5.3% to 98.9%, with their effectiveness supported by longitudinal studies, randomised controlled-trials (RCTs), cross-sectional surveys, and mixed-method studies. IPTp-SP demonstrates efficacy in reducing malaria during pregnancy, placental parasitaemia, and anaemia episodes, alongside improved birth outcomes. Common adverse effects of IPTp-SP include prematurity and low birth weight. Facilitators of IPTp-SP uptake include education and ANC attendance, while commonly reported barriers included inadequate knowledge and healthcare system challenges. The findings also suggest adverse effects such as prematurity, low birth weight, and maternal and perinatal mortality associated with IPTp-SP uptake. It is vital to strengthen antenatal care services by integrating comprehensive counselling on IPTp-SP and address healthcare system challenges. Community engagement, women's empowerment, and context-specific interventions are necessary for promoting IPTp-SP uptake and improving maternal and neonatal health outcomes in Africa.
Collapse
Affiliation(s)
- Gifty Osei Berchie
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Patience Fakornam Doe
- Department of Public Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (P.F.D.); (S.A.A.)
| | - Theodora Dedo Azu
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Joyce Agyeiwaa
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Gifty Owusu
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Christian Makafui Boso
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Naomi Kyeremaa Yeboa
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Dorcas Frempomaa Agyare
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Irene Korkoi Aboh
- Department of Maternal and Child Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (G.O.B.); (T.D.A.); (J.A.); (G.O.); (N.K.Y.); (I.K.A.)
| | - Bernard Nabe
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Godson Obeng Ofori
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Benjamin Anumel
- Center for Health Research and Policy Innovations, Legon, Accra P.O. Box LG 949, Ghana;
| | - Justice Enock Kagbo
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Amidu Alhassan
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Frank Odonkor Offei
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Rita Opoku-Danso
- Department of Adult Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (C.M.B.); (D.F.A.); (B.N.); (G.O.O.); (J.E.K.); (A.A.); (F.O.O.); (R.O.-D.)
| | - Susanna Aba Abraham
- Department of Public Health, School of Nursing and Midwifery, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana; (P.F.D.); (S.A.A.)
| | - Mustapha Amoadu
- Biomedical and Clinical Research Centre, College of Health and Allied Sciences, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana;
| | - John Elvis Hagan
- Department of Health, Physical Education and Recreation, University of Cape Coast, PMB, Cape Coast CC 3321, Ghana
- Neurocognition and Action-Biomechanics-Research Group, Faculty of Psychology and Sports Science, Bielefeld University, Postfach 10 01 31, 33501 Bielefeld, Germany
| |
Collapse
|
18
|
Azmi WA, Rizki AFM, Shidiq A, Djuardi Y, Artika IM, Siregar JE. Antimalarial drug sulfadoxine induces gametocytogenesis in Plasmodium berghei. Malar J 2024; 23:267. [PMID: 39223522 PMCID: PMC11367840 DOI: 10.1186/s12936-024-05071-1] [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: 06/03/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The spread of antimalarial drug resistance parasites is a major obstacle in eliminating malaria in endemic areas. This increases the urgency for developing novel antimalarial drugs with improved profiles to eliminate both sensitive and resistant parasites in populations. The invention of the drug candidates needs a model for sensitive and resistant parasites on a laboratory scale. METHODS Repeated Incomplete Treatment (RIcT) method was followed in raising the rodent malaria parasite, Plasmodium berghei, resistant to sulfadoxine. Plasmodium berghei were exposed to an adequate therapeutic dose of sulfadoxine without finishing the treatment to let the parasite recover. Cycles of drug treatment and parasite recovery were repeated until phenotypic resistance appeared. RESULTS After undergoing 3-4 cycles, phenotypic resistance was not yet found in mice treated with sulfadoxine. Nevertheless, the molecular biology of dhps gene (the target of sulfadoxine) was analyzed at the end of the RIcT cycle. There was no mutations found in the gene target. Interestingly, the appearance of gametocytes at the end of every cycle of drug treatment and parasite recovery was observed. These gametocytes later on would no longer extend their life in the RBC stage, unless mosquitoes bite the infected host. This phenomenon is similar to the case in human malaria infections treated with sulfadoxine-pyrimethamine (SP). CONCLUSIONS In this study, the antimalarial drug sulfadoxine induced gametocytogenesis in P. berghei, which could raise the risk factor for malaria transmission.
Collapse
Affiliation(s)
- Wihda Aisarul Azmi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor, 16911, Indonesia
- Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta, 10430, Indonesia
| | - Andita Fitri Mutiara Rizki
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor, 16911, Indonesia
| | - Achmad Shidiq
- Research Center for Preclinical and Clinical Medicine, National Research and Innovation Agency, Cibinong, Bogor, 16911, Indonesia
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine Universitas Indonesia, Jakarta, 10430, Indonesia
| | - I Made Artika
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Dramaga Campus, Bogor, 16680, Indonesia
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor, 16911, Indonesia.
| |
Collapse
|
19
|
Ranjit A, Wylie BJ. Malaria in Pregnancy, Current Challenges, and Emerging Prevention Strategies in a Warming Climate. Clin Obstet Gynecol 2024; 67:620-632. [PMID: 39061127 DOI: 10.1097/grf.0000000000000888] [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: 07/28/2024]
Abstract
Malaria still presents a grave threat to the health of pregnancies worldwide with prevention currently stalling as traditional control and prevention strategies are limited by both insecticide and drug resistance. Furthermore, climate change is bringing malaria to locations where it was once eradicated and intensifying malaria in other areas. Even where malaria is not currently common, obstetricians will need to understand the pathogenesis of the disease, how it is transmitted, methods for prevention and treatment in pregnancy, and promising emerging strategies such as vaccines. A renewed global response is needed for this age-old disease in which pregnancy poses specific susceptibility.
Collapse
Affiliation(s)
- Anju Ranjit
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of California, San Francisco
| | - Blair J Wylie
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Columbia University Medical Center, New York, New York
| |
Collapse
|
20
|
Fola AA, Ciubotariu II, Dorman J, Mwenda MC, Mambwe B, Mulube C, Kasaro R, Hawela MB, Hamainza B, Miller JM, Bailey JA, Moss WJ, Bridges DJ, Carpi G. National genomic profiling of Plasmodium falciparum antimalarial resistance in Zambian children participating in the 2018 Malaria Indicator Survey. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.05.24311512. [PMID: 39148823 PMCID: PMC11326323 DOI: 10.1101/2024.08.05.24311512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The emergence of antimalarial drug resistance is a major threat to malaria control and elimination. Using whole genome sequencing of 282 P. falciparum samples collected during the 2018 Zambia National Malaria Indicator Survey, we determined the prevalence and spatial distribution of known and candidate antimalarial drug resistance mutations. High levels of genotypic resistance were found across Zambia to pyrimethamine, with over 94% (n=266) of samples having the Pfdhfr triple mutant (N51I, C59R, and S108N), and sulfadoxine, with over 84% (n=238) having the Pfdhps double mutant (A437G and K540E). In northern Zambia, 5.3% (n=15) of samples also harbored the Pfdhps A581G mutation. Although 29 mutations were identified in Pfkelch13, these mutations were present at low frequency (<2.5%), and only three were WHO-validated artemisinin partial resistance mutations: P441L (n=1, 0.35%), V568M (n=2, 0.7%) and R622T (n=1, 0.35%). Notably, 91 (32%) of samples carried the E431K mutation in the Pfatpase6 gene, which is associated with artemisinin resistance. No specimens carried any known mutations associated with chloroquine resistance in the Pfcrt gene (codons 72-76). P. falciparum strains circulating in Zambia were highly resistant to sulfadoxine and pyrimethamine but remained susceptible to chloroquine and artemisinin. Despite this encouraging finding, early genetic signs of developing artemisinin resistance highlight the urgent need for continued vigilance and expanded routine genomic surveillance to monitor these changes.
Collapse
Affiliation(s)
- Abebe A. Fola
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - Ilinca I. Ciubotariu
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jack Dorman
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Mulenga C. Mwenda
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Brenda Mambwe
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Conceptor Mulube
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Rachael Kasaro
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Moonga B. Hawela
- National Malaria Elimination Centre, Zambia Ministry of Health, Chainama Hospital Grounds, Lusaka, Zambia
| | - Busiku Hamainza
- National Malaria Elimination Centre, Zambia Ministry of Health, Chainama Hospital Grounds, Lusaka, Zambia
| | - John M. Miller
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Jeffrey A. Bailey
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
| | - William J. Moss
- The Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daniel J. Bridges
- PATH Malaria Control and Elimination Partnership in Africa (MACEPA), Lusaka, Zambia
| | - Giovanna Carpi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
- The Johns Hopkins Malaria Research Institute, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
21
|
Joste V, Coppée R, Bailly J, Rakotoarivony Y, Toko Tchokoteu FG, Achache S, Pradines B, Cottrell G, Ariey F, Khim N, Popovici J, Mita T, Groger M, Ramharter M, Egbo T, Juma DW, Akala H, Houzé S, Clain J. Plasmodium ovale spp dhfr mutations associated with reduced susceptibility to pyrimethamine in sub-Saharan Africa: a retrospective genetic epidemiology and functional study. THE LANCET. MICROBE 2024; 5:669-678. [PMID: 38761813 DOI: 10.1016/s2666-5247(24)00054-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Mutations in the Plasmodium falciparum dhfr gene confer resistance to pyrimethamine, which is widely used for malaria chemoprevention in Africa. We aimed to evaluate the frequency and evolution of dhfr mutations in Plasmodium ovale spp in Africa and their functional consequences, which are incompletely characterised. METHODS We analysed dhfr mutations and their frequencies in P ovale spp isolates collected between Feb 1, 2004, and Aug 31, 2023, from the French National Malaria Reference Centre collection and from field studies in Benin, Gabon, and Kenya. Genetic patterns of positive selection were investigated. Full-length recombinant wild-type and mutant DHFR enzymes from both P ovale curtisi and P ovale wallikeri were expressed in bacteria to test whether the most common mutations reduced pyrimethamine susceptibility. FINDINGS We included 518 P ovale spp samples (314 P ovale curtisi and 204 P ovale wallikeri). In P ovale curtisi, Ala15Ser-Ser58Arg was the most common dhfr mutation (39%; 124 of 314 samples). In P ovale wallikeri, dhfr mutations were less frequent, with Phe57Leu-Ser58Arg reaching 17% (34 of 204 samples). These two mutants were the most prevalent in central and east Africa and were fixed in Kenyan isolates. We detected six and four other non-synonymous mutations, representing 8% (24 isolates) and 2% (five isolates) of the P ovale curtisi and P ovale wallikeri isolates, respectively. Whole-genome sequencing and microsatellite analyses revealed reduced genetic diversity around the mutant pocdhfr and powdhfr genes. The mutant DHFR proteins showed structural changes at the pyrimethamine binding site in-silico, confirmed by a 4-times increase in pyrimethamine half-maximal inhibitory concentration in an Escherichia coli growth assay for the Phe57Leu-Ser58Arg mutant and 50-times increase for the Ala15Ser-Ser58Arg mutant, compared with the wild-type counterparts. INTERPRETATION The widespread use of sulfadoxine-pyrimethamine for malaria chemoprevention might have exerted fortuitous selection pressure for dhfr mutations in P ovale spp. This calls for closer monitoring of dhfr and dhps mutations in P ovale spp. FUNDING French Ministry of Health, Agence Nationale de la Recherche, and Global Emerging Infections Surveillance branch of the Armed Forces Health Surveillance Division.
Collapse
Affiliation(s)
- Valentin Joste
- Université Paris Cité, IRD, MERIT, Paris, France; Centre National de Référence du Paludisme, AP-HP, Hôpital Bichat - Claude-Bernard, Paris, France.
| | - Romain Coppée
- Université de Rouen Normandie, Laboratoire de Parasitologie-Mycologie, UR 7510 ESCAPE, Centre Hospitalier Universitaire de Rouen, Rouen, France
| | | | - Yann Rakotoarivony
- Centre National de Référence du Paludisme, AP-HP, Hôpital Bichat - Claude-Bernard, Paris, France
| | | | | | - Bruno Pradines
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées (IRBA), Marseille, France; Université Aix-Marseille, IRD, SSA, VITROME, Marseille, France; IHU Méditerranée Infection, Marseille, France; Centre National de Référence du Paludisme, Marseille, France
| | | | - Frédéric Ariey
- INSERM U1016, Institut Cochin, Laboratoire de Parasitologie-Mycologie, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
| | - Nimol Khim
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Jean Popovici
- Malaria Research Unit, Institut Pasteur du Cambodge, Phnom Penh, Cambodia
| | - Toshihiro Mita
- Department of Tropical Medicine and Parasitology, Faculty of Medicine, Juntendo University, Tokyo, Japan
| | - Mirjam Groger
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Ramharter
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timothy Egbo
- Walter Reed Army Institute of Research - Africa (WRAIR-A), Kisumu, Kenya
| | - Dennis W Juma
- Department of Emerging and Infectious Diseases (DEID), Walter Reed Army Institute of Research - Africa (WRAIR-A), Kisumu, Kenya
| | - Hoseah Akala
- Department of Emerging and Infectious Diseases (DEID), Walter Reed Army Institute of Research - Africa (WRAIR-A), Kisumu, Kenya
| | - Sandrine Houzé
- Université Paris Cité, IRD, MERIT, Paris, France; Centre National de Référence du Paludisme, AP-HP, Hôpital Bichat - Claude-Bernard, Paris, France
| | - Jérôme Clain
- Université Paris Cité, IRD, MERIT, Paris, France; Centre National de Référence du Paludisme, AP-HP, Hôpital Bichat - Claude-Bernard, Paris, France
| |
Collapse
|
22
|
Tong Y, Ratnasiri K, Hanif S, Nguyen AT, Roh ME, Dorsey G, Kakuru A, Jagannathan P, Benjamin-Chung J. Pathways through which intermittent preventive treatment for malaria in pregnancy influences child growth faltering: a mediation analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.09.24308656. [PMID: 38947035 PMCID: PMC11213035 DOI: 10.1101/2024.06.09.24308656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Intermittent preventive treatment for malaria in pregnancy (IPTp) can improve birth outcomes, but whether it confers benefits to postnatal growth is unclear. We investigated the effect of IPTp on infant growth in Uganda and its pathways of effects using causal mediation analyses. Methods We analyzed data from 633 infants born to mothers enrolled in a randomized trial of monthly IPTp with dihydroartemisinin-piperaquine (DP) vs sulfadoxine-pyrimethamine (SP) (NCT02793622). Weight and length were measured from 0-12 months of age. Using generalized linear models, we estimated effects of DP vs. SP on gravidity-stratified mean length-for-age (LAZ) and weight-for-length Z-scores (WLZ). We investigated mediation by placental malaria, gestational weight change, maternal anemia, maternal inflammation-related proteins, preterm birth, birth length, and birth weight. Mediation models adjusted for infant sex, gravidity, gestational age at enrollment, maternal age, maternal parasitemia at enrollment, education, and wealth. Findings SP increased LAZ by 0.18-0.28 Z from birth through age 4 months compared to DP, while DP increased WLZ by 0.11-0.28 Z from 2-8 months compared to SP among infants of multigravidae. We did not observe these differences among primigravida. Mediators of SP included increased birth weight and length and maternal stem cell factor at delivery. Mediators of DP included placental malaria and birth length, maternal IL-18, CDCP1, and CD6 at delivery. Interpretation In high malaria transmission settings, different IPTp regimens influenced infant growth among multigravidae through distinct pathways in the period of exclusive breastfeeding, when few other interventions are available. Funding Stanford Center for Innovation and Global Health, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bill & Melinda Gates Foundation.
Collapse
Affiliation(s)
- Yanwei Tong
- Department of Statistics, Stanford University, Stanford, United States
| | - Kalani Ratnasiri
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
- Department of Microbiology and Immunology, Stanford University, Stanford, United States
| | - Suhi Hanif
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
| | - Anna T. Nguyen
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
| | - Michelle E. Roh
- Institute for Global Health Sciences, University of California San Francisco, San Francisco, United States
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, United States
| | - Grant Dorsey
- Department of Medicine, Division of HIV, ID, and Global Medicine, University of California San Francisco, San Francisco
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Jade Benjamin-Chung
- Department of Epidemiology and Population Health, Stanford University, Stanford, United States
- Chan Zuckerberg Biohub, San Francisco, United States
| |
Collapse
|
23
|
Matambisso G, Brokhattingen N, Maculuve S, Cístero P, Mbeve H, Escoda A, Bambo G, Cuna B, Melembe C, Ndimande N, Tetteh KKA, Drakeley C, Gamain B, Chitnis C, Chauhan V, Quintó L, Macete E, Mayor A. Sustained clinical benefit of malaria chemoprevention with sulfadoxine-pyrimethamine (SP) in pregnant women in a region with high SP resistance markers. J Infect 2024; 88:106144. [PMID: 38574776 DOI: 10.1016/j.jinf.2024.106144] [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: 10/04/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
OBJECTIVE The effectiveness of intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP) is threatened by increasing SP-resistance in Africa. We assessed the level of SP-resistance markers, and the clinical and parasitological effectiveness of IPTp-SP in southern Mozambique. METHODS P. falciparum infection, antimalarial antibodies and dhfr/dhps SP-resistance mutants were detected by quantitative polymerase chain reaction (qPCR), suspension array technology and targeted deep sequencing, respectively, among 4016 HIV-negative women in Maputo province (2016-2019). Univariate and multivariate regression models were used to assess the association between taking the recommended three or more IPTp-SP doses (IPTp3+) and parasitological and clinical outcomes. RESULTS 84.3% (3385/4016) women received three or more IPTp-SP doses. The prevalence of quintuple mutants at first antenatal care (ANC) visit was 94.2%. IPTp3+ was associated with a higher clearance rate of qPCR-detected infections from first ANC visit to delivery (adjusted odds ratio [aOR]=5.9, 95% CI: 1.5-33.3; p = 0.012), lower seroprevalence at delivery of antibodies against the pregnancy-specific antigen VAR2CSADBL34 (aOR=0.72, 95% CI: 0.54-0.95; p = 0.022), and lower prevalence of low birth weight deliveries (aOR: 0.61, 95% CI: 0.41-0.90; p = 0.013). CONCLUSION A sustained parasitological effect of IPTp-SP contributes to the clinical effectiveness of IPTp3+ in areas with high prevalence of SP-resistance markers.
Collapse
Affiliation(s)
- Glória Matambisso
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Nanna Brokhattingen
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Sónia Maculuve
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Pau Cístero
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Henriques Mbeve
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Anna Escoda
- ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Gizela Bambo
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Boaventura Cuna
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Cardoso Melembe
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Nelo Ndimande
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique
| | - Kevin K A Tetteh
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Chris Drakeley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Benoit Gamain
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, Paris, France
| | - Chetan Chitnis
- Malaria Parasite Biology and Vaccines, Department of Parasites & Insect Vectors, Institut Pasteur, Paris, France
| | - Virander Chauhan
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India
| | - Llorenç Quintó
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique; ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Eusébio Macete
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique; National Directare of Health, Ministry of Health, Maputo, Mozambique
| | - Alfredo Mayor
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Moçambique; ISGlobal, Barcelona Institute for Global Health, Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain; Department of Physiologic Sciences, Faculty of Medicine, Universidade Eduardo Mondlane, Maputo, Mozambique.
| |
Collapse
|
24
|
Rakotozandrindrainy R, Rakotoarivelo RA, Kislaya I, Marchese V, Rasamoelina T, Solonirina J, Ratiaharison EF, Razafindrakoto R, Razafindralava NM, Rakotozandrindrainy N, Radomanana M, Andrianarivelo MR, Klein P, Lorenz E, Jaeger A, Hoekstra PT, Corstjens PLAM, Schwarz NG, van Dam GJ, May J, Fusco D. Schistosome infection among pregnant women in the rural highlands of Madagascar: A cross-sectional study calling for public health interventions in vulnerable populations. PLoS Negl Trop Dis 2024; 18:e0011766. [PMID: 38626192 PMCID: PMC11051649 DOI: 10.1371/journal.pntd.0011766] [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: 11/03/2023] [Revised: 04/26/2024] [Accepted: 03/26/2024] [Indexed: 04/18/2024] Open
Abstract
INTRODUCTION Schistosomiasis is a parasitic infection highly prevalent in sub-Saharan Africa (SSA) with Madagascar being among the countries with highest burden of the disease worldwide. Despite WHO recommendations, suggesting treatment of pregnant women after the first trimester, this group is still excluded from Mass Drug Administration programs. Our study, had the objective to measure the prevalence of schistosome infection among pregnant women in Madagascar in order to inform public health policies for treatment in this vulnerable population. METHODS Women were recruited for this cross-sectional study between April 2019 and February 2020 when attending Antenatal Care Services (ANCs) at one of 42 included Primary Health Care Centers. The urine-based upconverting reporter particle, lateral flow (UCP-LF) test detecting circulating anodic antigen was used for the detection of schistosome infections. To identify factors associated with the prevalence of schistosome infection crude and adjusted prevalence ratios and 95% CIs were estimated using mixed-effect Poisson regression. RESULTS Among 4,448 participating women aged between 16 and 47 years, the majority (70.4%, 38 n = 3,133) resided in rural settings. Overall, the prevalence of schistosome infection was 55.9% (n = 2486, CI 95%: 53.3-58.5). A statistically significant association was found with age group (increased prevalence in 31-47 years old, compared to 16-20 years old (aPR = 1.15, CI 95%: 1.02-1.29) and with uptake of antimalaria preventive treatment (decreased prevalence, aPR = 0.85, CI 95%: 0.77-0.95). No other associations of any personal characteristics or contextual factors with schistosome infection were found in our multivariate regression analysis. DISCUSSION AND CONCLUSION The high prevalence of schistosome infection in pregnant women supports the consideration of preventive schistosomiasis treatment in ANCs of the Malagasy highlands. We strongly advocate for adapting schistosomiasis programs in highly endemic contexts. This, would contribute to both the WHO and SDGs agendas overall to improving the well-being of women and consequently breaking the vicious cycle of poverty perpetuated by schistosomiasis.
Collapse
Affiliation(s)
| | | | - Irina Kislaya
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Valentina Marchese
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | | | | | | | | | | | | - Mickael Radomanana
- Infectious diseases service, University Hospital Tambohobe, Fianarantsoa, Madagascar
| | | | - Philipp Klein
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Eva Lorenz
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Anna Jaeger
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | | | | - Norbert Georg Schwarz
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
| | | | - Jürgen May
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | - Daniela Fusco
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine (BNITM), Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg-Borstel-Lübeck-Riems, Germany
| | | |
Collapse
|
25
|
Lee JJ, Kakuru A, Jacobson KB, Kamya MR, Kajubi R, Ranjit A, Gaw SL, Parsonnet J, Benjamin-Chung J, Dorsey G, Jagannathan P, Roh ME. Monthly Sulfadoxine-Pyrimethamine During Pregnancy Prevents Febrile Respiratory Illnesses: A Secondary Analysis of a Malaria Chemoprevention Trial in Uganda. Open Forum Infect Dis 2024; 11:ofae143. [PMID: 38585183 PMCID: PMC10995957 DOI: 10.1093/ofid/ofae143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024] Open
Abstract
Background Trials evaluating antimalarials for intermittent preventive treatment in pregnancy (IPTp) have shown that dihydroartemisinin-piperaquine (DP) is a more efficacious antimalarial than sulfadoxine-pyrimethamine (SP); however, SP is associated with higher birthweight, suggesting that SP demonstrates "nonmalarial" effects. Chemoprevention of nonmalarial febrile illnesses (NMFIs) was explored as a possible mechanism. Methods In this secondary analysis, we leveraged data from 654 pregnant Ugandan women without HIV infection who participated in a randomized controlled trial comparing monthly IPTp-SP with IPTp-DP. Women were enrolled between 12 and 20 gestational weeks and followed through delivery. NMFIs were measured by active and passive surveillance and defined by the absence of malaria parasitemia. We quantified associations among IPTp regimens, incident NMFIs, antibiotic prescriptions, and birthweight. Results Mean "birthweight for gestational age" Z scores were 0.189 points (95% CI, .045-.333) higher in women randomized to IPTp-SP vs IPTp-DP. Women randomized to IPTp-SP had fewer incident NMFIs (incidence rate ratio, 0.74; 95% CI, .58-.95), mainly respiratory NMFIs (incidence rate ratio, 0.69; 95% CI, .48-1.00), vs IPTp-DP. Counterintuitively, respiratory NMFI incidence was positively correlated with birthweight in multigravidae. In total 75% of respiratory NMFIs were treated with antibiotics. Although overall antibiotic prescriptions were similar between arms, for each antibiotic prescribed, "birthweight for gestational age" Z scores increased by 0.038 points (95% CI, .001-.074). Conclusions Monthly IPTp-SP was associated with reduced respiratory NMFI incidence, revealing a potential nonmalarial mechanism of SP and supporting current World Health Organization recommendations for IPTp-SP, even in areas with high-grade SP resistance. While maternal respiratory NMFIs are known risk factors of lower birthweight, most women in our study were presumptively treated with antibiotics, masking the potential benefit of SP on birthweight mediated through preventing respiratory NMFIs.
Collapse
Affiliation(s)
- Jordan John Lee
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Karen B Jacobson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Kaiser Permanente Northern California Division of Research, Vaccine Study Center, Oakland, California, USA
| | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Anju Ranjit
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Stephanie L Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, California, USA
| | - Julie Parsonnet
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
| | - Jade Benjamin-Chung
- Department of Epidemiology and Population Health, Stanford University, Stanford, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Grant Dorsey
- Division of HIV, Infectious Diseases, and Global Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Prasanna Jagannathan
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, California, USA
| | - Michelle E Roh
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
- Malaria Elimination Initiative, Institute for Global Health Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
26
|
Pasricha SR, Moya E, Ataide R, Mzembe G, Harding R, Phiri K. The REVAMP trial: key questions remain - Authors' reply. Lancet 2024; 403:29-30. [PMID: 38184336 DOI: 10.1016/s0140-6736(23)01915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/08/2023] [Indexed: 01/08/2024]
Affiliation(s)
- Sant-Rayn Pasricha
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Diagnostic Haematology, The Royal Melbourne Hospital, Parkville, VIC, Australia; Clinical Haematology, The Peter MacCallum Cancer Centre and The Royal Melbourne Hospital, Parkville, VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Ernest Moya
- Training and Research Unit of Excellence, Blantyre, Malawi; Department of Public Health, School of Public and Global Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Ricardo Ataide
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medicine at the Peter Doherty Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Glory Mzembe
- Training and Research Unit of Excellence, Blantyre, Malawi; Department of Public Health, School of Public and Global Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Rebecca Harding
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Kamija Phiri
- Training and Research Unit of Excellence, Blantyre, Malawi; Department of Public Health, School of Public and Global Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| |
Collapse
|
27
|
Guémas E, Coppée R, Ménard S, du Manoir M, Nsango S, Makaba Mvumbi D, Nakoune E, Eboumbou Moukoko CE, Bouyou Akotet MK, Mirabeau TY, Manguin S, Malekita Yobi D, Akiana J, Kouna LC, Mawili Mboumba DP, Voumbo-Matoumona DF, Otam AL, Rubbo PA, Lombart JP, Kwanai E, Cohen O, Iriart X, Ayong L, Lekana-Douki JB, Ariey F, Berry A. Evolution and spread of Plasmodium falciparum mutations associated with resistance to sulfadoxine-pyrimethamine in central Africa: a cross-sectional study. THE LANCET. MICROBE 2023; 4:e983-e993. [PMID: 37865113 DOI: 10.1016/s2666-5247(23)00211-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Efficacy of sulfadoxine-pyrimethamine, the malaria chemoprophylaxis used in pregnant women, and in children when combined with amodiaquine, is threatened by the accumulation of mutations in the Plasmodium falciparum dihydropteroate synthase (pfdhps) and dihydrofolate reductase (pfdhfr) genes. Data on the prevalence of resistant alleles in central Africa and the new pfdhps I431V mutation, particularly associated with other mutations to form the pfdhps vagKgs allele, are scarce. We explored the frequency and geographical distribution of pfdhps and pfdhfr mutations in central Africa in 2014-18, and assessed the evolutionary origin of the vagKgs allele. METHODS Samples were collected at 18 health-care centres in seven countries (Angola, Cameroon, Central African Republic, Democratic Republic of the Congo, Gabon, Nigeria, and Republic of the Congo) from patients who showed possible symptoms of malaria between March 1, 2014, and Oct 31, 2018. Samples that were positive for P falciparum were transported to a laboratory in Toulouse, France, and genotyped. The frequency of pfdhfr and pfdhps mutations was studied in 1749 samples. Microsatellites in pfdhps flanking regions and whole-genome analysis compared with parasite genomes from the data-sharing network MalariaGEN were performed on samples carrying the vagKgs allele. FINDINGS Mapping of the prevalence of single nucleotide polymorphisms and corresponding alleles of pfdhfr and pfdhps showed a substantial spread of alleles associated with sulfadoxine-pyrimethamine resistance in central Africa during the 2014-18 period, especially an increase going west to east in pfdhps alleles carrying the K540E and A581G mutations. A high prevalence of the pfdhps I431V mutation was observed in Cameroon (exceeding 50% in the northern region) and Nigeria. Genomic analysis showed a recent African emergence and a clonal expansion of the most frequent pfdhps vagKgs allele. INTERPRETATION Reduced sulfadoxine-pyrimethamine efficacy due to increased resistance is a worrying situation, especially because the malaria transmission level is high in central Africa. Although the resistance phenotype remains to be confirmed, the emergence and spread of the vagKgs allele in west and central Africa could challenge the use of sulfadoxine-pyrimethamine. FUNDING Toulouse Institute for Infectious and Inflammatory Diseases.
Collapse
Affiliation(s)
- Emilie Guémas
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France; Département de Parasitologie et Mycologie, CHU Toulouse, Toulouse, France; LAAS-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Romain Coppée
- Université Paris Cité and Sorbonne Paris Nord, INSERM, IAME, Paris, France
| | - Sandie Ménard
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France
| | - Milena du Manoir
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France
| | - Sandrine Nsango
- Faculté de Médecine et des Sciences Pharmaceutiques, Université de Douala, Douala, Cameroon; Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Dieudonné Makaba Mvumbi
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo; Institute for Medical Immunology, Université Libre de Bruxelles, Brussells, Belgium
| | | | - Carole Else Eboumbou Moukoko
- Faculté de Médecine et des Sciences Pharmaceutiques, Université de Douala, Douala, Cameroon; Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Marielle Karine Bouyou Akotet
- Département de Parasitologie Mycologie Médecine Tropicale, Faculté de Médecine de l'Université des Sciences de la Santé, Libreville, Gabon; Centre de Recherche Biomédicale en Pathogènes Infectieux et Pathologies Associées, CREIPA, Université des Sciences de la Santé, Libreville, Gabon
| | - Tatfeng Youtchou Mirabeau
- Department of Medical Laboratory Science, Faculty of Basic Medical Sciences, College of Health Sciences, Niger Delta University, Wilberforce Island, Nigeria
| | - Sylvie Manguin
- Hydro Sciences Montpellier, Université de Montpellier, CNRS, IRD, Montpellier, France
| | - Doudou Malekita Yobi
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Jean Akiana
- Laboratoire National de Santé Publique, Université Marien Ngouabi, Brazzaville, Republic of the Congo
| | - Lady Charlène Kouna
- Unité d'Evolution Epidémiologie et Résistances Parasitaires, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon; Département de Parasitologie-Mycologie, Université des Sciences de la Santé, Libreville, Gabon
| | - Denise Patricia Mawili Mboumba
- Département de Parasitologie Mycologie Médecine Tropicale, Faculté de Médecine de l'Université des Sciences de la Santé, Libreville, Gabon; Centre de Recherche Biomédicale en Pathogènes Infectieux et Pathologies Associées, CREIPA, Université des Sciences de la Santé, Libreville, Gabon
| | - Dominique Fatima Voumbo-Matoumona
- Laboratoire National de Santé Publique, Université Marien Ngouabi, Brazzaville, Republic of the Congo; Unité d'Evolution Epidémiologie et Résistances Parasitaires, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon; Département de Parasitologie-Mycologie, Université des Sciences de la Santé, Libreville, Gabon
| | - Alliance-Laure Otam
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France
| | | | | | - Elisabeth Kwanai
- Coordination diocésaine de la Santé, Diocèse de Maroua-Mokolo, Maroua, Cameroon
| | - Olivia Cohen
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France
| | - Xavier Iriart
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France; Département de Parasitologie et Mycologie, CHU Toulouse, Toulouse, France
| | - Lawrence Ayong
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Jean Bernard Lekana-Douki
- Unité d'Evolution Epidémiologie et Résistances Parasitaires, Centre Interdisciplinaire de Recherches Médicales de Franceville, Franceville, Gabon; Département de Parasitologie-Mycologie, Université des Sciences de la Santé, Libreville, Gabon
| | - Frédéric Ariey
- INSERM U1016, Institut Cochin, Laboratoire de Parasitologie-Mycologie, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
| | - Antoine Berry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity), Université de Toulouse, CNRS UMR5051, INSERM UMR 1291, UPS, Toulouse, France; Département de Parasitologie et Mycologie, CHU Toulouse, Toulouse, France.
| |
Collapse
|
28
|
Kayiba NK, Tshibangu-Kabamba E, Rosas-Aguirre A, Kaku N, Nakagama Y, Kaneko A, Makaba DM, Malekita DY, Devleesschauwer B, Likwela JL, Zakayi PK, DeMol P, Lelo GM, Hayette MP, Dikassa PL, Kido Y, Speybroeck N. The landscape of drug resistance in Plasmodium falciparum malaria in the Democratic Republic of Congo: a mapping systematic review. Trop Med Health 2023; 51:64. [PMID: 37968745 PMCID: PMC10647042 DOI: 10.1186/s41182-023-00551-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023] Open
Abstract
CONTEXT The Democratic Republic of Congo (DRC), one of the most malaria-affected countries worldwide, is a potential hub for global drug-resistant malaria. This study aimed at summarizing and mapping surveys of malaria parasites carrying molecular markers of drug-resistance across the country. METHODS A systematic mapping review was carried out before July 2023 by searching for relevant articles through seven databases (PubMed, Embase, Scopus, African Journal Online, African Index Medicus, Bioline and Web of Science). RESULTS We identified 1541 primary studies of which 29 fulfilled inclusion criteria and provided information related to 6385 Plasmodium falciparum clinical isolates (collected from 2000 to 2020). We noted the PfCRT K76T mutation encoding for chloroquine-resistance in median 32.1% [interquartile interval, IQR: 45.2] of analyzed malaria parasites. The proportion of parasites carrying this mutation decreased overtime, but wide geographic variations persisted. A single isolate had encoded the PfK13 R561H substitution that is invoked in artemisinin-resistance emergence in the Great Lakes region of Africa. Parasites carrying various mutations linked to resistance to the sulfadoxine-pyrimethamine combination were widespread and reflected a moderate resistance profile (PfDHPS A437G: 99.5% [IQR: 3.9]; PfDHPS K540E: 38.9% [IQR: 47.7]) with median 13.1% [IQR: 10.3] of them being quintuple IRN-GE mutants (i.e., parasites carrying the PfDHFR N51I-C59R-S108N and PfDHPS A437G-K540E mutations). These quintuple mutants tended to prevail in eastern regions of the country. Among circulating parasites, we did not record any parasites harboring mutations related to mefloquine-resistance, but we could suspect those with decreased susceptibility to quinine, amodiaquine, and lumefantrine based on corresponding molecular surrogates. CONCLUSIONS Drug resistance poses a serious threat to existing malaria therapies and chemoprevention options in the DRC. This review provides a baseline for monitoring public health efforts as well as evidence for decision-making in support of national malaria policies and for implementing regionally tailored control measures across the country.
Collapse
Affiliation(s)
- Nadine Kalenda Kayiba
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
- Department of Public Health, Faculty of Medicine, University of Mbujimayi, Mbujimayi, Democratic Republic of Congo
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Evariste Tshibangu-Kabamba
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
- Department of Internal Medicine, Faculty of Medicine, University of Mbujimayi, Mbujimayi, Democratic Republic of Congo
| | - Angel Rosas-Aguirre
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
| | - Natsuko Kaku
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yu Nakagama
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Kaneko
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Dieudonné Mvumbi Makaba
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
- Department of Quality of Laboratories, Sciensano, Brussels, Belgium
| | - Doudou Yobi Malekita
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Brecht Devleesschauwer
- Department of Epidemiology and Public Health, Sciensano, Brussels, Belgium
- Department of Translational Physiology, Infectiology and Public Health, Ghent University, Merelbeke, Belgium
| | - Joris Losimba Likwela
- Department of Public Health, Faculty of Medicine, University of Kisangani, Kisangani, Democratic Republic of Congo
| | - Pius Kabututu Zakayi
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Patrick DeMol
- Laboratory of Clinical Microbiology, Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | - Georges Mvumbi Lelo
- Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Marie-Pierre Hayette
- Laboratory of Clinical Microbiology, Center for Interdisciplinary Research on Medicines, University of Liège, Liège, Belgium
| | - Paul Lusamba Dikassa
- School of Public Health, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Yasutoshi Kido
- Research Center for Infectious Disease Science & Department of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan.
| | - Niko Speybroeck
- Research Institute of Health and Society, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
29
|
Molina-de la Fuente I, Sagrado Benito MJ, Lasry E, Ousley J, García L, González V, Pasquale HA, Julla A, Uwiragiye P, Abdi AM, Chol BT, Abubakr B, Benito A, Casademont C, Berzosa P, Nanclares C. Seasonal malaria chemoprevention in a context of high presumed sulfadoxine-pyrimethamine resistance: malaria morbidity and molecular drug resistance profiles in South Sudan. Malar J 2023; 22:345. [PMID: 37950227 PMCID: PMC10637007 DOI: 10.1186/s12936-023-04740-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: 01/30/2023] [Accepted: 10/03/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Seasonal malaria chemoprevention (SMC) using sulfadoxine-pyrimethamine plus amodiaquine (SP-AQ), is a community-based malaria preventive strategy commonly used in the Sahel region of sub-Saharan Africa. However, to date it has not been implemented in East Africa due to high SP resistance levels. This paper is a report on the implementation of SMC outside of the Sahel in an environment with a high level of presumed SP-resistance: five cycles of SMC using SPAQ were administered to children 3-59 months during a period of high malaria transmission (July-December 2019) in 21 villages in South Sudan. METHODS A population-based SMC coverage survey was combined with a longitudinal time series analysis of health facility and community health data measured after each SMC cycle. SMC campaign effectiveness was assessed by Poisson model. SPAQ molecular resistance markers were additionally analysed from dried blood spots from malaria confirmed patients. RESULTS Incidence of uncomplicated malaria was reduced from 6.6 per 100 to an average of 3.2 per 100 after SMC administration (mean reduction: 53%) and incidence of severe malaria showed a reduction from 21 per 10,000 before SMC campaign to a mean of 3.3 per 10,000 after each cycle (mean reduction: 84%) in the target group when compared to before the SMC campaign. The most prevalent molecular haplotype associated with SP resistance was the IRNGE haplotype (quintuple mutant, with 51I/59R/108N mutation in pfdhfr + 437G/540E in pfdhps). In contrast, there was a low frequency of AQ resistance markers and haplotypes resistant to both drugs combined (< 2%). CONCLUSIONS The SMC campaign was effective and could be used as an additional preventive tool in seasonal malaria settings outside of the Sahel, especially in areas where access to health care is unstable. Malaria case load reduction was observed despite the high level of resistance to SP.
Collapse
Affiliation(s)
| | | | - Estrella Lasry
- Médecins Sans Frontières, Carrer de Zamora, 54, 08005, Barcelona, Spain
| | | | - Luz García
- Institute of Health Carlos III, Madrid, Spain
| | | | | | - Ahmed Julla
- National Malaria Control Programme, Ministry of Health, Juba, South Sudan
| | | | | | | | | | - Agustín Benito
- Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomedica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | | - Pedro Berzosa
- Institute of Health Carlos III, Madrid, Spain
- Centro de Investigación Biomedica en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | |
Collapse
|
30
|
Figueroa-Romero A, Bissombolo D, Meremikwu M, Ratsimbasoa A, Sacoor C, Arikpo I, Lemba E, Nhama A, Rakotosaona R, Llach M, Pons-Duran C, Sanz S, Ma L, Doderer-Lang C, Maly C, Roman E, Pagnoni F, Mayor A, Menard D, González R, Menéndez C. Prevalence of molecular markers of resistance to sulfadoxine-pyrimethamine before and after community delivery of intermittent preventive treatment of malaria in pregnancy in sub-Saharan Africa: a multi-country evaluation. Lancet Glob Health 2023; 11:e1765-e1774. [PMID: 37858587 DOI: 10.1016/s2214-109x(23)00414-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND The effectiveness of community delivery of intermittent preventive treatment (C-IPT) of malaria in pregnancy (IPTp) with sulfadoxine-pyrimethamine has been evaluated in selected areas of the Democratic Republic of the Congo, Madagascar, Mozambique, and Nigeria. We aimed to assess the effect of C-IPTp on the potential development of Plasmodium falciparum resistance to sulfadoxine-pyrimethamine, since it could threaten the effectiveness of this strategy. METHODS Health facility-based cross-sectional surveys were conducted at baseline and 3 years after C-IPTp implementation in two neighbouring areas per country, one with C-IPTp intervention, and one without, in the four project countries. Dried blood spots from children under five years of age with clinical malaria were collected. Sulfadoxine-pyrimethamine resistance-associated mutations of the P falciparum dhfr (Asn51Ile/Cys59Arg/Ser108Asn/Ile164Leu) and dhps (Ile431Val/Ser436Ala/Ala437Gly/Lys540Glu/Ala581Gly/Ala613Ser) genes were analysed. FINDINGS 2536 children were recruited between June 19 and Oct 10, 2018, during baseline surveys. Endline surveys were conducted among 2447 children between July 26 and Nov 30, 2021. In the Democratic Republic of the Congo, the dhfr/dhps IRNI/ISGEAA inferred haplotype remained lower than 10%, from 2% (5 of 296) at baseline to 8% (24 of 292) at endline, and from 3% (9 of 300) at baseline to 6% (18 of 309) at endline surveys in intervention and non-intervention areas respectively with no significant difference in the change between the areas. In Mozambique, the prevalence of this haplotype remained stable at over 60% (194 [64%] of 302 at baseline to 194 [64%] of 303 at endline, and 187 [61%] of 306 at baseline to 183 [61%] of 301 in endline surveys, in non-intervention and intervention areas respectively). No isolates harbouring the dhps ISGEAA genotype were found in Nigeria. In Madagascar, only five isolates with this haplotype were found in the non-intervention area (2 [>1%] of 300 at baseline and 3 [1%] of 300 at endline surveys). No isolates were found carrying the dhps ISGEGA genotype. INTERPRETATION C-IPTp did not increase the prevalence of molecular markers associated with sulfadoxine-pyrimethamine resistance after three years of programme implementation. These findings reinforce C-IPTp as a strategy to optimise the control of malaria during pregnancy, and support the WHO guidelines for prevention of malaria in pregnancy. FUNDING UNITAID [2017-13-TIPTOP].
Collapse
Affiliation(s)
- Antía Figueroa-Romero
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Kinshasa, Democratic Republic of the Congo
| | | | - Martin Meremikwu
- Cross River Health and Demographic Surveillance System, University of Calabar, Cross River State, Nigeria
| | | | - Charfudin Sacoor
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Iwara Arikpo
- Cross River Health and Demographic Surveillance System, University of Calabar, Cross River State, Nigeria
| | - Elsha Lemba
- Medecins d'Afrique, Kinshasa, Democratic Republic of the Congo
| | - Abel Nhama
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; Instituto Nacional de Saúde (INS), Maputo, Mozambique
| | | | - Mireia Llach
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Clara Pons-Duran
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Sergi Sanz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Kinshasa, Democratic Republic of the Congo; Department of Basic Clinical Practice, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Laurence Ma
- Institut Pasteur, Université Paris Cité, Biomics Platform, Paris, France
| | - Cécile Doderer-Lang
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, Strasbourg, France
| | - Christina Maly
- Jhpiego, John Hopkins University Affiliate, Baltimore MD, USA
| | - Elaine Roman
- Jhpiego, John Hopkins University Affiliate, Baltimore MD, USA
| | - Franco Pagnoni
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain
| | - Alfredo Mayor
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Didier Menard
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, Strasbourg, France; Malaria Genetics and Resistance Unit, Institut Pasteur, Paris, France; Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France; CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, Strasbourg, France
| | - Raquel González
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Kinshasa, Democratic Republic of the Congo; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.
| | - Clara Menéndez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Kinshasa, Democratic Republic of the Congo; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| |
Collapse
|
31
|
Frempong NA, Ahiabor C, Anyan WK, Mama A, Kusi KA, Ofori MF, Adu B, Debrah AY, Anang AK, Ndam NT, Courtin D. Malaria, Urogenital Schistosomiasis, and Anaemia in Pregnant Ghanaian Women. J Parasitol Res 2023; 2023:7500676. [PMID: 37808169 PMCID: PMC10558271 DOI: 10.1155/2023/7500676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/03/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
Background Anaemia is common in sub-Saharan Africa, and parasitic infections could worsen its burden during pregnancy. Moreover, women become susceptible to malaria during pregnancy. We investigated Plasmodium falciparum (P. falciparum) and Schistosoma haematobium (S. haematobium) infections and determined their association with anaemia during pregnancy. Methods A cross-sectional study involving 707 pregnant women attending antenatal care visits (ANC) and 446 at delivery was conducted in Battor and Adidome hospitals. Pregnant women were screened by microscopy and qPCR for P. falciparum and S. haematobium infections. Haemoglobin (Hb) levels were determined, and most participants received intermittent preventive treatment during pregnancy (IPTp) during ANC till delivery. Regression analyses were performed for associations between parasite infection and anaemia. Results P. falciparum microscopy prevalence at ANC and delivery was 8% and 2%, respectively, and by PCR 24% at ANC and 12% at delivery. Anaemia prevalence at ANC was 52% and 49% at delivery. There was an increased risk of anaemia with P. falciparum infection (aOR = 1.92; p = 0.04). IPTp (p = 0.003) and age (p = 0.004) were associated with increased Hb levels at delivery. S. haematobium prevalence by microscopy was 4% at ANC and 2% at delivery. No significant correlation between S. haematobium and Hb levels was observed (coef. = -0.62 g/dl; p = 0.07). Conclusion High anaemia prevalence was observed during pregnancy, and P. falciparum infection was associated with anaemia at ANC. Low S. haematobium prevalence could be attributed to previous praziquantel treatment during mass drug administration. Routine diagnosis and treatment of S. haematobium infections in endemic areas could be initiated to reduce schistosomiasis during pregnancy.
Collapse
Affiliation(s)
- Naa Adjeley Frempong
- Clinical Microbiology Department, Kwame Nkrumah University of Science and Technology, Ghana
- Parasitology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Charity Ahiabor
- Science Laboratory Technology, Accra Technical University, Ghana
| | - William K. Anyan
- Parasitology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Atikatou Mama
- Inserm U 1016, Institut Cochin, Université de Paris, 75014, France
| | - Kwadwo Asamoah Kusi
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Michael F. Ofori
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Bright Adu
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Alex Yaw Debrah
- Faculty of Health Sciences, Kwame Nkrumah University of Science and Technology, Ghana
| | - Abraham K. Anang
- Institute of Environment and Sanitation Studies(IESS), University of Ghana, Legon, Ghana
| | | | | |
Collapse
|
32
|
Rent S, Bauserman M, Laktabai J, Tshefu AK, Taylor SM. Malaria in Pregnancy: Key Points for the Neonatologist. Neoreviews 2023; 24:e539-e552. [PMID: 37653081 DOI: 10.1542/neo.24-9-e539] [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: 09/02/2023]
Abstract
In malaria-endemic regions, infection with the malaria parasite Plasmodium during pregnancy has been identified as a key modifiable factor in preterm birth, the delivery of low-birthweight infants, and stillbirth. Compared with their nonpregnant peers, pregnant persons are at higher risk for malaria infection. Malaria infection can occur at any time during pregnancy, with negative effects for the pregnant person and the fetus, depending on the trimester in which the infection is contracted. Pregnant patients who are younger, in their first or second pregnancy, and those coinfected with human immunodeficiency virus are at increased risk for malaria. Common infection prevention measures during pregnancy include the use of insecticide-treated bed nets and the use of intermittent preventive treatment with monthly doses of antimalarials, beginning in the second trimester in pregnant patients in endemic areas. In all trimesters, artemisinin-combination therapies are the first-line treatment for uncomplicated falciparum malaria, similar to treatment in nonpregnant adults. The World Health Organization recently revised its recommendations, now listing the specific medication artemether-lumefantrine as first-line treatment for uncomplicated malaria in the first trimester. While strong prevention and detection methods exist, use of these techniques remains below global targets. Ongoing work on approaches to treatment and prevention of malaria during pregnancy remains at the forefront of global maternal child health research.
Collapse
Affiliation(s)
- Sharla Rent
- Department of Pediatrics, Duke University School of Medicine, Durham, NC
| | | | | | - Antoinette K Tshefu
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | - Steve M Taylor
- Department of Medicine, Duke University School of Medicine, Durham, NC
| |
Collapse
|
33
|
Nana RRD, Hawadak J, Foko LPK, Kumar A, Chaudhry S, Arya A, Singh V. Intermittent preventive treatment with Sulfadoxine pyrimethamine for malaria: a global overview and challenges affecting optimal drug uptake in pregnant women. Pathog Glob Health 2023; 117:462-475. [PMID: 36177658 PMCID: PMC10337642 DOI: 10.1080/20477724.2022.2128563] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Malaria in Pregnancy (MiP) leading to morbidity and mortality is a major public health problem that poses significant risk to pregnant women and their fetus. To cope with this alarming situation, administration of Sulfadoxine-pyrimethamine (SP) drugs to pregnant women as an intermittent preventive treatment (IPT) from 16 weeks of gestation is recommended by the World Health Organization (WHO) guidelines. We conducted a comprehensive search of published articles related to MiP in last 10 years with predefined keywords or their synonyms. The mapping of malaria in pregnant women showed a prevalence rate up to 35% in many countries. Although IPTp-SP has been implemented in endemic regions since several years but the IPTp-SP coverage percentage vary from country to country and continue to remain below the target of 80%. Major reasons for low IPTp-SP involve gestational age at first prenatal visit, level of education, place of residence, knowledge of IPTp-SP benefits, and use of antenatal services. Several challenges including the emergence of septuple and octuple SP-resistant parasites is reported from many countries which make the prophylactic use of IPTp-SP currently debatable. This narrative review addresses the barriers for optimal use of IPTp-SP and discusses alternative approaches to increase the use and effectiveness of SP intervention for preventing MiP. The COVID pandemic has drastically affected the public health disrupting the management of diseases worldwide. In view of this, a brief summary of COVID impact on MiP situation is also included.
Collapse
Affiliation(s)
- Rodrigue Roman Dongang Nana
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
- Parasitology laboratory, Institute of Medical Research and Medicinal Plants Studies (IMPM), Yaoundé, Cameroon
| | - Joseph Hawadak
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| | - Loick Pradel Kojom Foko
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| | - Amit Kumar
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| | - Shewta Chaudhry
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| | - Aditi Arya
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| | - Vineeta Singh
- Parasite Host Biology group, ICMR-National Institute of Malaria Research (NIMR), New Delhi, India
| |
Collapse
|
34
|
Eboumbou Moukoko CE, Kojom Foko LP, Ayina A, Tornyigah B, Epote AR, Penda IC, Epee Eboumbou P, Ebong SB, Texier G, Nsango SE, Ayong L, Tuikue Ndam N, Same Ekobo A. Effectiveness of Intermittent Preventive Treatment with Sulfadoxine-Pyrimethamine in Pregnancy: Low Coverage and High Prevalence of Plasmodium falciparum dhfr-dhps Quintuple Mutants as Major Challenges in Douala, an Urban Setting in Cameroon. Pathogens 2023; 12:844. [PMID: 37375534 DOI: 10.3390/pathogens12060844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/02/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Intermittent preventive treatment in pregnancy with sulfadoxine and pyrimethamine (IPTp-SP) is a key component in the malaria control strategy implemented in Africa. The aim of this study was to determine IPTp-SP adherence and coverage, and the impact on maternal infection and birth outcomes in the context of widespread SP resistance in the city of Douala, Cameroon. Clinical and demographic information were documented among 888 pregnant women attending 3 health facilities, from the antenatal care visit to delivery. Positive samples were genotyped for P. falciparum gene (dhfr, dhps, and k13) mutations. The overall IPTp-SP coverage (≥three doses) was 17.5%, and 5.1% received no dose. P. falciparum prevalence was 16%, with a predominance of submicroscopic infections (89.3%). Malaria infection was significantly associated with locality and history of malaria, and it was reduced among women using indoor residual spraying. Optimal doses of IPTp-SP were significantly associated with reduced infection among newborns and women (secundiparous and multiparous), but there was no impact of IPTp-SP on the newborn bodyweight. Pfdhfr-Pfdhps quintuple mutants were over-represented (IRNI-FGKAA, IRNI-AGKAA), and sextuple mutants (IRNI-AGKAS, IRNI-FGEAA, IRNI-AGKGS) were also reported. The Pfk13 gene mutations associated with artemisinin resistance were not detected. This study highlights the role of ANC in achieving optimal SP coverage in pregnant women, the mitigated impact of IPTp-SP on malaria outcomes, and the high prevalence of multiple SP-resistant P. falciparum parasites in the city of Douala that could compromise the efficacy of IPTp-SP.
Collapse
Affiliation(s)
- Carole Else Eboumbou Moukoko
- Malaria Research Unit, Centre Pasteur Cameroon, Yaoundé P.O. Box 1274, Cameroon
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
- Laboratory of Parasitology, Mycology and Virology, Postgraduate Training Unit for Health Sciences, Postgraduate School for Pure and Applied Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| | | | - Angèle Ayina
- Malaria Research Unit, Centre Pasteur Cameroon, Yaoundé P.O. Box 1274, Cameroon
- Pharmaceutical Sciences Department, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| | - Bernard Tornyigah
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box 1181, Ghana
- UMR 261 MERIT, Institut de Recherche pour le Développement (IRD), Université de Paris, 75006 Paris, France
| | - Annie Rachel Epote
- Haematology Laboratory, Centre Pasteur Cameroon, Yaoundé P.O. Box 1274, Cameroon
| | - Ida Calixte Penda
- Clinical Sciences Department, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| | - Patricia Epee Eboumbou
- Clinical Sciences Department, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
- Pediatric Wards, Bonassama Hospital, Douala P.O. Box 9023, Cameroon
| | - Serge Bruno Ebong
- Animal Organisms Biology and Physiology Department, Faculty of Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| | - Gaetan Texier
- UMR 257-Vecteurs, Infections Tropicales et Méditerranéennes-VITROME-IRD/SSA/AP-HM, Aix-Marseille University, 13005 Marseille, France
| | - Sandrine Eveline Nsango
- Malaria Research Unit, Centre Pasteur Cameroon, Yaoundé P.O. Box 1274, Cameroon
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| | - Lawrence Ayong
- Malaria Research Unit, Centre Pasteur Cameroon, Yaoundé P.O. Box 1274, Cameroon
| | - Nicaise Tuikue Ndam
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra P.O. Box 1181, Ghana
- UMR 261 MERIT, Institut de Recherche pour le Développement (IRD), Université de Paris, 75006 Paris, France
| | - Albert Same Ekobo
- Department of Biological Sciences, Faculty of Medicine and Pharmaceutical Sciences, The University of Douala, Douala P.O. Box 24157, Cameroon
| |
Collapse
|
35
|
Madanitsa M, Barsosio HC, Minja DTR, Mtove G, Kavishe RA, Dodd J, Saidi Q, Onyango ED, Otieno K, Wang D, Ashorn U, Hill J, Mukerebe C, Gesase S, Msemo OA, Mwapasa V, Phiri KS, Maleta K, Klein N, Magnussen P, Lusingu JPA, Kariuki S, Mosha JF, Alifrangis M, Hansson H, Schmiegelow C, Gutman JR, Chico RM, Ter Kuile FO. Effect of monthly intermittent preventive treatment with dihydroartemisinin-piperaquine with and without azithromycin versus monthly sulfadoxine-pyrimethamine on adverse pregnancy outcomes in Africa: a double-blind randomised, partly placebo-controlled trial. Lancet 2023; 401:1020-1036. [PMID: 36913959 PMCID: PMC10063957 DOI: 10.1016/s0140-6736(22)02535-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/28/2022] [Accepted: 12/06/2022] [Indexed: 03/12/2023]
Abstract
BACKGROUND Intermittent preventive treatment in pregnancy (IPTp) with dihydroartemisinin-piperaquine is more effective than IPTp with sulfadoxine-pyrimethamine at reducing malaria infection during pregnancy in areas with high-grade resistance to sulfadoxine-pyrimethamine by Plasmodium falciparum in east Africa. We aimed to assess whether IPTp with dihydroartemisinin-piperaquine, alone or combined with azithromycin, can reduce adverse pregnancy outcomes compared with IPTp with sulfadoxine-pyrimethamine. METHODS We did an individually randomised, double-blind, three-arm, partly placebo-controlled trial in areas of high sulfadoxine-pyrimethamine resistance in Kenya, Malawi, and Tanzania. HIV-negative women with a viable singleton pregnancy were randomly assigned (1:1:1) by computer-generated block randomisation, stratified by site and gravidity, to receive monthly IPTp with sulfadoxine-pyrimethamine (500 mg of sulfadoxine and 25 mg of pyrimethamine for 1 day), monthly IPTp with dihydroartemisinin-piperaquine (dosed by weight; three to five tablets containing 40 mg of dihydroartemisinin and 320 mg of piperaquine once daily for 3 consecutive days) plus a single treatment course of placebo, or monthly IPTp with dihydroartemisinin-piperaquine plus a single treatment course of azithromycin (two tablets containing 500 mg once daily for 2 consecutive days). Outcome assessors in the delivery units were masked to treatment group. The composite primary endpoint was adverse pregnancy outcome, defined as fetal loss, adverse newborn baby outcomes (small for gestational age, low birthweight, or preterm), or neonatal death. The primary analysis was by modified intention to treat, consisting of all randomised participants with primary endpoint data. Women who received at least one dose of study drug were included in the safety analyses. This trial is registered with ClinicalTrials.gov, NCT03208179. FINDINGS From March-29, 2018, to July 5, 2019, 4680 women (mean age 25·0 years [SD 6·0]) were enrolled and randomly assigned: 1561 (33%; mean age 24·9 years [SD 6·1]) to the sulfadoxine-pyrimethamine group, 1561 (33%; mean age 25·1 years [6·1]) to the dihydroartemisinin-piperaquine group, and 1558 (33%; mean age 24·9 years [6.0]) to the dihydroartemisinin-piperaquine plus azithromycin group. Compared with 335 (23·3%) of 1435 women in the sulfadoxine-pyrimethamine group, the primary composite endpoint of adverse pregnancy outcomes was reported more frequently in the dihydroartemisinin-piperaquine group (403 [27·9%] of 1442; risk ratio 1·20, 95% CI 1·06-1·36; p=0·0040) and in the dihydroartemisinin-piperaquine plus azithromycin group (396 [27·6%] of 1433; 1·16, 1·03-1·32; p=0·017). The incidence of serious adverse events was similar in mothers (sulfadoxine-pyrimethamine group 17·7 per 100 person-years, dihydroartemisinin-piperaquine group 14·8 per 100 person-years, and dihydroartemisinin-piperaquine plus azithromycin group 16·9 per 100 person-years) and infants (sulfadoxine-pyrimethamine group 49·2 per 100 person-years, dihydroartemisinin-piperaquine group 42·4 per 100 person-years, and dihydroartemisinin-piperaquine plus azithromycin group 47·8 per 100 person-years) across treatment groups. 12 (0·2%) of 6685 sulfadoxine-pyrimethamine, 19 (0·3%) of 7014 dihydroartemisinin-piperaquine, and 23 (0·3%) of 6849 dihydroartemisinin-piperaquine plus azithromycin treatment courses were vomited within 30 min. INTERPRETATION Monthly IPTp with dihydroartemisinin-piperaquine did not improve pregnancy outcomes, and the addition of a single course of azithromycin did not enhance the effect of monthly IPTp with dihydroartemisinin-piperaquine. Trials that combine sulfadoxine-pyrimethamine and dihydroartemisinin-piperaquine for IPTp should be considered. FUNDING European & Developing Countries Clinical Trials Partnership 2, supported by the EU, and the UK Joint-Global-Health-Trials-Scheme of the Foreign, Commonwealth and Development Office, Medical Research Council, Department of Health and Social Care, Wellcome, and the Bill-&-Melinda-Gates-Foundation.
Collapse
Affiliation(s)
- Mwayiwawo Madanitsa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi; Department of Clinical Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | - Hellen C Barsosio
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Daniel T R Minja
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - George Mtove
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Reginald A Kavishe
- Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Queen Saidi
- Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - 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
| | - Ulla Ashorn
- Centre for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Samwel Gesase
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Omari A Msemo
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - 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
| | - Nigel Klein
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Pascal Magnussen
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - John P A Lusingu
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Simon Kariuki
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Jacklin F Mosha
- Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Helle Hansson
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christentze Schmiegelow
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - R Matthew Chico
- Department of Disease Control, London School of Hygiene & Tropical Medicine, London, UK
| | - 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
|
36
|
Mbacham HF, Mosume DM, Apinjoh TO, Ntui VN, Moyeh MN, Kalaji LN, Wepnje GB, Ghogomu SM, Dionne JA, Tita ATN, Achidi EA, Anchang-Kimbi JK. Sub-microscopic Plasmodium falciparum parasitaemia, dihydropteroate synthase (dhps) resistance mutations to sulfadoxine-pyrimethamine, transmission intensity and risk of malaria infection in pregnancy in Mount Cameroon Region. Malar J 2023; 22:73. [PMID: 36864514 PMCID: PMC9979436 DOI: 10.1186/s12936-023-04485-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Plasmodium falciparum resistance to intermittent preventive treatment with sulfadoxine-pyrimethamine (IPTp-SP) continues to spread throughout sub-Saharan Africa. This study assessed the occurrence of microscopic and sub-microscopic P. falciparum parasitaemia, dihydropteroate synthase mutations associated with resistance to SP and maternal anaemia in the Mount Cameroon area. METHODS Consenting pregnant women living in semi-rural and semi-urban/urbanized settings were enrolled in this cross-sectional study. Socio-demographic, antenatal and clinical data were documented. Microscopic and sub-microscopic parasitaemia were diagnosed using peripheral blood microscopy and nested polymerase chain reaction (PCR) respectively. The dhps mutations were genotyped by restriction fragment length polymorphism analysis. The presence of A437G, K540E, and A581G was considered a marker for high-level resistance. Haemoglobin levels and anaemia status were determined. RESULTS Among the women, the prevalence of microscopic and sub-microscopic P. falciparum infection were 7.7% (67/874) and 18.6% (93/500) respectively. Predictors of microscopic infection were younger age (< 21 years) (AOR = 2.89; 95% CI 1.29-6.46) and semi-rural settings (AOR = 2.27; 95% CI 1.31-3.96). Determinants of sub-microscopic infection were the rainy season (AOR, 3.01; 95% CI 1.77-5.13), primigravidity (AOR = 0.45; 95% CI 0.21-0.94) and regular ITN usage (AOR = 0.49; 95% CI 0.27-0.90). Of the145 P. falciparum isolates genotyped, 66.9% (97) carried mutations associated with resistance to SP; 33.8% (49), 0%, 52.4% (76) and 19.3% (28) for A437G, K540E, A581G and A437G + A581G respectively. The A581G mutation was associated with ≥ 3 SP doses evident only among sub-microscopic parasitaemia (P = 0.027) and multigravidae (P = 0.009). Women with microscopic infection were more likely from semi-rural settings (AOR = 7.09; 95% CI 2.59-19.42), to report history of fever (AOR = 2.6; 95% CI 1.07-6.31), to harbour parasites with double resistant mutations (AOR = 6.65; 95% CI 1.85-23.96) and were less likely to have received 2 SP doses (AOR = 0.29; 95% CI 1.07-6.31). Microscopic infection decreased Hb levels more than sub-microscopic infection. CONCLUSION The occurrence of sub-microscopic P. falciparum parasites resistant to SP and intense malaria transmission poses persistent risk of malaria infection during pregnancy in the area. ITN usage and monitoring spread of resistance are critical.
Collapse
Affiliation(s)
- Harry F Mbacham
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon
| | - Diange M Mosume
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon
| | - Tobias O Apinjoh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | - Vincent N Ntui
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | - Marcel N Moyeh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | - Laken N Kalaji
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon
| | - Godlove B Wepnje
- Department of Animal Biology and Conservation, University of Buea, Buea, Cameroon
| | - Stephen M Ghogomu
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | - Jodie A Dionne
- Department of Medicine, University of Alabama at Birmingham, Birmingham, USA
| | - Alan T N Tita
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, USA
| | - Eric A Achidi
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | | |
Collapse
|
37
|
Adegbola AJ, Ijarotimi OA, Ubom AE, Adesoji BA, Babalola OE, Hocke EF, Hansson H, Mousa A, Bolaji OO, Alifrangis M, Roper C. A snapshot of the prevalence of dihydropteroate synthase-431V mutation and other sulfadoxine-pyrimethamine resistance markers in Plasmodium falciparum isolates in Nigeria. Malar J 2023; 22:71. [PMID: 36859238 PMCID: PMC9976540 DOI: 10.1186/s12936-023-04487-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/11/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Malaria is a major public health issue with substantial risks among vulnerable populations. Currently, the World Health Organization (WHO) recommends SP-IPTp in the second and third trimesters. However, the efficacy of SP-IPTp is threatened by the emergence of sulfadoxine-pyrimethamine resistant malaria parasites due to single nucleotide polymorphisms in the Plasmodium falciparum dihydrofolate reductase and dihydropteroate synthetase genes. This study aimed to assess the current prevalence of Pfdhfr/Pfdhps mutations in P. falciparum isolates collected from individuals residing in Ile-Ife, Nigeria, and also present maps of the prevalence of Pfdhps 431V and 581G within Nigeria and surrounding countries. METHODS Between October 2020 and April 2021, samples were collected as dried blood spots among 188 participants who showed malaria positivity with a histidine-rich-protein-based rapid diagnostic test (RDT). Nested PCR assays were used to confirm falciparum in the samples with RDT positivity, and to amplify fragments of the Pfdhfr/Pfdhps genes followed by targeted amplicon sequencing. Published data since 2007 on the prevalence of the Pfdhps genotypes in Nigeria and the neighbouring countries were used to produce maps to show the distribution of the mutant genotypes. RESULTS Only 74 and 61 samples were successfully amplified for the Pfdhfr and Pfdhps genes, respectively. At codons resulting in N51I, C59R, and S108N, Pfdhfr carried mutant alleles of 97.3% (72/74), 97.3% (72/74) and 98.6% (73/74), respectively. The Pfdhps gene carried mutations at codons resulting in amino acid changes at 431-436-437-540-581-613; I431V [45.9%, (28/61)], A581G [31.1% (19/61)] and A613S [49.2% (30/61)]. Constructed haplotypes were mainly the triple Pfdhfr mutant 51I-59R-108N (95.9%), and the most common haplotypes observed for the Pfdhps gene were the ISGKAA (32.8%), ISGKGS (8.2%), VAGKAA (14.8%), VAGKAS (9.8%) and VAGKGS (14.8%). In the context of the previously published data, a high prevalence of 431V/581G mutations was found in the study population. It seems quite evident that the Pfdhps 431V, 581G and 613S often co-occur as Pfdhps-VAGKGS haplotype. CONCLUSION This study showed that the prevalence of VAGKGS haplotype seems to be increasing in prevalence. If this is similar in effect to the emergence of 581G in East Africa, the efficacy of SP-IPTp in the presence of these novel Pfdhps mutants should be re-assessed.
Collapse
Affiliation(s)
- Adebanjo J Adegbola
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile Ife, Nigeria.
| | - Omotade A Ijarotimi
- Department of Obstetrics, Gynaecology and Perinatology, Faculty of Clinical Sciences, College of Health Sciences, Obafemi Awolowo University, Ile-Ife, Nigeria.,Department of Obstetrics, Gynaecology and Perinatology, Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Nigeria
| | - Akaninyene E Ubom
- Department of Obstetrics, Gynaecology and Perinatology, Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Nigeria
| | - Bukola A Adesoji
- Department of Nursing Services, Obafemi Awolowo University Teaching Hospitals Complex, Ile-Ife, Nigeria
| | | | - Emma F Hocke
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Helle Hansson
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Andria Mousa
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Oluseye O Bolaji
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Obafemi Awolowo University, Ile Ife, Nigeria
| | - Michael Alifrangis
- Department of Immunology and Microbiology, Centre for Medical Parasitology, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Cally Roper
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
38
|
Kalenda NK, Tshibangu-Kabamba E, Nakagama Y, Kaku N, Kaneko A, Speybroeck N, Kido Y. Usefulness of seasonal malaria chemoprevention in the Sahel. THE LANCET. INFECTIOUS DISEASES 2023; 23:269-270. [PMID: 36327998 DOI: 10.1016/s1473-3099(22)00654-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/25/2023]
Affiliation(s)
- Nadine Kayiba Kalenda
- Institute of Health and Society, Faculty of Public Health, Université Catholique de Louvain, Brussels 1200, Belgium; Department of Public Health, University of Mbujimayi, Mbujimayi, DR Congo.
| | - Evariste Tshibangu-Kabamba
- Departments of Internal Medicine & Basic Sciences, University of Mbujimayi, Mbujimayi, DR Congo; Departments of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Research Center of Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Yu Nakagama
- Departments of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Research Center of Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Natsuko Kaku
- Departments of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Research Center of Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Akira Kaneko
- Departments of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Research Center of Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Niko Speybroeck
- Institute of Health and Society, Faculty of Public Health, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Yasutoshi Kido
- Departments of Virology and Parasitology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Research Center of Infectious Disease Sciences, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
39
|
Nuwa A, Baker K, Bonnington C, Odongo M, Kyagulanyi T, Bwanika JB, Richardson S, Nabakooza J, Achan J, Kajubi R, Odong DS, Nakirunda M, Magumba G, Beinomugisha G, Marasciulo-Rice M, Abio H, Rassi C, Rutazaana D, Rubahika D, Tibenderana J, Opigo J. A non-randomized controlled trial to assess the protective effect of SMC in the context of high parasite resistance in Uganda. Malar J 2023; 22:63. [PMID: 36814301 PMCID: PMC9945593 DOI: 10.1186/s12936-023-04488-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/11/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Until recently, due to widespread prevalence of molecular markers associated with sulfadoxine-pyrimethamine (SP) and amodiaquine (AQ) resistance in east and southern Africa, seasonal malaria chemoprevention (SMC) has not been used at scale in this region. This study assessed the protective effectiveness of monthly administration of SP + AQ (SPAQ) to children aged 3-59 months in Karamoja sub-region, Uganda, where parasite resistance is assumed to be high and malaria transmission is seasonal. METHODS A two-arm quasi-experimental, open-label prospective non-randomized control trial (nRCT) was conducted in three districts. In two intervention districts, 85,000 children aged 3-59 months were targeted to receive monthly courses of SMC using SPAQ during the peak transmission season (May to September) 2021. A third district served as a control, where SMC was not implemented. Communities with comparable malaria attack rates were selected from the three districts, and households with at least one SMC-eligible child were purposively selected. A total cohort of 600 children (200 children per district) were selected and followed using passive surveillance for breakthrough confirmed malaria episodes during the five-month peak transmission season. Malaria incidence rate per person-months and number of malaria episodes among children in the two arms were compared. Kaplan-Meier failure estimates were used to compare the probability of a positive malaria test. Other factors that may influence malaria transmission and infection among children in the two arms were also assessed using multivariable cox proportional hazards regression model. RESULTS The malaria incidence rate was 3.0 and 38.8 per 100 person-months in the intervention and control groups, respectively. In the intervention areas 90.0% (361/400) of children did not experience any malaria episodes during the study period, compared to 15% (29/200) in the control area. The incidence rate ratio was 0.078 (95% CI 0.063-0.096), which corresponds to a protective effectiveness of 92% (95% CI 90.0-94.0) among children in the intervention area. CONCLUSION SMC using SPAQ provided high protective effect against malaria during the peak transmission season in children aged 3-59 months in the Karamoja sub-region of Uganda.
Collapse
Affiliation(s)
| | - Kevin Baker
- grid.475304.10000 0004 6479 3388Malaria Consortium, London, UK ,grid.4714.60000 0004 1937 0626Department of Global Public Health, Karolinska Institute, Stockholm, Sweden
| | | | - Musa Odongo
- grid.452563.3Malaria Consortium Uganda, Kampala, Uganda
| | | | | | - Sol Richardson
- grid.475304.10000 0004 6479 3388Malaria Consortium, London, UK ,grid.12527.330000 0001 0662 3178Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Jane Nabakooza
- grid.415705.2National Malaria Control Division, Ministry of Health, Kampala, Uganda
| | - Jane Achan
- grid.475304.10000 0004 6479 3388Malaria Consortium, London, UK
| | | | | | | | | | | | | | - Hilda Abio
- grid.452563.3Malaria Consortium Uganda, Kampala, Uganda
| | - Christian Rassi
- grid.475304.10000 0004 6479 3388Malaria Consortium, London, UK
| | - Damian Rutazaana
- grid.415705.2National Malaria Control Division, Ministry of Health, Kampala, Uganda
| | - Denis Rubahika
- grid.415705.2National Malaria Control Division, Ministry of Health, Kampala, Uganda
| | | | - Jimmy Opigo
- grid.415705.2National Malaria Control Division, Ministry of Health, Kampala, Uganda
| |
Collapse
|
40
|
Adeleke OT, Oyenuga A, Slusher TM, Gbadero DA. Cluster-randomized controlled trial of intermittent preventive treatment in infancy using sulfadoxine-pyrimethamine (SP-IPTi): a pilot study in Nigeria. J Trop Pediatr 2022; 69:6986163. [PMID: 36633492 DOI: 10.1093/tropej/fmad001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Malaria kills a child in sub-Saharan Africa every 2 min despite widely available interventions including intermittent preventive treatment in infants (IPTi). Since 2010, when World Health Organization (WHO) recommended IPTi, no country has implemented it. To our knowledge, no IPTi study has been conducted in Nigeria. Considering severity of malaria in infancy and urgency to improve malaria prevention, we proposed a study to investigate the efficacy of this intervention in reducing malarial morbidity and mortality. OBJECTIVE(S) The aim of this was to determine the safety and efficacy of SP-IPTi in reducing the prevalence of asymptomatic malaria parasitemia and malarial-associated hospital admissions. METHODS We performed a cluster-randomized controlled trial in 1379 infants. SP was administered alongside routine vaccinations in immunization centers randomized to intervention groups. Infants in control groups received only routine vaccines. Malarial 'morbidity and adverse events were monitored through passive case-detection and cross-sectional surveys'. RESULTS SP-IPTi was safe. There was no statistically significant difference in terms of risks of asymptomatic parasitemia at 9 months, fever or hospitalization between our control and intervention groups. CONCLUSIONS Our study demonstrated that SP-IPTi had no benefit but was well tolerated. WHO and some researchers have also reported declining efficacy of SP, due to increasing drug resistance.
Collapse
Affiliation(s)
| | - Abayomi Oyenuga
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55414, USA
| | - Tina M Slusher
- Department of Pediatrics, Bowen University Teaching Hospital, Ogbomoso 232101, Nigeria.,Department of Pediatrics, University of Minnesota, Minneapolis, MN 55414, USA.,Department of Pediatrics, Hennepin Healthcare, Minneapolis, MN 55404, USA
| | - Daniel A Gbadero
- Department of Pediatrics, Bowen University Teaching Hospital, Ogbomoso 232101, Nigeria
| |
Collapse
|
41
|
Maketa V, Kabalu J, Kabena M, Luzolo F, Muhindo-Mavoko H, Schallig HDFH, Kayentao K, Mens PF, Lutumba P, Tinto H. Comparison of intermittent screening (using ultra-sensitive malaria rapid diagnostic test) and treatment (using a newly registered antimalarial pyronaridine-artesunate-PYRAMAX®) to standard intermittent preventive treatment with sulfadoxine-pyrimethamine for the prevention of malaria in pregnant women living in endemic areas: ULTRAPYRAPREG. Trials 2022; 23:963. [PMID: 36443882 PMCID: PMC9706843 DOI: 10.1186/s13063-022-06884-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is an important malaria control strategy in sub-Saharan Africa. Indeed, it overcomes the risk of misdiagnosis due to low peripheral parasitemia during pregnancy by treating women with SP on predetermined schedules. However, over time, the spread of Plasmodium-resistant strains has threatened this strategy in many countries. As an alternative, the intermittent screening and treatment for pregnancy (ISTp) aims at a monthly screening of pregnant women, preferably by using very sensitive tests such as ultrasensitive rapid diagnostic tests (us-RDTs) and the treatment of positive cases with artemisinin-based combination therapy (ACT) regardless of the presence of symptoms. Unlike IPTp-SP, ISTp prevents overuse of antimalarials limiting the drug pressure on parasites, an advantage which can be potentiated by using an ACT like pyronaridine-artesunate (Pyramax®) that is not yet used in pregnant women in the field. METHODS This study aims to compare the non-inferiority of ISTp using us-RDTs and Pyramax® versus IPTp-SP on malaria in pregnancy through a randomized clinical trial performed in Kisenso, Kinshasa, the Democratic Republic of the Congo, a malaria perennial transmission area. DISCUSSION The results will be essential for the National Malaria Control Program to update the malaria prevention policy in pregnant women in the Democratic Republic of the Congo. TRIAL REGISTRATION ClinicalTrials.gov NCT04783051.
Collapse
Affiliation(s)
- Vivi Maketa
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Japhet Kabalu
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
- Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam (AMC), Laboratory for Experimental Parasitology, Amsterdam Institute for Infection and Immunology, Amsterdam, Netherlands
| | - Melissa Kabena
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Flory Luzolo
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Hypolite Muhindo-Mavoko
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Henk D. F. H. Schallig
- Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam (AMC), Laboratory for Experimental Parasitology, Amsterdam Institute for Infection and Immunology, Amsterdam, Netherlands
| | - Kassoum Kayentao
- Malaria Research and Training Center (MRTC), University of Sciences of Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Petra F. Mens
- Amsterdam University Medical Centres, Academic Medical Centre at the University of Amsterdam (AMC), Laboratory for Experimental Parasitology, Amsterdam Institute for Infection and Immunology, Amsterdam, Netherlands
| | - Pascal Lutumba
- Department of Tropical Medicine, University of Kinshasa (UNIKIN), Kinshasa, Democratic Republic of the Congo
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé – Clinical Research Unit of Nanoro (IRSS-CRUN), Ouagadougou, Burkina Faso
| |
Collapse
|
42
|
Kayentao K, Ongoiba A, Preston AC, Healy SA, Doumbo S, Doumtabe D, Traore A, Traore H, Djiguiba A, Li S, Peterson ME, Telscher S, Idris AH, Kisalu NK, Carlton K, Serebryannyy L, Narpala S, McDermott AB, Gaudinski M, Traore S, Cisse H, Keita M, Skinner J, Hu Z, Zéguimé A, Ouattara A, Doucoure M, Dolo A, Djimdé A, Traore B, Seder RA, Crompton PD. Safety and Efficacy of a Monoclonal Antibody against Malaria in Mali. N Engl J Med 2022; 387:1833-1842. [PMID: 36317783 PMCID: PMC9881676 DOI: 10.1056/nejmoa2206966] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND CIS43LS is a monoclonal antibody that was shown to protect against controlled Plasmodium falciparum infection in a phase 1 clinical trial. Whether a monoclonal antibody can prevent P. falciparum infection in a region in which the infection is endemic is unknown. METHODS We conducted a phase 2 trial to assess the safety and efficacy of a single intravenous infusion of CIS43LS against P. falciparum infection in healthy adults in Mali over a 6-month malaria season. In Part A, safety was assessed at three escalating dose levels. In Part B, participants were randomly assigned (in a 1:1:1 ratio) to receive 10 mg of CIS43LS per kilogram of body weight, 40 mg of CIS43LS per kilogram, or placebo. The primary efficacy end point, assessed in a time-to-event analysis, was the first P. falciparum infection detected on blood-smear examination, which was performed at least every 2 weeks for 24 weeks. At enrollment, all the participants received artemether-lumefantrine to clear possible P. falciparum infection. RESULTS In Part B, 330 adults underwent randomization; 110 were assigned to each trial group. The risk of moderate headache was 3.3 times as high with 40 mg of CIS43LS per kilogram as with placebo. P. falciparum infections were detected on blood-smear examination in 39 participants (35.5%) who received 10 mg of CIS43LS per kilogram, 20 (18.2%) who received 40 mg of CIS43LS per kilogram, and 86 (78.2%) who received placebo. At 6 months, the efficacy of 40 mg of CIS43LS per kilogram as compared with placebo was 88.2% (adjusted 95% confidence interval [CI], 79.3 to 93.3; P<0.001), and the efficacy of 10 mg of CIS43LS per kilogram as compared with placebo was 75.0% (adjusted 95% CI, 61.0 to 84.0; P<0.001). CONCLUSIONS CIS43LS was protective against P. falciparum infection over a 6-month malaria season in Mali without evident safety concerns. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT04329104.).
Collapse
Affiliation(s)
- Kassoum Kayentao
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Aissata Ongoiba
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Anne C Preston
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Sara A Healy
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Safiatou Doumbo
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Didier Doumtabe
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Abdrahamane Traore
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Hamadi Traore
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Adama Djiguiba
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Shanping Li
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Mary E Peterson
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Shinyi Telscher
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Azza H Idris
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Neville K Kisalu
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Kevin Carlton
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Leonid Serebryannyy
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Sandeep Narpala
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Adrian B McDermott
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Martin Gaudinski
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Siriman Traore
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Hamidou Cisse
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Mamadou Keita
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Jeff Skinner
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Zonghui Hu
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Amatigué Zéguimé
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Adama Ouattara
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - M'Bouye Doucoure
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Amagana Dolo
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Abdoulaye Djimdé
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Boubacar Traore
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Robert A Seder
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| | - Peter D Crompton
- From the Malaria Research and Training Center, Mali International Center for Excellence in Research, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali (K.K., A. Ongoiba, S.D., D.D., A.T., H.T., A. Djiguiba, S. Traore, H.C., M.K., A.Z., A. Ouattara, M.D., A. Dolo, A. Djimdé, B.T.); and the Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, Division of Intramural Research (A.C.P., S.A.H., S.L., M.E.P., J.S., P.D.C.), and the Biostatistics Research Branch, Division of Clinical Research (Z.H.), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, and the Vaccine Research Center, NIAID, NIH, Bethesda (S. Telscher, A.H.I., N.K.K., K.C., L.S., S.N., A.B.M., M.G., R.A.S.) - all in Maryland
| |
Collapse
|
43
|
Temporal trends in molecular markers of drug resistance in Plasmodium falciparum in human blood and profiles of corresponding resistant markers in mosquito oocysts in Asembo, western Kenya. Malar J 2022; 21:265. [PMID: 36100912 PMCID: PMC9472345 DOI: 10.1186/s12936-022-04284-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/30/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over the last two decades, the scale-up of vector control and changes in the first-line anti-malarial, from chloroquine (CQ) to sulfadoxine-pyrimethamine (SP) and then to artemether-lumefantrine (AL), have resulted in significant decreases in malaria burden in western Kenya. This study evaluated the long-term effects of control interventions on molecular markers of Plasmodium falciparum drug resistance using parasites obtained from humans and mosquitoes at discrete time points. METHODS Dried blood spot samples collected in 2012 and 2017 community surveys in Asembo, Kenya were genotyped by Sanger sequencing for markers associated with resistance to SP (Pfdhfr, Pfdhps), CQ, AQ, lumefantrine (Pfcrt, Pfmdr1) and artemisinin (Pfk13). Temporal trends in the prevalence of these markers, including data from 2012 to 2017 as well as published data from 1996, 2001, 2007 from same area, were analysed. The same markers from mosquito oocysts collected in 2012 were compared with results from human blood samples. RESULTS The prevalence of SP dhfr/dhps quintuple mutant haplotype C50I51R59N108I164/S436G437E540A581A613 increased from 19.7% in 1996 to 86.0% in 2012, while an increase in the sextuple mutant haplotype C50I51R59N108I164/H436G437E540A581A613 containing Pfdhps-436H was found from 10.5% in 2012 to 34.6% in 2017. Resistant Pfcrt-76 T declined from 94.6% in 2007 to 18.3% in 2012 and 0.9% in 2017. Mutant Pfmdr1-86Y decreased across years from 74.8% in 1996 to zero in 2017, mutant Pfmdr1-184F and wild Pfmdr1-D1246 increased from 17.9% to 58.9% in 2007 to 55.9% and 90.1% in 2017, respectively. Pfmdr1 haplotype N86F184S1034N1042D1246 increased from 11.0% in 2007 to 49.6% in 2017. No resistant mutations in Pfk13 were found. Prevalence of Pfdhps-436H was lower while prevalence of Pfcrt-76 T was higher in mosquitoes than in human blood samples. CONCLUSION This study showed an increased prevalence of dhfr/dhps resistant markers over 20 years with the emergence of Pfdhps-436H mutant a decade ago in Asembo. The reversal of Pfcrt from CQ-resistant to CQ-sensitive genotype occurred following 19 years of CQ withdrawal. No Pfk13 markers associated with artemisinin resistance were detected, but the increased haplotype of Pfmdr1 N86F184S1034N1042D1246 was observed. The differences in prevalence of Pfdhps-436H and Pfcrt-76 T SNPs between two hosts and the role of mosquitoes in the transmission of drug resistant parasites require further investigation.
Collapse
|
44
|
Nana RRD, Bayengue SSB, Mogtomo MLK, Ngane ARN, Singh V. Anti-folate quintuple mutations in Plasmodium falciparum asymptomatic infections in Yaoundé, Cameroon. Parasitol Int 2022; 92:102657. [PMID: 36038059 DOI: 10.1016/j.parint.2022.102657] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/07/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022]
Abstract
A major challenge in the fight to effectively control malaria is the emergence of resistant parasite to drugs used in therapy as well as for chemoprevention. In this study, single nucleotide polymorphisms (SNPs) associated with Plasmodium falciparum resistance to sulfadoxine-pyrimethamine (SP), one of the partner drugs in artemisinin-based therapies (ACTs) were studied in asymptomatic P. falciparum isolates from Cameroon. Dried Blood spots were collected from children with asymptomatic malaria enrolled during a household survey. The P. falciparum dihydrofolate reductase (Pfdhfr), dihydropteroate synthase (Pfdhps) and Kelch 13 genes were amplified and point mutations in these gene sequences were analyzed by sequencing. Among a total of 234 samples collected, 51 showed parasitaemia after microscopic examination of which 47 were P. falciparum mono-infections. Molecular analysis revealed 97.3% of mutant alleles at codons 51I, 59R and 108 N in Pfdhfr gene. In Pfdhps gene the most common mutation was 437G (83.3%); followed by 436A (47.6%) and 436F (28.6%). The association of mutations in the two genes (dhfr + dhps) showed 11 different haplotypes including three sextuple mutants (IRNI + AGKGA, IRNI + AAKGS, IRNI + AGKAS) and one septuple mutant (IRNI + AGKGS). For K13 gene no SNPs were seen in the studied asymptomatic malaria samples. The findings revealed presence of SP-resistant alleles in asymptomatic infected individuals with presence of sextuples and septuple SNPs. This emphasizes that regular profiling of antimalarial drugs resistance markers in such population is essential for malaria control and elimination programmes.
Collapse
Affiliation(s)
- Rodrigue Roman Dongang Nana
- Institute of Medical Research and Medicinal Plants studies, PO Box 13033, Yaoundé, Cameroon; ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi 110077, India
| | | | | | - Anne Rosalie Ngono Ngane
- Department of Biochemistry, Faculty of Science, University of Douala, PO Box 24157, Douala, Cameroon
| | - Vineeta Singh
- ICMR-National Institute of Malaria Research, Dwarka, Sector 8, New Delhi 110077, India.
| |
Collapse
|
45
|
Spatiotemporal spread of Plasmodium falciparum mutations for resistance to sulfadoxine-pyrimethamine across Africa, 1990–2020. PLoS Comput Biol 2022; 18:e1010317. [PMID: 35951528 PMCID: PMC9371298 DOI: 10.1371/journal.pcbi.1010317] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 06/19/2022] [Indexed: 11/26/2022] Open
Abstract
Background Sulfadoxine-pyrimethamine (SP) is recommended in Africa in several antimalarial preventive regimens including Intermittent Preventive Treatment in pregnant women (IPTp), Intermittent Preventive Treatment in infants (IPTi) and Seasonal Malaria Chemoprevention (SMC). The effectiveness of SP-based preventive treatments are threatened in areas where Plasmodium falciparum resistance to SP is high. The prevalence of mutations in the dihydropteroate synthase gene (pfdhps) can be used to monitor SP effectiveness. IPTi-SP is recommended only in areas where the prevalence of the pfdhps540E mutation is below 50%. It has also been suggested that IPTp-SP does not have a protective effect in areas where the pfdhps581G mutation, exceeds 10%. However, pfdhps mutation prevalence data in Africa are extremely heterogenous and scattered, with data completely missing from many areas. Methods and findings The WWARN SP Molecular Surveyor database was designed to summarize dihydrofolate reductase (pfdhfr) and pfdhps gene mutation prevalence data. In this paper, pfdhps mutation prevalence data was used to generate continuous spatiotemporal surface maps of the estimated prevalence of the SP resistance markers pfdhps437G, pfdhps540E, and pfdhps581G in Africa from 1990 to 2020 using a geostatistical model, with a Bayesian inference framework to estimate uncertainty. The maps of estimated prevalence show an expansion of the pfdhps437G mutations across the entire continent over the last three decades. The pfdhps540E mutation emerged from limited foci in East Africa to currently exceeding 50% estimated prevalence in most of East and South East Africa. pfdhps540E distribution is expanding at low or moderate prevalence in central Africa and a predicted focus in West Africa. Although the pfdhps581G mutation spread from one focus in East Africa in 2000, to exceeding 10% estimated prevalence in several foci in 2010, the predicted distribution of the marker did not expand in 2020, however our analysis indicated high uncertainty in areas where pfdhps581G is present. Uncertainty was higher in spatial regions where the prevalence of a marker is intermediate or where prevalence is changing over time. Conclusions The WWARN SP Molecular Surveyor database and a set of continuous spatiotemporal surface maps were built to provide users with standardized, current information on resistance marker distribution and prevalence estimates. According to the maps, the high prevalence of pfdhps540E mutation was to date restricted to East and South East Africa, which is reassuring for continued use of IPTi and SMC in West Africa, but continuous monitoring is needed as the pfdhps540E distribution is expanding. Several foci where pfdhps581G prevalence exceeded 10% were identified. More data on the pfdhps581G distribution in these areas needs to be collected to guide IPTp-SP recommendations. Prevalence and uncertainty maps can be utilized together to strategically identify sites where increased surveillance can be most informative. This study combines a molecular marker database and predictive modelling to highlight areas of concern, which can be used to support decisions in public health, highlight knowledge gaps in certain regions, and guide future research. Despite great success in reducing death and illness from malaria over the last 20 years, the disease is still one of the main leading causes of death in low-income countries with estimated 229 million cases and 409,000 deaths annually. One of the main obstacles in malaria control is the development and spread of drug resistance. Several intermittent preventive treatments depend on the efficacy of the antimalarial drug sulfadoxine-pyrimethamine (SP); Intermittent Preventive Treatment in pregnant women (IPTp), Intermittent Preventive Treatment in infants (IPTi) and Seasonal Malaria Chemoprevention (SMC). Mutations in the dihydropteroate synthase gene (pfdhps) can cause resistance to SP treatment. In this paper, we use pfdhps mutation prevalence data to generate continuous spatiotemporal surface maps of the estimated prevalence of the SP resistance markers in Africa from 1990 to 2020 using a Bayesian geostatistical model. These predictive maps provide much needed insight about where SP can be used as part of preventive treatments. Spatial information on the spread of antimalarial resistance is critical for health organizations to prioritize surveillance measures, and plan control and elimination efforts.
Collapse
|
46
|
Kamau A, Musau M, Mwakio S, Amadi D, Nyaguara A, Bejon P, Seale AC, Berkley JA, Snow RW. Impact of Intermittent Presumptive Treatment for Malaria in Pregnancy on Hospital Birth Outcomes on the Kenyan Coast. Clin Infect Dis 2022; 76:e875-e883. [PMID: 35731850 PMCID: PMC9907553 DOI: 10.1093/cid/ciac509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/10/2022] [Accepted: 06/17/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Intermittent preventive treatment (IPTp) for pregnant women with sulfadoxine-pyrimethamine (SP) is widely implemented for the prevention of malaria in pregnancy and adverse birth outcomes. The efficacy of SP is declining, and there are concerns that IPTp may have reduced impact in areas of high resistance. We sought to determine the protection afforded by SP as part of IPTp against adverse birth outcomes in an area with high levels of SP resistance on the Kenyan coast. METHODS A secondary analysis of surveillance data on deliveries at the Kilifi County Hospital between 2015 and 2021 was undertaken in an area of low malaria transmission and high parasite mutations associated with SP resistance. A multivariable logistic regression model was developed to estimate the effect of SP doses on the risk of low birthweight (LBW) deliveries and stillbirths. RESULTS Among 27 786 deliveries, 3 or more doses of IPTp-SP were associated with a 27% reduction in the risk of LBW (adjusted odds ratio [aOR], 0.73; 95% confidence interval [CI], .64-.83; P < .001) compared with no dose. A dose-response association was observed with increasing doses of SP from the second trimester linked to increasing protection against LBW deliveries. Three or more doses of IPTp-SP were also associated with a 21% reduction in stillbirth deliveries (aOR, 0.79; 95% CI, .65-.97; P = .044) compared with women who did not take any dose of IPTp-SP. CONCLUSIONS The continued significant association of SP on LBW deliveries suggests that the intervention may have a non-malaria impact on pregnancy outcomes.
Collapse
Affiliation(s)
- Alice Kamau
- Correspondence: A. Kamau, KEMRI/Wellcome Trust Research Programme, PO Box 43640-00100, Nairobi, Kenya ()
| | - Moses Musau
- Public Health Research, Kenya Medical Research Institute–Wellcome Trust Research Programme, Nairobi, Kenya
| | - Stella Mwakio
- Epidemiology and Demography, Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - David Amadi
- Epidemiology and Demography, Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Amek Nyaguara
- Epidemiology and Demography, Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya
| | - Philip Bejon
- Epidemiology and Demography, Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya,Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Anna C Seale
- Epidemiology and Demography, Kenya Medical Research Institute–Wellcome Trust Research Programme, Kilifi, Kenya,Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom,College of Health and Medical Sciences, Haramaya University, Harar, Ethiopia,Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - James A Berkley
- Public Health Research, Kenya Medical Research Institute–Wellcome Trust Research Programme, Nairobi, Kenya,Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
47
|
Adderley J, Boulet C, McCann K, McHugh E, Ioannidis LJ, Yeoh LM. Advances in Plasmodium research, an update: highlights from the Malaria in Melbourne 2021 conference. Mol Biochem Parasitol 2022; 250:111487. [DOI: 10.1016/j.molbiopara.2022.111487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/05/2022] [Accepted: 05/18/2022] [Indexed: 11/28/2022]
|
48
|
Vincenz C, Dolo Z, Saye S, Lovett JL, Strassmann BI. Risk factors for placental malaria, sulfadoxine-pyrimethamine doses, and birth outcomes in a rural to urban prospective cohort study on the Bandiagara Escarpment and Bamako, Mali. Malar J 2022; 21:110. [PMID: 35361195 PMCID: PMC8974163 DOI: 10.1186/s12936-022-04125-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria in Mali remains a primary cause of morbidity and mortality, with women at high risk during pregnancy for placental malaria (PM). Risk for PM and its association with birth outcomes was evaluated in a rural to urban longitudinal cohort on the Bandiagara Escarpment and the District of Bamako. METHODS Placental samples (N = 317) were collected from 249 mothers who were participants in a prospective cohort study directed by BIS in the years 2011 to 2019. A placental pathologist and research assistant evaluated the samples by histology in blinded fashion to assess PM infection stage and parasite density. Generalized estimating equations (GEE) were used to model the odds of PM infection. RESULTS In a multivariable model, pregnancies in Bamako, beyond secondary education, births in the rainy season (instead of the hot dry season), and births to women who had ≥ 3 doses of sulfadoxine-pyrimethamine (SP) instead of no doses were associated with reduced odds of experiencing PM (active and past infections combined). Births in later years of the study were strongly associated with reduced odds of PM. Maternal age, which was positively associated with offspring year of birth, was significant as a predictor of PM only if offspring year of birth was omitted from the model. Gravidity was positively associated with both maternal age and offspring year of birth such that if either variable was included in the model, then gravidity was no longer significant. However, if maternal age or year of offspring birth were not adjusted for, then the odds of PM were nearly two-fold higher in primigravida compared to multigravida. Birth outcomes improved (+ 285 g birth weight, + 2 cm birth length, + 75 g placental weight) for women who had ≥ 3 doses of SP compared to no doses, but no difference was detected in birth weight or length for women who had 2 instead of ≥ 3 SP doses. However, at 2 instead of ≥ 3 doses placentas were 36 g lighter and the odds of low birth weight (< 2500 g) were 14% higher. Severe parasite densities (> 10% erythrocytes infected) were significantly associated with decreases in birth weight, birth length, and placental weight, as were chronic PM infections. The women who received no SP during pregnancy (7% of the study total) were younger and lacked primary school education. The women who received ≥ 3 doses of SP came from more affluent families. CONCLUSIONS Women who received no doses of SP during pregnancy experienced the most disadvantageous birth outcomes in both Bamako and on the Bandiagara Escarpment. Such women tended to be younger and to have had no primary school education. Targeting such women for antenatal care, which is the setting in which SP is most commonly administered in Mali, will have a more positive impact on public health than focusing on the increment from two to three doses of SP, although that increment is also desirable.
Collapse
Affiliation(s)
- Claudius Vincenz
- Research Center for Group Dynamics, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Zachary Dolo
- Independent Investigator, Bandiagara Cercle, Mali
| | - Serou Saye
- Independent Investigator, Bandiagara Cercle, Mali
| | - Jennie L Lovett
- Department of Anthropology, University of Michigan, Ann Arbor, MI, USA
| | - Beverly I Strassmann
- Research Center for Group Dynamics, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA. .,Department of Anthropology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Plowe CV. Malaria chemoprevention and drug resistance: a review of the literature and policy implications. Malar J 2022; 21:104. [PMID: 35331231 PMCID: PMC8943514 DOI: 10.1186/s12936-022-04115-8] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/03/2022] [Indexed: 01/19/2023] Open
Abstract
Chemoprevention strategies reduce malaria disease and death, but the efficacy of anti-malarial drugs used for chemoprevention is perennially threatened by drug resistance. This review examines the current impact of chemoprevention on the emergence and spread of drug resistant malaria, and the impact of drug resistance on the efficacy of each of the chemoprevention strategies currently recommended by the World Health Organization, namely, intermittent preventive treatment in pregnancy (IPTp); intermittent preventive treatment in infants (IPTi); seasonal malaria chemoprevention (SMC); and mass drug administration (MDA) for the reduction of disease burden in emergency situations. While the use of drugs to prevent malaria often results in increased prevalence of genetic mutations associated with resistance, malaria chemoprevention interventions do not inevitably lead to meaningful increases in resistance, and even high rates of resistance do not necessarily impair chemoprevention efficacy. At the same time, it can reasonably be anticipated that, over time, as drugs are widely used, resistance will generally increase and efficacy will eventually be lost. Decisions about whether, where and when chemoprevention strategies should be deployed or changed will continue to need to be made on the basis of imperfect evidence, but practical considerations such as prevalence patterns of resistance markers can help guide policy recommendations.
Collapse
|
50
|
Lingani M, Zango SH, Valéa I, Somé G, Sanou M, Samadoulougou SO, Ouoba S, Rouamba E, Robert A, Dramaix M, Donnen P, Tinto H. Low birth weight and its associated risk factors in a rural health district of Burkina Faso: a cross sectional study. BMC Pregnancy Childbirth 2022; 22:228. [PMID: 35313840 PMCID: PMC8935822 DOI: 10.1186/s12884-022-04554-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 03/09/2022] [Indexed: 12/30/2022] Open
Abstract
Background Low birth weight (LBW) is a major factor of neonate mortality that particularly affects developing countries. However, the scarcity of data to support decision making to reduce LBW occurrence is a major obstacle in sub-Saharan Africa. The aim of this research was to determine the prevalence and associated factors of LBW at the Yako health district in a rural area of Burkina Faso. Methods A cross sectional survey was conducted at four peripheral health centers among mothers and their newly delivered babies. The mothers’ socio-demographic and obstetrical characteristics were collected by face-to-face interview or by review of antenatal care books. Maternal malaria was tested by standard microscopy and neonates’ birth weights were documented. Multivariate logistic regression was used to determine factors associated with LBW. A p-value < 0.05 was considered statistically significant. Results Of 600 neonates examined, the prevalence of low birth weight was 11.0%. Adjustment for socio-demographic characteristic, medical conditions, obstetrical history, malaria prevention measures by multivariate logistic regression found that being a primigravid mother (aOR = 1.8, [95% CI: 1.1–3.0]), the presence of malaria infection (aOR = 1.9, [95% CI: 1.1–3.5]), the uptake of less than three doses of sulfadoxine-pyrimethamine for the intermittent preventive treatment of malaria in pregnancy (IPTp-SP) (aOR = 2.2, [95% CI: 1.3–3.9]), the presence of maternal fever at the time of delivery (aOR = 2.8, [95% CI: 1.5–5.3]) and being a female neonate (aOR = 1.9, [95% CI: 1.1–3.3]) were independently associated with an increased risk of LBW occurrence. The number of antenatal visits performed by the mother during her pregnancy did not provide any direct protection for low birth weight. Conclusion The prevalence of LBW remained high in the study area. Maternal malaria, fever and low uptake of sulfadoxine-pyrimethamine doses were significantly associated with LBW and should be adequately addressed by public health interventions.
Collapse
Affiliation(s)
- Moussa Lingani
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso. .,École de Santé publique, Université Libre de Bruxelles, Bruxelles, Belgium.
| | - Serge Henri Zango
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso.,Epidemiology and Biostatistics Research Division, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Innocent Valéa
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Georges Somé
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Maïmouna Sanou
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Sékou O Samadoulougou
- Evaluation Platform on Obesity Prevention, Quebec Heart and Lung Institute Research Center, Quebec, Canada
| | - Serge Ouoba
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Eli Rouamba
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| | - Annie Robert
- Epidemiology and Biostatistics Research Division, Institut de recherche expérimentale et clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Michèle Dramaix
- École de Santé publique, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Philippe Donnen
- École de Santé publique, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), Nanoro, Burkina Faso
| |
Collapse
|