1
|
Gebreegziabher EA, Ouattara M, Bountogo M, Coulibaly B, Boudo V, Ouedraogo T, Lebas E, Hu H, O'Brien KS, Hsiang MS, Glidden DV, Arnold BF, Lietman TM, Sié A, Oldenburg CE. The role of Seasonal Malaria Chemoprevention in the effect of Azithromycin on Child Mortality: A Secondary Analysis of the CHAT Cluster Randomized Clinical Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.30.25326740. [PMID: 40343013 PMCID: PMC12060949 DOI: 10.1101/2025.04.30.25326740] [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: 05/11/2025]
Abstract
Objective Mass treatment with azithromycin (AZ) and administration of seasonal malaria chemoprevention (SMC) are both effective in reducing mortality among children under 5. However, it is not clear whether the benefit of AZ for mortality varies in the presence of routine SMC administration. The objective of this study was to examine whether the effect of mass AZ distribution on all-cause mortality among children less than 5 years of age varies with SMC administration season or SMC coverage. Methods This was a secondary analysis of the Community Health with Azithromycin Trial (CHAT), a cluster randomized placebo-controlled trial of 341 communities in the Nouna District of Burkina Faso. Communities randomized to intervention received treatment with twice yearly mass AZ while control communities receive placebo. All communities received SMC as standard-of-care. SMC administration and coverage data were provided from National Malaria Control Program. SMC administration season was defined as the period during and immediately following SMC (July-December) versus the months of no SMC (January-June). SMC coverage was assessed as proportion of the population covered and by whether it was below or above a threshold of 80%. We used Poisson regression models with person-time at risk used as an offset and robust standard error to analyze mortality rates by treatment group and SMC subgroups and assessed interaction on both the multiplicative and additive scales. Results Mortality was higher in SMC seasons for both arms, with a mortality rate of 10.3 per 1,000 person-years (95% CI: 9.0 to 11.6) in SMC seasons and 7.9 (95% CI: 6.9 to 9.0) in non-SMC seasons. Compared to placebo, the mortality rate in AZ clusters was 0.77 (95% CI: 0.60 to 0.98) during SMC season, while it was 0.89 (95% CI: 0.68 to 1.15) during the non-SMC seasons. The effect of AZ compared to placebo in clusters with <80% SMC coverage was 0.73 95%CI (0.56 to 0.96) and in clusters with ≥80% SMC coverage, it was 1.0 95%CI (0.59 to 1.69). The interaction between AZ and SMC season or coverage was not statistically significant on the additive or multiplicative scales. Conclusion While our findings did not reach statistical significance, they raise the question of whether prioritizing MDA AZ during high transmission periods or in regions with low SMC coverage could be beneficial. Further research is needed to determine if targeting these periods or areas could lead to greater reductions in child mortality. Trial Registration ClinicalTrials.gov Identifier: NCT03676764.
Collapse
Affiliation(s)
- Elisabeth A Gebreegziabher
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | | | | | | | - Valentin Boudo
- Centre de Recherche en Sante de Nouna, Nouna, Burkina Faso
| | | | - Elodie Lebas
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
| | - Huiyu Hu
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
| | - Kieran S O'Brien
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle S Hsiang
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Malaria Elimination Initiative, Institute for Global Health Sciences, University of California San Francisco, San Francisco, CA, USA
- Division of Pediatric Infectious Diseases, Department of Pediatrics, San Francisco, CA, USA
| | - David V Glidden
- Department of Epidemiology and Biostatistics, University of California, San Francisco
| | - Benjamin F Arnold
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas M Lietman
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Ali Sié
- Centre de Recherche en Sante de Nouna, Nouna, Burkina Faso
| | - Catherine E Oldenburg
- Francis I. Proctor Foundation, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
2
|
Roh ME, Gutman JR, Murphy M, Hill J, Madanitsa M, Kakuru A, Barsosio HC, Kariuki S, Lusingu JP, Mosha F, Kajubi R, Kamya MR, Mathanga D, Chinkhumba J, Laufer MK, Mlugu E, Kamuhabwa AA, Aklillu E, Minzi O, Okoro RN, Geidam AD, Ohieku JD, Desai M, Jagannathan P, Dorsey G, ter Kuile FO. Dihydroartemisinin-piperaquine versus sulfadoxine-pyrimethamine for intermittent preventive treatment of malaria in pregnancy: a systematic review and individual participant data meta-analysis. EClinicalMedicine 2025; 83:103202. [PMID: 40370584 PMCID: PMC12076784 DOI: 10.1016/j.eclinm.2025.103202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 05/16/2025] Open
Abstract
Background High-grade Plasmodium falciparum resistance to sulfadoxine-pyrimethamine in east and southern Africa has prompted trials evaluating intermittent preventive treatment in pregnancy (IPTp) with dihydroartemisinin-piperaquine as an alternative to sulfadoxine-pyrimethamine. We aimed to provide an updated and comprehensive review of trials conducted in areas of high P. falciparum resistance that compared the efficacy of two types of IPTp regimens on maternal, birth, and infant outcomes. Methods We conducted two-stage, individual participant data meta-analyses of randomised trials comparing IPTp with dihydroartemisinin-piperaquine to sulfadoxine-pyrimethamine on maternal, birth, and infant outcomes. We searched the WHO International Clinical Trials Registry Platform, ClinicalTrials.Gov, PubMed, and the Malaria in Pregnancy Consortium Library, on July 30, 2020 (updated on September 24, 2024), without restrictions by publication date, peer-review status, or language. Eligible trials enrolled HIV-uninfected pregnant women, followed participants to delivery, included participants with no prior IPTp use during the current pregnancy, and were conducted in areas with high-level parasite resistance to sulfadoxine-pyrimethamine (i.e., PfDHPS 540E ≥ 90% and/or 581G>0%). Only singleton pregnancies were analysed. The primary endpoint was a composite measure of any adverse pregnancy outcome defined as fetal or neonatal loss, small-for-gestational age, low birthweight, or preterm birth. Summary estimates were generated using a random-effects model. Gravidity subgroup analyses were performed. Causal mediation analyses were used to investigate the maternal mechanisms underlying the effect of IPTp regimens on birth outcomes. The meta-analysis is registered in PROSPERO (CRD42020196127). Findings Of 85 screened records, six trials (one multi-country trial) from Kenya, Malawi, Uganda and Tanzania contributed data on 6646 pregnancies. Compared to sulfadoxine-pyrimethamine, dihydroarteminsinin-piperaquine was associated with a 69% [95% CI: 45%-82%] lower incidence of clinical malaria during pregnancy, a 62% [37%-77%] lower risk of placental parasitaemia, and a 17% [0%-31%] lower incidence of moderate maternal anaemia. In contrast, sulfadoxine-pyrimethamine was associated with higher mean maternal weight gain (34 g/week [17-51]). There were no statistically significant differences in the composite adverse pregnancy outcome (RR = 1.05 [0.92-1.19]; I 2 = 48%). Individual components of the primary outcome showed no statistically significant differences in the risks of fetal loss (RR = 0.94 [0.61-1.46]), preterm birth (RR = 0.93 [0.76-1.14]), low birthweight (RR = 1.09 [0.83-1.43]), or neonatal loss (RR = 0.73 [0.42-1.26]), though findings may have been underpowered. Small-for-gestational-age risk was 15% (3%-24%) lower in the sulfadoxine-pyrimethamine arm, particularly among multigravidae (a 22% reduction vs 9% in primigravidae). Among multigravidae, infant stunting and underweight by two months was 20% [8%-30%] and 35% [17%-49%] lower in the sulfadoxine-pyrimethamine arm compared to dihydroartemisinin-piperaquine. Compared to dihydroartemisinin-piperaquine, sulfadoxine-pyrimethamine was associated with higher mean newborn birthweight (mean difference (MD) = 50 g [95% CI: 13-88]; p = 0.0090, I2 = 61%) and BWGA z-scores (MD = 0.12 [95% CI: 0.05-0.20]; p = 0.0012, I2 = 51%), but not gestational age at birth (MD = 0 weeks [95% CI: -0.11 to 0.12]; p = 0.94; I2 = 42%). Infant wasting by two months was 13% [3%-22%] lower in the sulfadoxine-pyrimethamine arm, regardless of gravidity. Mediation analyses indicated that 15% [0%-19%] of sulfadoxine-pyrimethamine's superior effect on small-for-gestational-age risk was mediated by its greater impact on gestational weight gain. Interpretation In areas with high P. falciparum sulfadoxine-pyrimethamine resistance, dihydroartemisinin-piperaquine offers superior antimalarial efficacy than sulfadoxine-pyrimethamine. However, replacing sulfadoxine-pyrimethamine with dihydroartemisinin-piperaquine alone may not lead to improved maternal and infant health outcomes. Instead, it could result in slightly reduced gestational weight gain and a modest increase in the risk of small-for-gestational age births, and poor infant growth by two months of age. Future research evaluating alternative strategies for IPTp are needed. Funding This work was supported by the Bill and Melinda Gates Foundation and Eunice Kennedy Shriver National Institute of Child Health and Human Development.
Collapse
Affiliation(s)
- Michelle E. Roh
- Institute for Global Health Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Julie R. Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Maxwell Murphy
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Mywayiwawo Madanitsa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Hellen C. Barsosio
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - John P.A. Lusingu
- National Institute for Medical Research (NIMR), Tanga Medical Research Centre, Tanga, Tanzania
| | - Frank Mosha
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Moses R. Kamya
- School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Don Mathanga
- Malaria Alert Center, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Jobiba Chinkhumba
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Miriam K. Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eulambius Mlugu
- Department of Pharmaceutics, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Appolinary A.R. Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Eleni Aklillu
- Department of Global Public Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Omary Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Roland Nnaemeka Okoro
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy, University of Maiduguri, Maiduguri, Nigeria
| | - Ado Danazumi Geidam
- Department of Obstetrics and Gynaecology, University of Maiduguri Teaching Hospital, Maiduguri, Nigeria
| | - John David Ohieku
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy, University of Maiduguri, Maiduguri, Nigeria
| | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | | | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Feiko O. ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
3
|
Luntamo M, Hallamaa L, Kulmala T, Maleta K, Ashorn P. Effect of Antenatal Monthly Sulfadoxine-Pyrimethamine, Alone or with Azithromycin, on Gestational Weight Gain and Anemia during Pregnancy and One Month Postpartum in Malawi: A Randomized Controlled Trial Secondary Analysis. Am J Trop Med Hyg 2025; 112:931-941. [PMID: 39903927 PMCID: PMC11965741 DOI: 10.4269/ajtmh.23-0829] [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: 11/26/2023] [Accepted: 09/24/2024] [Indexed: 02/06/2025] Open
Abstract
Low gestational weight gain (GWG) and prenatal anemia are associated with adverse pregnancy, maternal and infant health outcomes. In a secondary analysis of a single-center, randomized, partially placebo-controlled, outcome assessor-blinded, controlled trial conducted in Malawi from 2003 to 2006, when antiretroviral treatment (ART) for HIV was not widely available, we studied whether GWG can be increased and the prevalence of maternal anemia decreased during pregnancy and at 1 month postpartum through the intermittent preventive treatment in pregnancy (IPTp) of maternal malaria and reproductive tract infections. The participants (≥15-year-old women with uncomplicated second trimester single pregnancies) received either sulfadoxine (1,500 mg) and pyrimethamine (75 mg; SP) twice (control group, n = 433), monthly SP (n = 439), or monthly SP and azithromycin (1,000 mg) twice (AZI-SP, n = 441) during pregnancy. The mean weekly GWG in the sample was 256 g/week. The participants in the monthly SP group gained, on average (95% CI), 4 g (-13 to 20; P = 0.671), and those in the AZI-SP group gained 25 g (8-41; P = 0.003) more weight per week than control group participants. Among HIV-positive participants (12%), the differences were larger and also significant between the monthly SP group and control group. Mean hemoglobin and anemia prevalence did not differ between the groups during pregnancy or postnatally. The data support a hypothesis that IPTp with monthly SP and two doses of azithromycin can increase GWG, especially among HIV-positive women who are not on ART, possibly through the reduction of infections, inflammation, and effects on the maternal gut microbiome.
Collapse
Affiliation(s)
- Mari Luntamo
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| | - Lotta Hallamaa
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| | - Teija Kulmala
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
| | - Kenneth Maleta
- Department of Nutrition and Dietetics, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Per Ashorn
- Faculty of Medicine and Health Technology, Center for Child, Adolescent, and Maternal Health Research, Tampere University, Tampere, Finland
- Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
4
|
Mwebaza N, Roh ME, Geng YZ, Opio L, Opira B, Marzan F, Mwima MW, Ssemukuye T, Guo K, Gingrich D, Fotaki N, Kakuru A, Kizza J, Aguti M, Adrama H, Kamya M, Dorsey G, Rosenthal PJ, Aweeka FT, Huang L. Drug-Drug Interaction Between Dihydroartemisinin-Piperaquine and Sulfadoxine-Pyrimethamine During Malaria Chemoprevention in Pregnant Women. Clin Pharmacol Ther 2025; 117:506-514. [PMID: 39402742 PMCID: PMC11747899 DOI: 10.1002/cpt.3471] [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: 06/06/2024] [Accepted: 09/21/2024] [Indexed: 11/20/2024]
Abstract
Co-administration of dihydroartemisinin-piperaquine (DP) and sulfadoxine-pyrimethamine (SP) for intermittent preventive treatment of malaria in pregnancy (IPTp) may be superior in preventing adverse birth outcomes compared with either therapy alone, but potential drug-drug interactions require investigation. We conducted intensive and sparse pharmacokinetic (PK) studies in a subset of Ugandan women participating in a randomized controlled trial of monthly IPTp with SP vs. DP vs. DP + SP. Intensive PK sampling was performed from day 0 to 23 after dosing at 28 weeks gestation in 87 participants across treatment arms. Sparse sampling was performed on day 28 (trough) after dosing at 20 and 28 gestational weeks in additional 196 participants receiving SP vs. DP + SP. Intensive PK analysis demonstrated that compared with SP alone, co-administration of DP + SP was associated with lower maximal concentrations, the area under the concentration-time curves (AUC), and day 23 concentrations of sulfadoxine (25%, 25%, and 27%) and pyrimethamine (26%, 34%, and 32%) (P < 0.05 for all comparisons). Sparse PK results demonstrated participants co-administered DP + SP had lower trough concentrations after dosing at 20 and 28 gestational weeks for sulfadoxine (6%, P = 0.68 and 31%, P = 0.023, respectively) and pyrimethamine (18%, P = 0.032 and 33%, P < 0.001, respectively) compared with SP alone. Co-administration of DP + SP was associated with a 19% reduction in piperaquine AUC (P = 0.046), but no significant difference in other PK parameters compared with DP alone. In summary, co-administration of DP + SP was associated with significantly reduced SP exposure, with a greater magnitude during the third vs. second trimester. The clinical consequences of this interaction are yet unknown.
Collapse
Affiliation(s)
- Norah Mwebaza
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Pharmacology and Therapeutics, Makerere University, Kampala, Uganda
| | - Michelle E. Roh
- Institute for Global Health Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Yourong Z Geng
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Leonard Opio
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Bishop Opira
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Florence Marzan
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Moses W. Mwima
- Infectious Disease Research Collaboration, Kampala, Uganda
| | | | - Kevin Guo
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - David Gingrich
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Nikoletta Fotaki
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - Abel Kakuru
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Jimmy Kizza
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Miriam Aguti
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Harriet Adrama
- Infectious Disease Research Collaboration, Kampala, Uganda
| | - Moses Kamya
- Infectious Disease Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, CA, USA
| | | | - Francesca T. Aweeka
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| | - Liusheng Huang
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Gore-Langton GR, Madanitsa M, Barsosio HC, Minja DTR, Mosha J, Kavishe RA, Mtove G, Gesase S, Msemo OA, Kariuki S, Otieno K, Phiri KS, Lusingu JPA, Mukerebe C, Manjurano A, Ikigo P, Saidi Q, Onyango ED, Schmiegelow C, Dodd J, Hill J, Hansson H, Alifrangis M, Gutman J, Hunter PJ, Klein N, Ashorn U, Khalil A, Cairns M, ter Kuile FO, Chico RM. Prevalence and risk factors of curable sexually transmitted and reproductive tract infections and malaria co-infection among pregnant women at antenatal care booking in Kenya, Malawi and Tanzania: a cross-sectional study of randomised controlled trial data. BMJ PUBLIC HEALTH 2024; 2:e000501. [PMID: 40018559 PMCID: PMC11816199 DOI: 10.1136/bmjph-2023-000501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 07/30/2024] [Indexed: 03/01/2025]
Abstract
Objectives Malaria and curable sexually transmitted and reproductive tract infections (STIs/RTIs) are associated with adverse pregnancy outcomes. This study reports the prevalence and risk factors of curable STIs/RTIs, STI/RTI co-infection and STI/RTI and malaria co-infection among HIV-negative pregnant women at their first antenatal care visit in Kenya, Malawi and Tanzania. Methods HIV-negative pregnant women of all gravidae (n=4680) were screened for syphilis with point-of-care tests and treated if positive. Separately, women provided blood samples (n=4569) for rapid plasma reagin (RPR) testing; positive cases were confirmation by Treponema pallidum particle agglutination (TPPA). Women also provided dried blood spots for batch testing of malaria by retrospective polymerase chain reaction (PCR (n=4226) methods. A randomly selected subgroup of women provided vaginal swabs for chlamydia, gonorrhoea and trichomoniasis testing by retrospective PCR batch testing (n=1431), and bacterial vaginosis diagnosis by Nugent scoring (n=1402). Results Malaria prevalence was 14.6% (95% CI 13.6 to 15.7), 45.9% (43.4 to 48.4) of women were positive for at least one curable STI/RTI and 6.7% (5.5 to 8.1) were co-infected with malaria and a curable STI/RTI. Prevalence of individual STIs/RTIs ranged from 28.5% (26.2 to 30.9) for bacterial vaginosis to 14.5% (12.7 to 16.4) for trichomoniasis, 13.8% (12.1 to 15.7) for chlamydia, 2.7% (1.9 to 3.6) for gonorrhoea and 1.7% (1.4 to 2.2) for RPR/TPPA-confirmed syphilis. The prevalence of STI/RTI co-infection was 10.1% (8.7 to 11.8). Paucigravidae, at highest risk of malaria, were also at greater risk of having chlamydia, gonorrhoea and bacterial vaginosis than multigravidae. Conclusions Of women infected with malaria, 49.0% also had a curable STI/RTI and one in five women with at least one STI/RTI were co-infected with more than one STI/RTI. Current antenatal interventions that address malaria and curable STIs/RTIs remain suboptimal. New approaches to preventing and managing these infections in pregnancy are urgently needed. Trial registration number NCT03208179.
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, Center for Global Health Research, Kisumu, Kenya
| | - Daniel T R Minja
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Jacklin Mosha
- National Institute for Medical Research, Mwanza Centre, Mwanza, Tanzania
| | - Reginald A Kavishe
- National Institute for Medical Research, Mwanza Centre, Mwanza, Tanzania
| | - George Mtove
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Samwel Gesase
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Omari A Msemo
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Simon Kariuki
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Kephas Otieno
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Kamija S Phiri
- Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - John P A Lusingu
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania
| | - Crispin Mukerebe
- National Institute for Medical Research, Mwanza Centre, Mwanza, Tanzania
| | | | - Pius Ikigo
- National Institute for Medical Research, Mwanza Centre, Mwanza, Tanzania
| | - Queen Saidi
- Kilimanjaro Clinical Research Institute and Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Eric D Onyango
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
| | - Christentze Schmiegelow
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Gynaecology and Obstetrics, Copenhagen University Hospital—North Zealand, Hilleroed, Denmark
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Helle Hansson
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen and Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| | - Julie Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centre for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Patricia Jean Hunter
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Nigel Klein
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ulla Ashorn
- Centre for Child Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Asma Khalil
- Fetal Medicine Unit, Saint George’s Hospital, London, UK
| | - Matt Cairns
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| | - Feiko O ter Kuile
- Kenya Medical Research Institute, Center for Global Health Research, Kisumu, Kenya
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - R Matthew Chico
- Department of Disease Control, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
8
|
Roh ME, Gutman J, Murphy M, Hill J, Madanitsa M, Kakuru A, Barsosio HC, Kariuki S, Lusingu JPA, Mosha F, Kajubi R, Kamya MR, Mathanga D, Chinkhumba J, Laufer MK, Mlugu E, Kamuhabwa AAR, Aklillu E, Minzi O, Okoro RN, Geidam AD, Ohieku JD, Desai M, Jagannathan P, Dorsey G, Ter Kuile FO. Dihydroartemisinin-piperaquine versus sulfadoxine-pyrimethamine for intermittent preventive treatment of malaria in pregnancy: a systematic review and individual participant data meta-analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.23.24315401. [PMID: 39649586 PMCID: PMC11623747 DOI: 10.1101/2024.11.23.24315401] [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: 12/11/2024]
Abstract
Background High-grade Plasmodium falciparum resistance to sulfadoxine-pyrimethamine in East and Southern Africa has prompted numerous trials evaluating intermittent preventive treatment in pregnancy (IPTp) with dihydroartemisinin-piperaquine as an alternative to sulfadoxine-pyrimethamine. Methods We conducted individual participant data meta-analyses of randomised trials comparing IPTp with dihydroartemisinin-piperaquine to sulfadoxine-pyrimethamine on maternal, birth, and infant outcomes. We searched the WHO International Clinical Trials Registry Platform, ClinicalTrials.Gov, PubMed, and the Malaria in Pregnancy Consortium Library. Eligible trials enrolled HIV-uninfected pregnant women, followed participants to delivery, included participants with no prior IPTp use during the current pregnancy, and were conducted in areas with high-level parasite resistance to sulfadoxine-pyrimethamine (i.e., PfDHPS 540E≥90% and/or 581G>0%). Only singleton pregnancies were analysed. Meta-analyses used a two-stage approach: first, study-specific estimates were generated and then pooled using a random-effects model. Gravidity subgroup analyses were performed. Causal mediation analyses were used to investigate the maternal mechanisms underlying the effect of IPTp regimens on birth outcomes. The meta-analysis is registered in PROSPERO (CRD42020196127). Findings Of 85 screened records, six trials (one multi-country trial) contributed data on 6646 pregnancies. Compared to sulfadoxine-pyrimethamine, dihydroarteminsinin-piperaquine was associated with a 69% [95% CI: 45%-82%] lower incidence of clinical malaria during pregnancy, a 62% [37%-77%] lower risk of placental parasitaemia, and a 17% [0%-31%] lower incidence of moderate maternal anaemia (Hb<9 g/dL). In contrast, sulfadoxine-pyrimethamine was associated with higher mean weekly maternal weight gain (34 grams/week [17-51]). There were no statistically significant differences in the composite adverse pregnancy outcome between the two IPTp regimens (RR=1·05 [95% CI: 0·92-1·19]; I 2=48%), although the risk of small-for-gestational-age was 15% [3%-24%] lower in the sulfadoxine-pyrimethamine arm. Among multigravidae, participants of the sulfadoxine-pyrimethamine arm were 20% [8%-30%] and 35% [17%-49%] less likely to have stunted and underweight infants by two months compared to the dihydroartemisinin-piperaquine arm. Infant wasting by two months was 13% [3%-22%] lower in the sulfadoxine-pyrimethamine arm, regardless of gravidity. Mediation analyses indicated that 15% [0%-19%] of sulfadoxine-pyrimethamine's superior effect on reducing small-for-gestational-age risk was mediated by its greater impact on gestational weight gain. Interpretation In areas of high P. falciparum sulfadoxine-pyrimethamine resistance, dihydroartemisin-inpiperaquine is a more efficacious antimalarial than sulfadoxine-pyrimethamine. However, replacing sulfadoxine-pyrimethamine with dihydroartemisinin-piperaquine alone will not result in better maternal, birth, or infant outcomes. It could increase the risk of SGA, since much of the effect of sulfadoxine-pyrimethamine may be exerted through non-malarial mechanisms. Future research evaluating the alternative strategies for IPTp are needed, including with the combination of sulfadoxine-pyrimethamine and dihydroartemisinin-piperaquine. Funding This work was supported by the Bill and Melinda Gates Foundation and Eunice Kennedy Shriver National Institute of Child Health and Human Development.
Collapse
Affiliation(s)
- Michelle E Roh
- Institute for Global Health Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Julie Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - Maxwell Murphy
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Mywayiwawo Madanitsa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Hellen C Barsosio
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - John P A Lusingu
- National Institute for Medical Research (NIMR), Tanga Medical Research Centre, Tanga, Tanzania
| | - Frank Mosha
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Moses R Kamya
- School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Don Mathanga
- Malaria Alert Center, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Jobiba Chinkhumba
- Department of Health Systems and Policy, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Miriam K Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eulambius Mlugu
- Department of Pharmaceutics, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Appolinary A R Kamuhabwa
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Eleni Aklillu
- Department of Global Public Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Omary Minzi
- Department of Clinical Pharmacy and Pharmacology, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Roland Nnaemeka Okoro
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy, University of Maiduguri, Maiduguri, Nigeria
| | - Ado Danazumi Geidam
- Department of Obstetrics and Gynaecology, University of Maiduguri Teaching Hospital, Maiduguri, Nigeria
| | - John David Ohieku
- Department of Clinical Pharmacy and Pharmacy Administration, Faculty of Pharmacy, University of Maiduguri, Maiduguri, Nigeria
| | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | | | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Adegbola AJ, Ndwiga L, Wamae K, Osoti V, Bolaji OO, Bejon P, Ochola-Oyier LI. ONT sequencing identifies a high prevalence of crt sensitive, triple mutant dhfr and single mutant dhps parasites within an ANC population in Nigeria. Front Genet 2024; 15:1470156. [PMID: 39483850 PMCID: PMC11525066 DOI: 10.3389/fgene.2024.1470156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Background Malaria in pregnancy is a major public health issue, particularly among vulnerable populations in malaria-endemic sub-Saharan African countries. To mitigate its risks, WHO recommends sulphadoxine-pyrimethamine (SP) for chemoprevention and artemisinin-based combination therapy (ACT) to treat uncomplicated Plasmodium falciparum malaria. These interventions have helped to alleviate the risk associated with malaria in pregnancy; however, in the context of the emergence of SP- and ACT-resistant P. falciparum, maintained efficacy is under threat. Molecular surveillance is a reliable tool to monitor the emergence of resistance where molecular markers are known. Thus, the objective of the study was to use a multiplexed amplicon Oxford Nanopore sequencing approach to assess the molecular markers for antimalarial resistance among pregnant women in Nigeria. Methods Dried blood spots (DBS) were collected from pregnant women who received IPTp-SP at the enrollment and follow-up visits. P. falciparum genomic DNA was extracted by the Chelex® method and Pf18S qPCR was used to detect parasite DNA in each sample. With nested PCR assays, fragments of Pfdhps, Pfdhfr, Pfmdr1, Pfcrt, Pfk13 and Pfama1 genes were amplified and multiplexed amplicon-based sequencing was conducted on the minION Oxford Nanopore Technology. Result In total, 251 pregnant women were enrolled in the study and 457 DBS samples were collected. P. falciparum genomic DNA was detected in 12% (56/457) of the samples, 31 at baseline and the remaining during the follow-up visits. Pfama1, pfk13, Pfdhps, Pfdhfr, Pfmdr1 and Pfcrt were successfully sequenced in a single run. Notably, k13 artemisinin resistance mutations were absent, the frequencies of Pfdhfr and Pfdhps SP resistance haplotypes, IRN for pyrimethamine resistance and ISGKA/IAGKA associated with sulphadoxine resistance were 82% (36/44) and 64% (27/42), respectively, and the Pfcrt CVIET resistant haplotype was at approximately 22% (7/32). Conclusion and recommendations Here a multiplexed amplicon-based ONT assay established that triple mutant Pfdfhr-IRN, double mutant Pfdhps-SG haplotypes and the chloroquine sensitive strain were prevalent among pregnant women in Nigeria.
Collapse
Affiliation(s)
- Adebanjo Jonathan Adegbola
- Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme (KWTRP), Kilifi, Kenya
| | - Leonard Ndwiga
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme (KWTRP), Kilifi, Kenya
| | - Kevin Wamae
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme (KWTRP), Kilifi, Kenya
| | - Victor Osoti
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme (KWTRP), Kilifi, Kenya
| | | | - Philip Bejon
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme (KWTRP), Kilifi, Kenya
| | | |
Collapse
|
11
|
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
|
12
|
Rogerson SJ, Aitken EH. Malaria in pregnancy: baby steps. Curr Opin Infect Dis 2024; 37:320-326. [PMID: 39018104 DOI: 10.1097/qco.0000000000001037] [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: 07/18/2024]
Abstract
PURPOSE OF REVIEW Malaria threatens pregnant women and their babies, particularly in Africa. RECENT FINDINGS This century, the number of women at risk of malaria in pregnancy has decreased globally, apart from in Africa, where it has increased. Low and sub microscopic infections are increasingly documented but remain hard to diagnose with current point-of-care tests, and their contribution to morbidity and transmission are unclear. Artemether-lumefantrine has been endorsed for treatment in first trimester, but many women attend antenatal clinics later in pregnancy, and reaching high-risk young, first-time mothers is particularly difficult. Small-for-gestational-age babies frequently result from malaria, which affects the placenta's development and its functions such as nutrient transport. Resistance to continues to increase to sulphadoxine-pyrimethamine, the mainstay of intermittent preventive treatment in pregnancy. The alternative, dihydroartemisinin-piperaquine controls malaria better, but does not improve pregnancy outcomes, suggesting that sulphadoxine-pyrimethamine may have nonmalarial effects including improving gut function or reducing dangerous inflammation. Understanding of how the malaria parasite uses the VAR2CSA protein to bind to its placental receptor is increasing, informing the search for a vaccine to prevent pregnancy malaria. SUMMARY Progress in several areas increases optimism that improved prevention and control of malaria in pregnancy is possible, but obstacles remain.
Collapse
Affiliation(s)
- Stephen J Rogerson
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne
- Department of Medicine (RMH), The University of Melbourne, 4 Floor, Clinical Sciences Building, Royal Melbourne Hospital, Royal Parade, Parkville
| | - Elizabeth H Aitken
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Cellich P, Unger HW, Rogerson SJ, Mola GDL. Impact on pregnancy outcomes of intermittent preventive treatment with sulfadoxine-pyrimethamine in urban and peri-urban Papua New Guinea: a retrospective cohort study. Malar J 2024; 23:201. [PMID: 38970076 PMCID: PMC11225125 DOI: 10.1186/s12936-024-05010-0] [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: 03/25/2024] [Accepted: 06/04/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Intermittent preventive treatment in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP) reduces malaria-attributable adverse pregnancy outcomes and may also prevent low birth weight (< 2,500 g) through mechanisms independent of malaria. Malaria transmission in Papua New Guinea (PNG) is highly heterogeneous. The impact of IPTp-SP on adverse birth outcomes in settings with little or no malaria transmission, such as PNG's capital city Port Moresby, is unknown. METHODS A retrospective cohort study was conducted amongst HIV-negative women with a singleton pregnancy who delivered at Port Moresby General Hospital between 18 July and 21 August 2022. The impact of IPTp-SP doses on adverse birth outcomes and anaemia was assessed using logistic and linear regression models, as appropriate. RESULTS Of 1,140 eligible women amongst 1,228 consecutive births, 1,110 had a live birth with a documented birth weight. A total of 156 women (13.7%) did not receive any IPTp-SP, 347 women (30.4%) received one, 333 (29.2%) received two, and 304 (26.7%) received the recommended ≥ 3 doses of IPTp-SP. A total of 65 of 1,110 liveborn babies (5.9%) had low birth weight and there were 34 perinatal deaths (3.0%). Anaemia (haemoglobin < 100 g/L) was observed in 30.6% (243/793) of women, and 14 (1.2%) had clinical malaria in pregnancy. Compared to women receiving 0-1 dose of IPTp-SP, women receiving ≥ 2 doses had lower odds of LBW (adjusted odds ratio [aOR] 0.50; 95% confidence interval [CI] 0.26, 0.96), preterm birth (aOR 0.58; 95% CI 0.32, 1.04), perinatal death (aOR 0.49; 95% CI 0.18, 1.38), LBW/perinatal death (aOR 0.55; 95% CI 0.27, 1.12), and anaemia (OR 0.50; 95% CI 0.36, 0.69). Women who received 2 doses versus 0-1 had 45% lower odds of LBW (aOR 0.55, 95% CI 0.27, 1.10), and a 16% further (total 61%) reduction with ≥ 3 doses (aOR 0.39, 95% CI 0.14, 1.05). Birth weights for women who received 2 or ≥ 3 doses versus 0-1 were 81 g (95% CI -3, 166) higher, and 151 g (58, 246) higher, respectively. CONCLUSIONS Provision of IPTp-SP in a low malaria-transmission setting in PNG appears to translate into substantial health benefits, in a dose-response manner, supporting the strengthening IPTp-SP uptake across all transmission settings in PNG.
Collapse
Affiliation(s)
- Philip Cellich
- Division of Obstetrics and Gynaecology, School of Medicine and Health Sciences, Port Moresby General Hospital, University of Papua New Guinea, Port Moresby, Papua New Guinea.
- Department of Obstetrics and Gynaecology, Canterbury Hospital, 575 Canterbury Road, Campsie 2194, NSW, Australia.
| | - Holger W Unger
- Menzies School of Health Research, Charles Darwin University, PO Box 41096, Casuarina 0810, NT, Australia.
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, NT, Australia.
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
- Department of Infectious Diseases, University of Melbourne, The Doherty Institute, Melbourne, VIC, Australia.
| | - 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
| | - Glen D L Mola
- Division of Obstetrics and Gynaecology, School of Medicine and Health Sciences, Port Moresby General Hospital, University of Papua New Guinea, Port Moresby, Papua New Guinea
- School of Medicine and Health Sciences, University of Papua New Guinea, Port Moresby, Papua New Guinea
| |
Collapse
|
16
|
Opoku KB, Tompkins K, Waltmann A, Ciccone EJ, Bartlelt L, Andermann T, Chinkhumba J, Mathanga DP, Gutman JR, Juliano JJ. Impact of Sulfadoxine-Pyrimethamine and Dihydroartemisinin-Piperaquine as Intermittent Preventive Treatment in Pregnancy on Stool Antimicrobial Resistance Gene Abundance. Am J Trop Med Hyg 2024; 111:43-47. [PMID: 38806022 PMCID: PMC11229632 DOI: 10.4269/ajtmh.23-0824] [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: 11/27/2023] [Accepted: 03/11/2024] [Indexed: 05/30/2024] Open
Abstract
Increasing antimicrobial resistance (AMR) is a global public health emergency. Although chemoprevention has improved malaria-related pregnancy outcomes, the downstream effects on AMR have not been characterized. We compared the abundance of 10 AMR genes in stool samples from pregnant women receiving sulfadoxine-pyrimethamine (SP) as intermittent preventive treatment against malaria in pregnancy (IPTp) to that in samples from women receiving dihydroartemisinin-piperaquine (DP) for IPTp. All participants had at least one AMR gene at baseline. Mean quantities of the antifolate gene dfrA17 were increased after two or more doses of SP (mean difference = 1.6, 95% CI: 0.4-2.7, P = 0.008). Antimicrobial resistance gene abundance tended to increase from baseline in SP recipients compared with a downward trend in the DP group. Overall, IPTp-SP had minimal effects on the abundance of antifolate resistance genes (except for dfrA17), potentially owing to a high starting prevalence. However, the trend toward increasing AMR in SP recipients warrants further studies.
Collapse
Affiliation(s)
- Kofi B. Opoku
- Applied Epidemiology Program, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen Tompkins
- Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Andreea Waltmann
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Emily J. Ciccone
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Luther Bartlelt
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Tessa Andermann
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | | | - Don P. Mathanga
- Malaria Alert Center, Malawi College of Medicine, Blantyre, Malawi
| | - Julie R. Gutman
- Division of Parasitic Diseases and Malaria, National Center for Emerging and Zoonotic Infectious Diseases, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jonathan J. Juliano
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
- Division of Infectious Diseases, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
17
|
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
|
18
|
Cheng K, Aitken EH, Hasang W, Meagher N, Price DJ, Madanitsa M, Mwapasa V, Phiri KS, Dodd J, ter Kuile FO, Rogerson SJ. Intermittent preventive treatment with sulphadoxine-pyrimethamine but not dihydroartemisinin-piperaquine modulates the relationship between inflammatory markers and adverse pregnancy outcomes in Malawi. PLOS GLOBAL PUBLIC HEALTH 2024; 4:e0003198. [PMID: 38753813 PMCID: PMC11098340 DOI: 10.1371/journal.pgph.0003198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
Women in malaria-endemic areas receive sulphadoxine-pyrimethamine (SP) as Intermittent Preventive Treatment in Pregnancy (IPTp) to reduce malaria. While dihydroartemisinin-piperaquine (DP) has superior antimalarial properties as IPTp, SP is associated with superior fetal growth. As maternal inflammation influences fetal growth, we investigated whether SP alters the relationship between inflammation and birth outcomes. We measured C-reactive protein (CRP) and alpha-1-acid glycoprotein (AGP) at enrollment (16-28 gestation weeks (gw)), visit 3 (24-36 gw) and delivery in 1319 Malawian women randomized to receive monthly SP, DP, or DP and single-dose azithromycin (AZ) in the IMPROVE trial (NCT03208179). Logistic regression was used to assess the relationship between adverse outcomes, inflammation, and treatment arm. Elevated AGP at enrollment was associated with adverse birth outcome (aRR 1.40, 95% CI: 1.15, 1.70), with similar associations observed across treatment arms, exceptions being that elevated AGP was associated with low maternal weight gain in SP recipients (aRR 1.94, 95% CI: 1.36, 2.76) and with small for gestational age in DP+AZ recepients (aRR 1.49, 95% CI 1.02, 2.17). At visit 3 there were few associations between inflammation andoutcomes. At delivery, women with elevated AGP receiving either DP or DP+AZ had an increased risk of adverse birth outcomes (aRR 1.60, 95% CI: 1.28, 2.00), including low birth weight, pre-term birth and foetal loss, this was not seen in women receiving SP (aRR 0.82, 95% CI: 0.54, 1.26). The risk of an association between elevated AGP and adverse birth outcome was higher in those receiving DP or DP+AZ compared to those receiving SP (aRR 1.95, 95% CI: 1.21, 3.13). No clear associations between CRP and adverse outcomes were observed. AGP identified women at risk of adverse pregnancy outcomes. SP modifies the relationship between inflammatory biomarkers and adverse outcomes. Our findings provide insights into potential mechanisms by which SP may improve pregnancy outcomes.
Collapse
Affiliation(s)
- Kaylene Cheng
- Department of Medicine (RMH), The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth H. Aitken
- Department of Infectious Diseases, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Wina Hasang
- Department of Infectious Diseases, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Niamh Meagher
- Department of Infectious Diseases, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - David J. Price
- Department of Infectious Diseases, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Epidemiology & Biostatistics, Melbourne School of Population & Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Mwayiwawo Madanitsa
- Department of Clinical Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Victor Mwapasa
- Department of Epidemiology and Biostatistics, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Kamija S. Phiri
- Department of Epidemiology and Biostatistics, School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
- Training and Research Unit of Excellence, Blantyre, Malawi
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Feiko O. ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Stephen J. Rogerson
- Department of Medicine (RMH), The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The Peter Doherty Institute of Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
19
|
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
|
20
|
Riddell MA, Vallely LM, Mengi A, Badman SG, Low N, Wand H, Bolnga JW, Babona D, Mola GDL, Wiseman V, Kelly-Hanku A, Homer CSE, Morgan C, Luchters S, Whiley DM, Robinson LJ, Au L, Pukai-Gani I, Laman M, Kariwiga G, Toliman PJ, Batura N, Tabrizi SN, Rogerson SJ, Garland SM, Guy RJ, Peeling RW, Pomat WS, Kaldor JM, Vallely AJB. Point-of-care testing and treatment of sexually transmitted and genital infections to improve birth outcomes in high-burden, low-resource settings (WANTAIM): a pragmatic cluster randomised crossover trial in Papua New Guinea. Lancet Glob Health 2024; 12:e641-e651. [PMID: 38485431 DOI: 10.1016/s2214-109x(24)00004-4] [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: 11/19/2023] [Revised: 12/13/2023] [Accepted: 01/02/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis, and bacterial vaginosis have been associated with adverse maternal and perinatal outcomes, but there is conflicting evidence on the benefits of antenatal screening and treatment for these conditions. We aimed to determine the effect of antenatal point-of-care testing and immediate treatment of C trachomatis, N gonorrhoeae, T vaginalis, and bacterial vaginosis on preterm birth, low birthweight, and other adverse maternal and perinatal outcomes compared with current standard of care, which included symptom-based treatment without laboratory confirmation. METHODS In this pragmatic cluster randomised crossover trial, we enrolled women (aged ≥16 years) attending an antenatal clinic at 26 weeks' gestation or earlier (confirmed by obstetric ultrasound), living within approximately 1 h drive of a study clinic, and able to provide reliable contact details at ten primary health facilities and their catchment communities (clusters) in Papua New Guinea. Clusters were randomly allocated 1:1 to receive either the intervention or control (standard care) in the first phase of the trial. Following an interval (washout period) of 2-3 months at the end of the first phase, each cluster crossed over to the other group. Randomisation was stratified by province. Individual participants were informed about trial group allocation only after completing informed consent procedures. The primary outcome was a composite of preterm birth (livebirth before 37 weeks' gestation), low birthweight (<2500 g), or both, analysed according to the intention-to-treat population. This study is registered with ISRCTN Registry, ISRCTN37134032, and is completed. FINDINGS Between July 26, 2017, and Aug 30, 2021, 4526 women were enrolled (2210 [63·3%] of 3492 women in the intervention group and 2316 [62·8%] of 3687 in the control group). Primary outcome data were available for 4297 (94·9%) newborn babies of 4526 women. The proportion of preterm birth, low birthweight, or both, in the intervention group, expressed as the mean of crude proportions across clusters, was 18·8% (SD 4·7%) compared with 17·8% in the control group (risk ratio [RR] 1·06, 95% CI 0·78-1·42; p=0·67). There were 1052 serious adverse events reported (566 in the intervention group and 486 in the control group) among 929 trial participants, and no differences by trial group. INTERPRETATION Point-of-care testing and treatment of C trachomatis, N gonorrhoeae, T vaginalis, and bacterial vaginosis did not reduce preterm birth or low birthweight compared with standard care. Within the subgroup of women with N gonorrhoeae, there was a substantial reduction in the primary outcome. FUNDING UK Department of Health and Social Care; UK Foreign, Commonwealth and Development Office; UK Medical Research Council; the Wellcome Trust; the Australian National Health and Medical Research Council; and Swiss National Science Foundation.
Collapse
Affiliation(s)
- Michaela A Riddell
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - Lisa M Vallely
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - Alice Mengi
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Steven G Badman
- The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Handan Wand
- The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - John W Bolnga
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; Modilon General Hospital, Madang, Papua New Guinea
| | - Delly Babona
- St Mary's Hospital Vunapope, Kokopo, Papua New Guinea
| | - Glen D L Mola
- School of Medicine and Health Sciences, University of Papua New Guinea, National Capital District, Papua New Guinea
| | - Virginia Wiseman
- The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia; London School of Hygiene & Tropical Medicine, London, UK
| | - Angela Kelly-Hanku
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | | | - Christopher Morgan
- Burnet Institute, Melbourne, VIC, Australia; Jhpiego the Johns Hopkins University affiliate, Baltimore, MD, USA
| | - Stanley Luchters
- Centre for Sexual Health and HIV/AIDS Research, Harare, Zimbabwe; Liverpool School of Tropical Medicine, Liverpool, UK
| | - David M Whiley
- UQ Centre for Clinical Research, University of Queensland, Herston, QLD, Australia; Pathology Queensland Central Laboratory, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Leanne J Robinson
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; Burnet Institute, Melbourne, VIC, Australia
| | - Lucy Au
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Irene Pukai-Gani
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Moses Laman
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Grace Kariwiga
- Alotau Provincial Hospital, Alotau, Milne Bay Province, Papua New Guinea
| | - Pamela J Toliman
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - Neha Batura
- University College London Institute for Global Health, London, UK
| | - Sepehr N Tabrizi
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia; Centre for Women's Infectious Diseases Research, The Royal Women's Hospital Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Infectious Diseases, University of Melbourne, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Suzanne M Garland
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia; Centre for Women's Infectious Diseases Research, The Royal Women's Hospital Melbourne, VIC, Australia
| | - Rebecca J Guy
- The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | | | - William S Pomat
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - John M Kaldor
- The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia
| | - Andrew J B Vallely
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea; The Kirby Institute, University of New South Wales Sydney, Kensington, NSW, Australia.
| |
Collapse
|
21
|
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
|
22
|
Barsosio HC, Madanitsa M, Ondieki ED, Dodd J, Onyango ED, Otieno K, Wang D, Hill J, Mwapasa V, Phiri KS, Maleta K, Taegtmeyer M, Kariuki S, Schmiegelow C, Gutman JR, Ter Kuile FO. Chemoprevention for malaria with monthly intermittent preventive treatment with dihydroartemisinin-piperaquine in pregnant women living with HIV on daily co-trimoxazole in Kenya and Malawi: a randomised, double-blind, placebo-controlled trial. Lancet 2024; 403:365-378. [PMID: 38224710 PMCID: PMC10865779 DOI: 10.1016/s0140-6736(23)02631-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/08/2023] [Accepted: 11/21/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND The efficacy of daily co-trimoxazole, an antifolate used for malaria chemoprevention in pregnant women living with HIV, is threatened by cross-resistance of Plasmodium falciparum to the antifolate sulfadoxine-pyrimethamine. We assessed whether addition of monthly dihydroartemisinin-piperaquine to daily co-trimoxazole is more effective at preventing malaria infection than monthly placebo plus daily co-trimoxazole in pregnant women living with HIV. METHODS We did an individually randomised, two-arm, placebo-controlled trial in areas with high-grade sulfadoxine-pyrimethamine resistance in Kenya and Malawi. Pregnant women living with HIV on dolutegravir-based combination antiretroviral therapy (cART) who had singleton pregnancies between 16 weeks' and 28 weeks' gestation were randomly assigned (1:1) by computer-generated block randomisation, stratified by site and HIV status (known positive vs newly diagnosed), to daily co-trimoxazole plus monthly dihydroartemisinin-piperaquine (three tablets of 40 mg dihydroartemisinin and 320 mg piperaquine given daily for 3 days) or daily co-trimoxazole plus monthly placebo. Daily co-trimoxazole consisted of one tablet of 160 mg sulfamethoxazole and 800 mg trimethoprim. The primary endpoint was the incidence of Plasmodium infection detected in the peripheral (maternal) or placental (maternal) blood or tissue by PCR, microscopy, rapid diagnostic test, or placental histology (active infection) from 2 weeks after the first dose of dihydroartemisinin-piperaquine or placebo to delivery. Log-binomial regression was used for binary outcomes, and Poisson regression for count outcomes. The primary analysis was by modified intention to treat, consisting of all randomised eligible participants with primary endpoint data. The safety analysis included all women who received at least one dose of study drug. All investigators, laboratory staff, data analysts, and participants were masked to treatment assignment. This trial is registered with ClinicalTrials.gov, NCT04158713. FINDINGS From Nov 11, 2019, to Aug 3, 2021, 904 women were enrolled and randomly assigned to co-trimoxazole plus dihydroartemisinin-piperaquine (n=448) or co-trimoxazole plus placebo (n=456), of whom 895 (99%) contributed to the primary analysis (co-trimoxazole plus dihydroartemisinin-piperaquine, n=443; co-trimoxazole plus placebo, n=452). The cumulative risk of any malaria infection during pregnancy or delivery was lower in the co-trimoxazole plus dihydroartemisinin-piperaquine group than in the co-trimoxazole plus placebo group (31 [7%] of 443 women vs 70 [15%] of 452 women, risk ratio 0·45, 95% CI 0·30-0·67; p=0·0001). The incidence of any malaria infection during pregnancy or delivery was 25·4 per 100 person-years in the co-trimoxazole plus dihydroartemisinin-piperaquine group versus 77·3 per 100 person-years in the co-trimoxazole plus placebo group (incidence rate ratio 0·32, 95% CI 0·22-0·47, p<0·0001). The number needed to treat to avert one malaria infection per pregnancy was 7 (95% CI 5-10). The incidence of serious adverse events was similar between groups in mothers (17·7 per 100 person-years in the co-trimoxazole plus dihydroartemisinin-piperaquine group [23 events] vs 17·8 per 100 person-years in the co-trimoxazole group [25 events]) and infants (45·4 per 100 person-years [23 events] vs 40·2 per 100 person-years [21 events]). Nausea within the first 4 days after the start of treatment was reported by 29 (7%) of 446 women in the co-trimoxazole plus dihydroartemisinin-piperaquine group versus 12 (3%) of 445 women in the co-trimoxazole plus placebo group. The risk of adverse pregnancy outcomes did not differ between groups. INTERPRETATION Addition of monthly intermittent preventive treatment with dihydroartemisinin-piperaquine to the standard of care with daily unsupervised co-trimoxazole in areas of high antifolate resistance substantially improves malaria chemoprevention in pregnant women living with HIV on dolutegravir-based cART and should be considered for policy. FUNDING European and Developing Countries Clinical Trials Partnership 2; UK Joint Global Health Trials Scheme (UK Foreign, Commonwealth and Development Office; Medical Research Council; National Institute for Health Research; Wellcome); and Swedish International Development Cooperation Agency.
Collapse
Affiliation(s)
- Hellen C Barsosio
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Mwayiwawo Madanitsa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi; Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | - Everlyne D Ondieki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - James Dodd
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Eric D Onyango
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Duolao Wang
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Jenny Hill
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Victor Mwapasa
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Kamija S Phiri
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Kenneth Maleta
- School of Global and Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | - Miriam Taegtmeyer
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Christentze Schmiegelow
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark; Department of Gynaecology and Obstetrics, Copenhagen University Hospital - North Zealand, Hillerød, Denmark
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Feiko O Ter Kuile
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| |
Collapse
|
23
|
Unger HW, Acharya S, Arnold L, Wu C, van Eijk AM, Gore-Langton GR, Ter Kuile FO, Lufele E, Chico RM, Price RN, Moore BR, Thriemer K, Rogerson SJ. The effect and control of malaria in pregnancy and lactating women in the Asia-Pacific region. Lancet Glob Health 2023; 11:e1805-e1818. [PMID: 37858590 DOI: 10.1016/s2214-109x(23)00415-1] [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: 05/21/2023] [Revised: 08/10/2023] [Accepted: 08/23/2023] [Indexed: 10/21/2023]
Abstract
Half of all pregnancies at risk of malaria worldwide occur in the Asia-Pacific region, where Plasmodium falciparum and Plasmodium vivax co-exist. Despite substantial reductions in transmission, malaria remains an important cause of adverse health outcomes for mothers and offspring, including pre-eclampsia. Malaria transmission is heterogeneous, and infections are commonly subpatent and asymptomatic. High-grade antimalarial resistance poses a formidable challenge to malaria control in pregnancy in the region. Intermittent preventive treatment in pregnancy reduces infection risk in meso-endemic New Guinea, whereas screen-and-treat strategies will require more sensitive point-of-care tests to control malaria in pregnancy. In the first trimester, artemether-lumefantrine is approved, and safety data are accumulating for other artemisinin-based combinations. Safety of novel antimalarials to treat artemisinin-resistant P falciparum during pregnancy, and of 8-aminoquinolines during lactation, needs to be established. A more systematic approach to the prevention of malaria in pregnancy in the Asia-Pacific is required.
Collapse
Affiliation(s)
- Holger W Unger
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Tiwi, NT, Australia; Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK.
| | - Sanjaya Acharya
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Lachlan Arnold
- Royal Melbourne Hospital Clinical School, The University of Melbourne, Parkville, VIC, Australia
| | - Connie Wu
- Royal Melbourne Hospital Clinical School, The University of Melbourne, Parkville, VIC, Australia
| | - Anna Maria van Eijk
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Georgia R Gore-Langton
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - 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, Papua New Guinea
| | - R Matthew Chico
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia; Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Brioni R Moore
- Curtin Medical School, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia; Telethon Kids Institute, Perth Children's Hospital, Nedlands, WA, Australia
| | - Kamala Thriemer
- Global and Tropical Health Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - 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
| |
Collapse
|
24
|
Alruwaili M, Uwimana A, Sethi R, Murindahabi M, Piercefield E, Umulisa N, Abram A, Eckert E, Munguti K, Mbituyumuremyi A, Gutman JR, Sullivan DJ. Peripheral and Placental Prevalence of Sulfadoxine-Pyrimethamine Resistance Markers in Plasmodium falciparum among Pregnant Women in Southern Province, Rwanda. Am J Trop Med Hyg 2023; 109:1057-1062. [PMID: 37783456 PMCID: PMC10622487 DOI: 10.4269/ajtmh.23-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/22/2023] [Indexed: 10/04/2023] Open
Abstract
Intermittent preventive therapy during pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is recommended in areas of moderate to high malaria transmission intensity. As a result of the increasing prevalence of SP resistance markers, IPTp-SP was withdrawn from Rwanda in 2008. Nonetheless, more recent findings suggest that SP may improve birthweight even in the face of parasite resistance, through alternative mechanisms that are independent of antimalarial effects. The prevalence of single nucleotide polymorphisms in Plasmodium falciparum dihydropteroate synthase (pfdhps) and dihydrofolate reductase (pfdhfr) genes associated with SP resistance among 148 pregnant women from 2016 to 2018 within Rwanda's Southern Province (Huye and Kamonyi districts) was measured using a ligase detection reaction-fluorescent microsphere assay. The frequency of pfdhps K540E, A581G, and the quintuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E) and sextuple (pfdhfr N51I + C59R + S108N/pfdhps A437G + K540E + A581G) mutant genotypes was 90%, 38%, 75%, and 28%, respectively. No significant genotype difference was seen between the two districts, which are approximately 50 km apart. Observed agreements for matched peripheral to placental blood were reported and found to be 207 of 208 (99%) for pfdhfr and 239 of 260 (92%) for pfdhps. The peripheral blood sample did not miss any pfdhfr drug-resistant mutants or pfdhps except at the S436 loci. At this level of the sextuple mutant, the antimalarial efficacy of SP for preventing low birthweight is reduced, although overall SP still exerts a nonmalarial benefit during pregnancy. This study further reveals the need to intensify preventive measures to sustain malaria control in Rwanda to keep the overall incidence of malaria during pregnancy low.
Collapse
Affiliation(s)
- Muharib Alruwaili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Aline Uwimana
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
| | - Reena Sethi
- Maternal and Child Survival Program/Jhpiego, Washington, District of Columbia
| | - Monique Murindahabi
- Malaria and Other Parasitic Diseases Division, Rwanda Biomedical Center, Kigali, Rwanda
- Roll Back Malaria, Ouagadougou, Burkina Faso
| | - Emily Piercefield
- U.S. President’s Malaria Initiative, Malaria Branch, U.S. Centers for Disease Control and Prevention, Kigali, Rwanda
| | - Noella Umulisa
- Maternal and Child Survival Program/Jhpiego, Kigali, Rwanda
| | | | - Erin Eckert
- RTI International, Washington, District of Columbia
| | - Kaendi Munguti
- U.S. President’s Malaria Initiative, U.S. Agency for International Development, Kigali, Rwanda
| | | | - Julie R. Gutman
- Malaria Branch, U.S. Centers for Disease Control and Prevention, Atlanta, Georgia
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
25
|
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
|
26
|
Sorano S, Gore-Langton G, Opondo C, Smith C, Matsui M, Chaponda EB, Chandramohan D, Chico RM. Coinfections of malaria and sexually transmitted and reproductive tract infections in pregnancy in sub-Saharan Africa: a systematic review and individual participant data meta-analysis protocol. BMJ Open 2023; 13:e074896. [PMID: 37339835 DOI: 10.1136/bmjopen-2023-074896] [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: 06/22/2023] Open
Abstract
INTRODUCTION Malaria infection and curable sexually transmitted infections and reproductive tract infections (STIs/RTIs) adversely impact pregnancy outcomes. In sub-Saharan Africa, the prevalence of malaria and curable STIs/RTIs is high and, where coinfection is common, combination interventions may be needed to improve pregnancy outcomes. The aim of this systematic review is to estimate the prevalence of malaria and curable STI/RTI coinfection during pregnancy, risk factors for coinfection and prevalence of associated adverse pregnancy outcomes. METHODS AND ANALYSIS We will use three electronic databases, PubMed, EMBASE and Malaria in Pregnancy Library to identify studies involving pregnant women attending routine antenatal care facilities in sub-Saharan Africa and reporting malaria and curable STI/RTI test results, published in any language since 2000. We will search databases in the second quarter of 2023 and repeat the search before completion of our analyses. The first two authors will screen titles and abstracts, selecting studies that meet inclusion criteria and qualify for full-text screening. If agreement on inclusion/exclusion cannot be reached, the last author will serve as arbiter. We will extract data from eligible publications for a study-level meta-analysis. We will contact research groups of included studies and request individual participant data for meta-analysis. The first two authors will conduct a quality appraisal of included studies using the GRADE system. The last author will adjudicate if the first two authors do not agree on any appraisals. We will conduct sensitivity analyses to test the robustness of effect estimates over time (by decade and half-decade periods), geography (East/Southern Africa vs West/Central Africa), gravidity (primigravidae, secundigravidae, multigravidae), treatment type and dosing frequency, and malaria transmission intensity. ETHICS AND DISSEMINATION We obtained ethics approval from the London School of Hygiene & Tropical Medicine (LSHTM Ethics Ref: 26167). Results of this study will be disseminated via peer-reviewed publication and presentation at scientific conferences. PROSPERO REGISTRATION NUMBER CRD42021224294.
Collapse
Affiliation(s)
- Sumire Sorano
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of Disease Control, London School of Hygiene & Tropical Medicine Faculty of Infectious and Tropical Diseases, London, UK
| | - Georgia Gore-Langton
- Department of Disease Control, London School of Hygiene & Tropical Medicine Faculty of Infectious and Tropical Diseases, London, UK
| | - Charles Opondo
- Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine Faculty of Epidemiology and Population Health, London, UK
| | - Chris Smith
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Clinical Research Department, London School of Hygiene & Tropical Medicine Faculty of Infectious and Tropical Diseases, London, UK
| | - Mitsuaki Matsui
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
- Department of Public Health, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | | | - Daniel Chandramohan
- Disease Control, London School of Hygiene & Tropical Medicine Faculty of Infectious and Tropical Diseases, London, UK
| | - R Matthew Chico
- Department of Disease Control, London School of Hygiene & Tropical Medicine Faculty of Infectious and Tropical Diseases, London, UK
| |
Collapse
|
27
|
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
|
28
|
Weckman AM, Elphinstone RE, Ssenkusu JM, Tran V, Zhong K, Madanitsa M, Khairallah C, Kalilani-Phiri L, Mwapasa V, Conroy AL, Ter Kuile FO, McDonald CR, Kain KC. Sequential disruptions to inflammatory and angiogenic pathways and risk of spontaneous preterm birth in Malawian women. iScience 2023; 26:106912. [PMID: 37332611 PMCID: PMC10275952 DOI: 10.1016/j.isci.2023.106912] [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: 06/08/2022] [Revised: 11/11/2022] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
Preterm birth is a leading cause of death in children under five years of age. We hypothesized that sequential disruptions to inflammatory and angiogenic pathways during pregnancy increase the risk of placental insufficiency and spontaneous preterm labor and delivery. We conducted a secondary analysis of inflammatory and angiogenic analytes measured in plasma samples collected across pregnancy from 1462 Malawian women. Women with concentrations of the inflammatory markers sTNFR2, CHI3L1, and IL18BP in the highest quartile before 24 weeks gestation and women with anti-angiogenic factors sEndoglin and sFlt-1/PlGF ratio in the highest quartile at 28-33 weeks gestation had an increased relative risk of preterm birth. Mediation analysis further supported a potential causal link between early inflammation, subsequent angiogenic dysregulation detrimental to placental vascular development, and earlier gestational age at delivery. Interventions designed to reduce the burden of preterm birth may need to be implemented before 24 weeks of gestation.
Collapse
Affiliation(s)
- Andrea M. Weckman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Robyn E. Elphinstone
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - John M. Ssenkusu
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University, Kampala, Uganda
| | - Vanessa Tran
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Kathleen Zhong
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | | | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | | | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Andrea L. Conroy
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Feiko O. Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Chloe R. McDonald
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| | - Kevin C. Kain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, ON, Canada
- Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada
- Toronto General Research Institute, University Health Network-Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
29
|
Muthiani Y, Hunter PJ, Näsänen-Gilmore PK, Koivu AM, Isojärvi J, Luoma J, Salenius M, Hadji M, Ashorn U, Ashorn P. Antenatal interventions to reduce risk of low birth weight related to maternal infections during pregnancy. Am J Clin Nutr 2023; 117 Suppl 2:S118-S133. [PMID: 37331759 DOI: 10.1016/j.ajcnut.2023.02.025] [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: 11/13/2022] [Revised: 01/27/2023] [Accepted: 02/09/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND Maternal infections during pregnancy have been linked to increased risk of adverse birth outcomes, including low birth weight (LBW), preterm birth (PTB), small for gestational age (SGA), and stillbirth (SB). OBJECTIVES The purpose of this article was to summarize evidence from published literature on the effect of key interventions targeting maternal infections on adverse birth outcomes. METHODS We searched MEDLINE, Embase, Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, and CINAHL Complete between March 2020 and May 2020 with an update to cover until August 2022. We included randomized controlled trials (RCTs) and reviews of RCTs of 15 antenatal interventions for pregnant women reporting LBW, PTB, SGA, or SB as outcomes. RESULTS Of the 15 reviewed interventions, the administration of 3 or more doses of intermittent preventive treatment in pregnancy with sulphadoxine-pyrimethamine [IPTp-SP; RR: 0.80 (95% CI: 0.69, 0.94)] can reduce risk of LBW compared with 2 doses. The provision of insecticide-treated bed nets, periodontal treatment, and screening and treatment of asymptomatic bacteriuria may reduce risk of LBW. Maternal viral influenza vaccination, treatment of bacterial vaginosis, intermittent preventive treatment with dihydroartemisinin-piperaquine compared with IPTp-SP, and intermittent screening and treatment of malaria during pregnancy compared with IPTp were deemed unlikely to reduce the prevalence of adverse birth outcomes. CONCLUSIONS At present, there is limited evidence from RCTs available for some potentially relevant interventions targeting maternal infections, which could be prioritized for future research.
Collapse
Affiliation(s)
- Yvonne Muthiani
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Patricia J Hunter
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Pieta K Näsänen-Gilmore
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Public Health and Welfare, Finnish Institute for Health and Welfare, FI-00271, Helsinki, Finland
| | - Annariina M Koivu
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jaana Isojärvi
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Juho Luoma
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Meeri Salenius
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Maryam Hadji
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ulla Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
30
|
Kakuru A, Jagannathan P. Can we reduce malaria in pregnancy and improve birth outcomes? Lancet 2023; 401:973-975. [PMID: 36913960 DOI: 10.1016/s0140-6736(23)00101-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/13/2022] [Indexed: 03/12/2023]
Affiliation(s)
- Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda; Department of Community and Public Health, Busitema University, Tororo, Uganda.
| | | |
Collapse
|
31
|
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
|
32
|
Bucher S, Nowak K, Otieno K, Tenge C, Marete I, Rutto F, Kemboi M, Achieng E, Ekhaguere OA, Nyongesa P, Esamai FO, Liechty EA. Birth weight and gestational age distributions in a rural Kenyan population. BMC Pediatr 2023; 23:112. [PMID: 36890485 PMCID: PMC9993805 DOI: 10.1186/s12887-023-03925-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 02/21/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND With the increased availability of access to prenatal ultrasound in low/middle-income countries, there is opportunity to better characterize the association between fetal growth and birth weight across global settings. This is important, as fetal growth curves and birthweight charts are often used as proxy health indicators. As part of a randomized control trial, in which ultrasonography was utilized to establish accurate gestational age of pregnancies, we explored the association between gestational age and birthweight among a cohort in Western Kenya, then compared our results to data reported by the INTERGROWTH-21st study. METHODS This study was conducted in 8 geographical clusters across 3 counties in Western Kenya. Eligible subjects were nulliparous women carrying singleton pregnancies. An early ultrasound was performed between 6 + 0/7 and 13 + 6/7 weeks gestational age. At birth, infants were weighed on platform scales provided either by the study team (community births), or the Government of Kenya (public health facilities). The 10th, 25th, median, 75th, and 90th BW percentiles for 36 to 42 weeks gestation were determined; resulting percentile points were plotted, and curves determined using a cubic spline technique. A signed rank test was used to quantify the comparison of the percentiles generated in the rural Kenyan sample with those of the INTERGROWTH-21st study. RESULTS A total of 1291 infants (of 1408 pregnant women randomized) were included. Ninety-three infants did not have a measured birth weight. The majority of these were due to miscarriage (n = 49) or stillbirth (n = 27). No significant differences were found between subjects who were lost to follow-up. Signed rank comparisons of the observed median of the Western Kenya data at 10th, 50th, and 90th birthweight percentiles, as compared to medians reported in the INTERGROWTH-21st distributions, revealed close alignment between the two datasets, with significant differences at 36 and 37 weeks. Limitations of the current study include small sample size, and detection of potential digit preference bias. CONCLUSIONS A comparison of birthweight percentiles by gestational age estimation, among a sample of infants from rural Kenya, revealed slight differences as compared to those from the global population (INTERGROWTH-21st). TRIAL REGISTRATION This is a single site sub-study of data collected in conjunction with the Aspirin Supplementation for Pregnancy Indicated Risk Reduction In Nulliparas (ASPIRIN) Trial, which is listed at ClinicalTrials.gov , NCT02409680 (07/04/2015).
Collapse
Affiliation(s)
- Sherri Bucher
- School of Medicine, Indiana University, Indianapolis, IN, USA.
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Indianapolis, IN, USA.
| | - Kayla Nowak
- RTI International, 3040 E. Cornwallis Road, Research Triangle Park, Durham, NC, USA
| | - Kevin Otieno
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Constance Tenge
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Irene Marete
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Faith Rutto
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Millsort Kemboi
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Emmah Achieng
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | | | - Paul Nyongesa
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | - Fabian O Esamai
- Department of Child Health and Paediatrics, Moi University School of Medicine, Eldoret, Kenya
| | | |
Collapse
|
33
|
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
|
34
|
Okoro RN, Geidam AD, Bukar AA, Zarami AB, Ohieku JD, Musa AB, Yerima TS. Superiority trial of intermittent treatment with dihydroartemisinin–piperaquine versus sulfadoxine–pyrimethamine for the prevention of malaria during pregnancy. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-023-00460-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Abstract
Background
Malaria in pregnancy is responsible for various adverse maternal and birth outcomes. The emerging resistance to sulfadoxine–pyrimethamine (SP) raises important concerns about its use for intermittent preventive treatment in pregnancy (IPTp) in Africa. This trial aimed to assess the efficacy and safety of IPTp with dihydroartemisinin–piperaquine (DP) as an alternative to IPTp with SP.
Results
The double-blind, randomized, and controlled superiority trial was conducted between July 2020 and June 2021. A total of 250 women were enrolled and randomly assigned to receive SP (n = 125) or DP (n = 125). Two hundred and six (82.4%) participants that contributed to the outcomes were included in the modified intention-to-treat (ITT) analysis, while 84 participants that completed the three courses of the study drugs were included in the per protocol (PP) analysis. The ITT analysis results showed that the incidence of histopathologically confirmed placental malaria was nonsignificantly higher in the DP group compared with the SP group (62.5% vs. 51.1%, P = 0.098). After adjusting for confounders, the risk of histopathologically confirmed placental malaria was also nonsignificantly higher in the DP group (Adjusted Relative Risk [RR] = 1.27, 95% CI 0.94–1.71) compared with the SP group. In contrast, the risk of a low APGAR score was significantly lower in the DP group (RR = 0.45, 95% CI 0.38–0.52) compared with the SP group. Also, the risk of a composite adverse birth outcome (low birth weight or preterm delivery or neonates small for the gestational age) was nonsignificantly lower in the DP group (Adjusted RR = 0.82, 95% CI 0.55–1.21) compared with the SP group. Both drugs were well tolerated, although nausea and vomiting occurred in a significant number of participants in the SP group.
Conclusions
A three-course IPTp with DP was safe and was not found to be superior to IPTp with SP in the prevention of placental malaria. Although IPTp with DP was associated with a significant lower risk of low APGAR score and nonsignificant lower risks of other adverse birth outcomes compared with IPTp with SP.
Trial registration
PACTR, PACTR202002644579177. Registered 20 February 2020, https://pactr.samrc.ac.za/TrialDisplay.aspx?TrialID=9753.
Collapse
|
35
|
Briggs J, Murray M, Nideffer J, Jagannathan P. Sex-Linked Differences in Malaria Risk Across the Lifespan. Curr Top Microbiol Immunol 2023; 441:185-208. [PMID: 37695429 DOI: 10.1007/978-3-031-35139-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Despite the high burden of malaria worldwide, there is surprisingly scarce research on sex-based differences in malaria outside of pregnancy. A more thorough understanding of sexual dimorphism in malaria, and what underlies these sex-based differences, could elucidate the underlying mechanisms driving malaria pathogenesis and has the potential to inform malaria control efforts, including new vaccines. This review summarizes our current understanding of sex-based differences in the epidemiology of malaria across the lifespan, potential sex- or gender-based mechanisms driving these differences, and the knowledge gaps that need to be addressed.
Collapse
Affiliation(s)
- Jessica Briggs
- Department of Medicine, University of California, San Francisco, California, United States
| | - Margaret Murray
- Department of Medicine, University of California, San Francisco, California, United States
| | - Jason Nideffer
- Department of Medicine, Stanford University, Stanford, California, United States
| | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, California, United States.
| |
Collapse
|
36
|
Gore-Langton GR, Cano J, Simpson H, Tatem A, Tejedor-Garavito N, Wigley A, Carioli A, Gething P, Weiss DJ, Chandramohan D, Walker PGT, Cairns ME, Chico RM. Global estimates of pregnancies at risk of Plasmodium falciparum and Plasmodium vivax infection in 2020 and changes in risk patterns since 2000. PLOS GLOBAL PUBLIC HEALTH 2022; 2:e0001061. [PMID: 36962612 PMCID: PMC10022219 DOI: 10.1371/journal.pgph.0001061] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 10/03/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Women are at risk of severe adverse pregnancy outcomes attributable to Plasmodium spp. infection in malaria-endemic areas. Malaria control efforts since 2000 have aimed to reduce this burden of disease. METHODS We used data from the Malaria Atlas Project and WorldPop to calculate global pregnancies at-risk of Plasmodium spp. infection. We categorised pregnancies as occurring in areas of stable and unstable P. falciparum and P. vivax transmission. We further stratified stable endemicity as hypo-endemic, meso-endemic, hyper-endemic, or holo-endemic, and estimated pregnancies at risk in 2000, 2005, 2010, 2015, 2017, and 2020. FINDINGS In 2020, globally 120.4M pregnancies were at risk of P. falciparum, two-thirds (81.0M, 67.3%) were in areas of stable transmission; 85 2M pregnancies were at risk of P. vivax, 93.9% (80.0M) were in areas of stable transmission. An estimated 64.6M pregnancies were in areas with both P. falciparum and P. vivax transmission. The number of pregnancies at risk of each of P. falciparum and P. vivax worldwide decreased between 2000 and 2020, with the exception of sub-Saharan Africa, where the total number of pregnancies at risk of P. falciparum increased from 37 3M in 2000 to 52 4M in 2020. INTERPRETATION Historic investments in malaria control have reduced the number of women at risk of malaria in pregnancy in all endemic regions except sub-Saharan Africa. Population growth in Africa has outpaced reductions in malaria prevalence. Interventions that reduce the risk of malaria in pregnancy are needed as much today as ever.
Collapse
Affiliation(s)
- Georgia R. Gore-Langton
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jorge Cano
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Expanded Special Project for Elimination of Neglected Tropical Diseases, WHO Regional Office for Africa, Brazzaville, Democratic Republic of the Congo
| | - Hope Simpson
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Andrew Tatem
- WorldPop, Geography and Environmental Science, University of Southampton, Southampton, United Kingdom
| | - Natalia Tejedor-Garavito
- WorldPop, Geography and Environmental Science, University of Southampton, Southampton, United Kingdom
| | - Adelle Wigley
- WorldPop, Geography and Environmental Science, University of Southampton, Southampton, United Kingdom
| | - Alessandra Carioli
- WorldPop, Geography and Environmental Science, University of Southampton, Southampton, United Kingdom
| | - Peter Gething
- Malaria Atlas Project, Telethon Kids Institute, Perth Children’s Hospital, Nedlands, Australia
- Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Daniel J. Weiss
- Malaria Atlas Project, Telethon Kids Institute, Perth Children’s Hospital, Nedlands, Australia
- Faculty of Health Sciences, Curtin University, Bentley, Australia
| | - Daniel Chandramohan
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Patrick G. T. Walker
- Faculty of Medicine, School of Public Health, Imperial College London, London, United Kingdom
| | - Matthew E. Cairns
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - R. Matthew Chico
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Malaria in pregnancy continues to exert a toll on pregnant women and their offspring. RECENT FINDINGS The burden of Plasmodium falciparum infection is especially large in Africa, and new data show lasting effects of maternal infection on the infant's neurocognitive development. Elsewhere, P. vivax infection causes relapsing infections that are challenging to prevent. Infection in first trimester of pregnancy is an area of increasing focus, and its adverse effects on pregnancy outcome are increasingly recognised. First-trimester infection is common and frequently acquired prior to conception. Although newer rapid diagnostic tests still have limited sensitivity, they may be useful in detection of early pregnancy malaria for treatment. Artemisinin-based combination therapies are efficacious in later pregnancy but have yet to be recommended in first trimester because of limited safety data. In Africa, intermittent preventive treatment in pregnancy (IPTp) with monthly sulfadoxine-pyrimethamine improves pregnancy outcomes, but sulfadoxine-pyrimethamine resistance is worsening. The alternative, IPTp with dihydroartemisinin-piperaquine, has greater antimalarial efficacy, but does not appear to improve pregnancy outcomes, because sulfadoxine-pyrimethamine has poorly understood nonmalarial benefits on birthweight. SUMMARY Novel IPTp regimens must be combined with interventions to strengthen protection from malaria infection acquired before and in early pregnancy.
Collapse
|
38
|
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
|
39
|
Jagannathan P, Kakuru A. Malaria in 2022: Increasing challenges, cautious optimism. Nat Commun 2022; 13:2678. [PMID: 35562368 PMCID: PMC9106727 DOI: 10.1038/s41467-022-30133-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/12/2022] [Indexed: 01/29/2023] Open
Affiliation(s)
- Prasanna Jagannathan
- grid.168010.e0000000419368956Department of Medicine, Stanford University, Stanford, CA United States ,grid.168010.e0000000419368956Department of Microbiology and Immunology, Stanford University, Stanford, CA United States
| | - Abel Kakuru
- grid.463352.50000 0004 8340 3103Infectious Diseases Research Collaboration, Kampala, Uganda
| |
Collapse
|
40
|
Mama A, Ahiabor C, Tornyigah B, Frempong NA, Kusi KA, Adu B, Courtin D, Houzé S, Deloron P, Ofori MF, Anang AK, Ariey F, Ndam NT. Intermittent preventive treatment in pregnancy with sulfadoxine–pyrimethamine and parasite resistance: cross-sectional surveys from antenatal care visit and delivery in rural Ghana. Malar J 2022; 21:107. [PMID: 35346205 PMCID: PMC8962208 DOI: 10.1186/s12936-022-04124-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 03/13/2022] [Indexed: 11/28/2022] Open
Abstract
Background Despite decades of prevention efforts, the burden of malaria in pregnancy (MiP) remains a great public health concern. Sulfadoxine-pyrimethamine (SP), used as intermittent preventive treatment in pregnancy (IPTp-SP) is an important component of the malaria prevention strategy implemented in Africa. However, IPTp-SP is under constant threat from parasite resistance, thus requires regular evaluation to inform decision-making bodies. Methods In two malaria endemic communities in the Volta region (Adidome and Battor), a cross-sectional hospital-based study was conducted in pregnant women recruited at their first antenatal care (ANC) visit and at delivery. Basic clinical and demographic information were documented and their antenatal records were reviewed to confirm IPTp-SP adherence. Peripheral and placental blood were assayed for the presence of Plasmodium falciparum parasites by quantitative polymerase chain reaction (qPCR). One hundred and twenty (120) positive samples were genotyped for mutations associated with SP resistance. Results At first ANC visit, P. falciparum prevalence was 28.8% in Adidome and 18.2% in Battor. At delivery, this decreased to 14.2% and 8.2%, respectively. At delivery, 66.2% of the women had taken at least the recommended 3 or more doses of IPTp-SP and there was no difference between the two communities. Taking at least 3 IPTp-SP doses was associated with an average birth weight increase of more than 360 g at both study sites compared to women who did not take treatment (p = 0.003). The Pfdhfr/Pfdhps quintuple mutant IRNI-A/FGKAA was the most prevalent (46.7%) haplotype found and the nonsynonymous Pfdhps mutation at codon A581G was higher at delivery among post-SP treatment isolates (40.6%) compared to those of first ANC (10.22%). There was also an increase in the A581G mutation in isolates from women who took 3 or more IPTp-SP. Conclusions This study confirms a positive impact following the implementation of the new IPTp-SP policy in Ghana in increasing the birth weight of newborns. However, the selection pressure exerted by the recommended 3 or more doses of IPTp-SP results in the emergence of parasites carrying the non-synonymous mutation on codon A581G. This constant selective pressure calls into question the time remaining for the clinical utility of IPTp-SP treatment during pregnancy in Africa.
Collapse
|
41
|
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
|
42
|
Waltmann A, McQuade ETR, Chinkhumba J, Operario DJ, Mzembe E, Itoh M, Kayange M, Puerto-Meredith SM, Mathanga DP, Juliano JJ, Carroll I, Bartelt LA, Gutman JR, Meshnick SR. The positive effect of malaria IPTp-SP on birthweight is mediated by gestational weight gain but modifiable by maternal carriage of enteric pathogens. EBioMedicine 2022; 77:103871. [PMID: 35217408 PMCID: PMC8866062 DOI: 10.1016/j.ebiom.2022.103871] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Poor pregnancy and birth outcomes are common in sub-Saharan Africa and have complex aetiologies. Sulfadoxine-pyrimethamine (SP), given for intermittent preventive therapy of malaria in pregnancy (IPTp), is one of few existing interventions that improves outcomes of both mother and baby despite widespread SP-resistant malaria. Compelling evidence exists that malaria-independent pathways contribute to this protective effect, but the exact sources of non anti-malarial protection remained unknown. We hypothesized that the beneficial effect of SP on birthweight is mediated by SP activity on maternal factors, including increased gestational weight gain and antibiotic activity on pathogens in the maternal gut. METHODS Expectant mothers from a larger randomized control trial comparing the efficacy of IPTp-SP to IPTp with dihydroartemisinin-piperaquine (DP) were also enrolled in this sub-study study at their first antenatal care visit before commencement of IPTp (n = 105). Participants were followed monthly until delivery. Weights and mid-to-upper-arm circumferences (MUAC) were recorded. Monthly stool samples were collected and screened for five Escherichia coli pathotypes, Shigella spp., Vibrio cholerae, Salmonella, Campylobacter coli/jejuni, and three protozoa (Giardia spp., Entameba histolytica, and Cryptosporidium spp.) using previously validated molecular assays. FINDINGS IPTp-SP vs. IPTP-DP was associated with higher maternal gestational weight gain (GWG) and nutritional indicators (MUAC and body-mass index, BMI). GWG was found to be a mediator of the birthweight and IPTp-SP relationship, as the birthweight of SP infants, but not DP infants, varied according to maternal GWG. The burden of maternal enteric infections was high. The three most commonly observed pathogens were enteroaggregative E. coli (EAEC), atypical enteropathogenic E.coli/enterohaemorrhagic E. coli (aEPEC/EHEC), and typical enteropathogenic E.coli (tEPEC). We found that SP reduced the prevalence of EAEC in a dose-dependent manner. After 3 or more doses, SP-recipients were 90% less likely to be infected with EAEC compared to DP-recipients (ORadj = 0.07, CI95 = 0.12, 0.39, p = 0.002). Compared to DP, this coincided with higher maternal gestational weight gain (GWG) and nutritional indicators (MUAC and body-mass index, BMI). The beneficial effect of SP on maternal GWG, MUAC and BMI, was lower if SP mothers had detectable EAEC, aEPEC/EHEC, tEPEC, and LT-ETEC at baseline. Maternal EAEC and tEPEC at baseline associated with lower birthweight for babies of both SP mothers and DP mothers. When comparing IPTp regimens, the positive effect of SP on birthweight compared to DP was only observed for infants of women who did not test positive for EAEC at baseline (adjusted mean birthweight difference SP vs. DP = 156.0 g, CI95 = -18.0 g, 336.9 g, p = 0.087), though confidence intervals crossed the null. INTERPRETATION Our findings indicate that in pregnant Malawian women, IPTp-SP vs. IPTp-DP is consistently associated with higher MUAC, BMI, and GWG following the WHO-recommended regimen of at least 3 doses, but carriage of maternal gut pathogens before initiation of IPTp lessens this effect. Because GWG was a mediator of the association between birthweight and SP, we show that SP's previously proven positive effect on birthweight is by promoting maternal weight gain. Overall, our results present one plausible pathway SP exerts malaria-independent protection against poor birth outcomes in the context of its waning antimalarial activity and warrants further investigation. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Andreea Waltmann
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| | | | - Jobiba Chinkhumba
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Darwin J Operario
- Division of Infectious Diseases & International Health, Department of Medicine, University of Virginia, VA, USA
| | - Enala Mzembe
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Megumi Itoh
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | | - Don P Mathanga
- Malaria Alert Centre (MAC), University of Malawi College of Medicine, Blantyre, Malawi
| | - Jonathan J Juliano
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Ian Carroll
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Luther A Bartelt
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Steven R Meshnick
- Institute for Global Health and Infectious Diseases, School of Medicine, University of North Carolina, Chapel Hill, NC, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
43
|
Mahamar A, Sumner KM, Levitt B, Freedman B, Traore A, Barry A, Issiaka D, Dembele AB, Kanoute MB, Attaher O, Diarra BN, Sagara I, Djimde A, Duffy PE, Fried M, Taylor SM, Dicko A. Effect of three years' seasonal malaria chemoprevention on molecular markers of resistance of Plasmodium falciparum to sulfadoxine-pyrimethamine and amodiaquine in Ouelessebougou, Mali. Malar J 2022; 21:39. [PMID: 35135546 PMCID: PMC8822718 DOI: 10.1186/s12936-022-04059-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 01/21/2022] [Indexed: 11/11/2022] Open
Abstract
Background In 2012, seasonal malaria chemoprevention (SMC) was recommended as policy for malaria control by the World Health Organization (WHO) in areas of highly seasonal malaria transmission across the Sahel sub-region in Africa along with monitoring of drug resistance. We assessed the long-term impact of SMC on Plasmodium falciparum resistance to sulfadoxine-pyrimethamine (SP) and amodiaquine (AQ) over a 3-year period of SMC implementation in the health district of Ouelessebougou, Mali. Methods In 8 randomly selected sub-districts of Ouelessebougou, Mali, children aged 0–5 years were randomly selected during cross-sectional surveys at baseline (August 2014) and 1, 2 and 3 years post-SMC, at the beginning and end of the malaria transmission season. Blood smears and blood spots on filter paper were obtained and frequencies of mutation in P. falciparum genes related to resistance to SP and AQ (Pfdhfr, Pfdhps, Pfmdr1, and Pfcrt) were assessed by PCR amplification on individual samples and PCR amplification followed by deep sequencing on pooled (by site and year) samples. Results At each survey, approximately 50–100 individual samples were analysed by PCR amplification and a total of 1,164 samples were analysed by deep sequencing with an average read depth of 18,018–36,918 after pooling by site and year. Most molecular markers of resistance did not increase in frequency over the period of study (2014–2016). After 3 years of SMC, the frequencies of Pfdhps 540E, Pfdhps 437G and Pfcrt K76T remained similar compared to baseline (4.0 vs 1.4%, p = 0.41; 74.5 vs 64.6%, p = 0.22; 71.3 vs 67.4%, p = 0.69). Nearly all samples tested carried Pfdhfr 59R, and this proportion remained similar 3 years after SMC implementation (98.8 vs 100%, p = 1). The frequency of Pfmdr1 N86Y increased significantly over time from 5.6% at baseline to 18.6% after 3 years of SMC (p = 0.016). Results of pooled analysis using deep sequencing were consistent with those by individual analysis with standard PCR, but also indicated for the first time the presence of mutations at the Pfdhps A581G allele at a frequency of 11.7% after 2 years of SMC, as well as the Pfdhps I431V allele at frequencies of 1.6–9.3% following 1 and 2 years of SMC, respectively. Conclusion Two and 3 years of SMC implementation were associated with increased frequency of the Pfmdr1 N86Y mutation but not Pfdhps 540E, Pfdhps 437G and Pfcrt K76T. The first-time detection of the Pfdhps haplotype bearing the I431V and A581G mutations in Mali, even at low frequency, warrants further long-term surveillance.
Collapse
Affiliation(s)
- Almahamoudou Mahamar
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali.
| | - Kelsey M Sumner
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA.,Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Brandt Levitt
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Betsy Freedman
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Aliou Traore
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Amadou Barry
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Djibrilla Issiaka
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Adama B Dembele
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Moussa B Kanoute
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Oumar Attaher
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | | | - Issaka Sagara
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Abdoulaye Djimde
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology (LMIV), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology (LMIV), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Steve M Taylor
- Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA.,Duke Global Health Institute, Duke University, Durham, NC, USA
| | - Alassane Dicko
- Malaria Research & Training Center, Faculty of Medicine, Pharmacy and Dentistry, University of Science, Techniques and Technologies (USTT), Bamako, Mali
| |
Collapse
|
44
|
Sundararaman SA, Odom John AR. Prevention of malaria in pregnancy: The threat of sulfadoxine-pyrimethamine resistance. Front Pediatr 2022; 10:966402. [PMID: 36061376 PMCID: PMC9433640 DOI: 10.3389/fped.2022.966402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Malaria infection in pregnancy can lead to adverse outcomes for both the pregnant person and fetus. The administration of intermittent preventative therapy (IPTp) with sulfadoxine-pyrimethamine (SP) during pregnancy (IPTp-SP) improves outcomes, including severe maternal anemia, placental malaria infection, and low infant birth weight. The WHO recommends IPTp-SP for pregnant individuals living in areas of moderate or high malaria transmission in Africa. The current regimen consists of two or more doses of SP starting as early as possible in the second trimester, at least 1 month apart. Unfortunately, rising Plasmodium falciparum SP resistance throughout Africa threatens to erode the benefits of SP. Recent studies have shown a decrease in IPTp-SP efficacy in areas with high SP resistance. Thus, there is an urgent need to identify new drug regimens that can be used for intermittent preventative therapy in pregnancy. In this review, we discuss recent data on P. falciparum SP resistance in Africa, the effect of resistance on IPTp-SP, and studies of alternative IPTp regimens. Finally, we present a framework for the ideal pharmacokinetic and pharmacodynamic properties for future IPTp regimens.
Collapse
Affiliation(s)
- Sesh A Sundararaman
- Department of Pediatrics, Children's Hospital of Philadelphia, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Audrey R Odom John
- Department of Pediatrics, Children's Hospital of Philadelphia, The Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
45
|
Gutman JR, Khairallah C, Stepniewska K, Tagbor H, Madanitsa M, Cairns M, L'lanziva AJ, Kalilani L, Otieno K, Mwapasa V, Meshnick S, Kariuki S, Chandramohan D, Desai M, Taylor SM, Greenwood B, ter Kuile FO. Intermittent screening and treatment with artemisinin-combination therapy versus intermittent preventive treatment with sulphadoxine-pyrimethamine for malaria in pregnancy: a systematic review and individual participant data meta-analysis of randomised clinical trials. EClinicalMedicine 2021; 41:101160. [PMID: 34746720 PMCID: PMC8556518 DOI: 10.1016/j.eclinm.2021.101160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/16/2021] [Accepted: 09/30/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND In sub-Saharan Africa, the efficacy of intermittent preventive therapy in pregnancy with sulphadoxine-pyrimethamine (IPTp-SP) for malaria in pregnancy is threatened by parasite resistance. We conducted an individual-participant data (IPD) meta-analysis to assess the efficacy of intermittent screening with malaria rapid diagnostic tests (RDTs) and treatment of RDT-positive women with artemisinin-based combination therapy (ISTp-ACT) compared to IPTp-SP, and understand the importance of subpatent infections. METHODS We searched MEDLINE and the Malaria-in-Pregnancy Library on May 6, 2021 for trials comparing ISTp-ACT and IPTp-SP. Generalised linear regression was used to compare adverse pregnancy outcomes (composite of small-for-gestational-age, low birthweight (LBW), or preterm delivery) and peripheral or placental Plasmodium falciparum at delivery. The effects of subpatent (PCR-positive, RDT/microscopy-negative) infections were assessed in both arms pooled using multi-variable fixed-effect models adjusting for the number of patent infections. PROSPERO registration: CRD42016043789. FINDINGS Five trials conducted between 2007 and 2014 contributed (10,821 pregnancies), two from high SP-resistance areas where dhfr/dhps quintuple mutant parasites are saturated, but sextuple mutants are still rare (Kenya and Malawi), and three from low-resistance areas (West-Africa). Four trials contributed IPD data (N=10,362). At delivery, the prevalence of any malaria infection (relative risk [RR]=1.08, 95% CI 1.00-1.16, I2=67.0 %) and patent infection (RR=1.02, 0.61-1.16, I2=0.0%) were similar. Subpatent infections were more common in ISTp recipients (RR=1.31, 1.05-1.62, I2=0.0%). There was no difference in adverse pregnancy outcome (RR=1.00, 0.96-1.05; studies=4, N=9,191, I2=54.5%). Subpatent infections were associated with LBW (adjusted RR=1.13, 1.07-1.19), lower mean birthweight (adjusted mean difference=32g, 15-49), and preterm delivery (aRR=1.35, 1.15-1.57). INTERPRETATION ISTp-ACT was not superior to IPTp-SP and may result in more subpatent infections than the existing IPTp-SP policy. Subpatent infections were associated with increased LBW and preterm delivery. More sensitive diagnostic tests are needed to detect and treat low-grade infections. FUNDING Centers for Disease Control and Prevention and Worldwide Antimalarial Resistance Network.
Collapse
Affiliation(s)
- Julie R Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Kasia Stepniewska
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
| | - Harry Tagbor
- University of Health and Allied Science, Ho, Ghana
| | | | | | - Anne Joan L'lanziva
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Linda Kalilani
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kephas Otieno
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Steve Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Simon Kariuki
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | | | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Steve M. Taylor
- Division of Infectious Diseases and Duke Global Health Institute, Duke University Medical Center, Durham, NC, USA
| | | | - 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
| |
Collapse
|
46
|
Chu X, Yan P, Zhang N, Chen N, Liu Y, Feng L, Li M, Zhang Z, Wang Q, Wang S, Yang K. The efficacy and safety of intermittent preventive treatment with sulphadoxine-pyrimethamine vs artemisinin-based drugs for malaria: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg 2021; 116:298-309. [PMID: 34651193 DOI: 10.1093/trstmh/trab158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/27/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Malaria is one of the most serious global problems. The objective of this study is to assess whether intermittent preventive treatment (IPT) using artemisinin-based combination therapies (ACTs) was a promising alternative to IPT with sulphadoxine-pyrimethamine (IPT-SP). METHODS We searched the following sources up to 12 August 2020: PubMed, The Cochrane Library, Embase, Web of Science, CNKI, CBM, VIP and WanFang Database from inception. The randomized controlled trials comparing SP with ACTs for malaria were included. Data were pooled using Stata.14 software. We performed subgroup analysis based on the different types of ACTs groups and participants. RESULTS A total of 13 studies comprising 5180 people were included. The meta-analysis showed that ACTs had the lower risk of number of any parasitemia (RR=0.46; 95% CI 0.22 to 0.96, p=0.039; I2=90.50%, p<0.001), early treatment failure (RR=0.17; 95% CI 0.06 to 0.48, p<0.001; I2=66.60%, p=0.011) and late treatment failure (RR=0.34; 95% CI 0.13 to 0.92, p<0.001; I2=87.80%, p<0.001) compared with SP. There was no significant difference in adequate clinical response, average hemoglobin and adverse neonatal outcomes. CONCLUSION Combinations with ACTs appear promising as suitable alternatives for IPT-SP.
Collapse
Affiliation(s)
- Xiajing Chu
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Peijing Yan
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610044, China
| | - Na Zhang
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Nan Chen
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Yang Liu
- Institute for Health Toxicology, School of Public Health, Lanzhou University, Lanzhou,730000, China
| | - Lufang Feng
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Meixuan Li
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| | - Ziyao Zhang
- School of Foreign Language, Lanzhou University of Arts and Science, Lanzhou, 730000, China
| | - Qi Wang
- Health Policy PhD Program, McMaster University, Hamilton, Ontario, L8S 4K1, Canada.,McMaster Health Forum, McMaster University, Hamilton, Ontario, L8S 4L6, Canada.,Department of Health Research Methods, Evidence and Impact, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| | | | - Kehu Yang
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, 730000, China.,Health Technology Assessment Center of Lanzhou University, School of Public Health, Lanzhou University, Lanzhou,730000, China.,Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, 730000, China
| |
Collapse
|
47
|
Unger HW, Laurita Longo V, Bleicher A, Ome-Kaius M, Karl S, Simpson JA, Karahalios A, Aitken EH, Rogerson SJ. The relationship between markers of antenatal iron stores and birth outcomes differs by malaria prevention regimen-a prospective cohort study. BMC Med 2021; 19:236. [PMID: 34607575 PMCID: PMC8491429 DOI: 10.1186/s12916-021-02114-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/31/2021] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Iron deficiency (ID) has been associated with adverse pregnancy outcomes, maternal anaemia, and altered susceptibility to infection. In Papua New Guinea (PNG), monthly treatment with sulphadoxine-pyrimethamine plus azithromycin (SPAZ) prevented low birthweight (LBW; <2500 g) through a combination of anti-malarial and non-malarial effects when compared to a single treatment with SP plus chloroquine (SPCQ) at first antenatal visit. We assessed the relationship between ID and adverse birth outcomes in women receiving SPAZ or SPCQ, and the mediating effects of malaria infection and haemoglobin levels during pregnancy. METHODS Plasma ferritin levels measured at antenatal enrolment in a cohort of 1892 women were adjusted for concomitant inflammation using C-reactive protein and α-1-acid glycoprotein. Associations of ID (defined as ferritin <15 μg/L) or ferritin levels with birth outcomes (birthweight, LBW, preterm birth, small-for-gestational-age birthweight [SGA]) were determined using linear or logistic regression analysis, as appropriate. Mediation analysis assessed the degree of mediation of ID-birth outcome relationships by malaria infection or haemoglobin levels. RESULTS At first antenatal visit (median gestational age, 22 weeks), 1256 women (66.4%) had ID. Overall, ID or ferritin levels at first antenatal visit were not associated with birth outcomes. There was effect modification by treatment arm. Amongst SPCQ recipients, ID was associated with a 81-g higher mean birthweight (95% confidence interval [CI] 10, 152; P = 0.025), and a twofold increase in ferritin levels was associated with increased odds of SGA (adjusted odds ratio [aOR] 1.25; 95% CI 1.06, 1.46; P = 0.007). By contrast, amongst SPAZ recipients, a twofold increase in ferritin was associated with reduced odds of LBW (aOR 0.80; 95% CI 0.67, 0.94; P = 0.009). Mediation analyses suggested that malaria infection or haemoglobin levels during pregnancy do not substantially mediate the association of ID with birth outcomes amongst SPCQ recipients. CONCLUSIONS Improved antenatal iron stores do not confer a benefit for the prevention of adverse birth outcomes in the context of malaria chemoprevention strategies that lack the non-malarial properties of monthly SPAZ. Research to determine the mechanisms by which ID protects from suboptimal foetal growth is needed to guide the design of new malaria prevention strategies and to inform iron supplementation policy in malaria-endemic settings. TRIAL REGISTRATION ClinicalTrials.gov NCT01136850 .
Collapse
Affiliation(s)
- Holger W Unger
- Department of Obstetrics and Gynaecology, Royal Darwin Hospital, Darwin, Northern Territory, Australia.,Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia.,Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Valentina Laurita Longo
- Catholic University of Sacred Heart, Rome, Italy.,Department of Obstetrics and Gynaecology, San Pietro-Fatebenefratelli Hospital, Rome, Italy
| | - Andie Bleicher
- Department of Medicine (RMH), Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Maria Ome-Kaius
- Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Stephan Karl
- Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Amalia Karahalios
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth H Aitken
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen J Rogerson
- Department of Medicine (RMH), Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia. .,Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
48
|
Hume-Nixon M, Quach A, Reyburn R, Nguyen C, Steer A, Russell F. A Systematic Review and meta-analysis of the effect of administration of azithromycin during pregnancy on perinatal and neonatal outcomes. EClinicalMedicine 2021; 40:101123. [PMID: 34541478 PMCID: PMC8436060 DOI: 10.1016/j.eclinm.2021.101123] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Currently there are trials in Africa and Asia investigating whether prophylactic azithromycin during pregnancy reduces infection-related neonatal morbidity and mortality. We undertook a systematic review and meta-analysis to determine the effect of azithromycin during pregnancy on perinatal and neonatal outcomes. METHODS We identified articles between January 1990 and 13th June 2021 by searching five electronic databases. Randomised control trials (RCTs) that included pregnant women administered azithromycin alone or in combination with other medications, and that reported outcomes of low birthweight (LBW), prematurity, stillbirth, and neonatal deaths, infections, and admissions, were eligible. Fixed effects meta-analyses were used for primary analysis. Quality appraisal was performed using Cochrane's Risk of Bias 2 tool. This review was registered with PROSPERO, CRD42019127099. FINDINGS The search generated 5777 studies, of which 14 studies were included involving 17,594 participants. Most studies investigated azithromycin as Intermittent Preventive Treatment in Pregnancy (IPTp) for malaria. More than 50% of the studies had low risk of bias for all outcomes, except for LBW and neonatal admissions. Fixed-effects meta-analyses found that azithromycin reduced the risk of LBW (seven studies, Pooled RR 0·79; 95% CI 0·68-0·93; I2 = 0·00%), and prematurity compared to controls (eight studies, Pooled RR 0·87; 95% CI 0·78-0·98; I2 = 23·28%). There was no strong evidence of any effect on neonatal mortality, infections and admissions. There was an increase in stillbirth but the 95% CI crossed the null value (seven studies, Pooled RR 1·39; 95% CI 0·94 - 2.07; I2=0·00%). However this review was limited by differences in the types of intervention and study populations, and inconsistency in outcome reporting between studies. INTERPRETATION Prophylactic azithromycin during pregnancy reduces LBW and prematurity. However, as azithromycin has been investigated as part of IPTp, it is unclear whether it would improve perinatal and neonatal outcomes in non-malaria endemic settings. The potential harm on stillbirth rates needs further investigation. FUNDING None.
Collapse
Affiliation(s)
- Maeve Hume-Nixon
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Alicia Quach
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Rita Reyburn
- Asia-Pacific Health, Murdoch Children's Research Institute, Melbourne, Australia
| | - Cattram Nguyen
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Andrew Steer
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Tropical Diseases Research Group, Murdoch Children's Research Institute, Melbourne, Australia
- Department of General Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Fiona Russell
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Asia-Pacific Health, Murdoch Children's Research Institute, Melbourne, Australia
| |
Collapse
|
49
|
Greenwood B, Cairns M, Chaponda M, Chico RM, Dicko A, Ouedraogo JB, Phiri KS, Ter Kuile FO, Chandramohan D. Combining malaria vaccination with chemoprevention: a promising new approach to malaria control. Malar J 2021; 20:361. [PMID: 34488784 PMCID: PMC8419817 DOI: 10.1186/s12936-021-03888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/20/2021] [Indexed: 11/13/2022] Open
Abstract
Malaria control has stalled in a number of African countries and novel approaches to malaria control are needed for these areas. The encouraging results of a recent trial conducted in young children in Burkina Faso and Mali in which a combination of the RTS,S/AS01E malaria vaccine and seasonal malaria chemoprevention led to a substantial reduction in clinical cases of malaria, severe malaria, and malaria deaths compared with the administration of either intervention given alone suggests that there may be other epidemiological/clinical situations in which a combination of malaria vaccination and chemoprevention could be beneficial. Some of these potential opportunities are considered in this paper. These include combining vaccination with intermittent preventive treatment of malaria in infants, with intermittent preventive treatment of malaria in pregnancy (through vaccination of women of child-bearing age before or during pregnancy), or with post-discharge malaria chemoprevention in the management of children recently admitted to hospital with severe anaemia. Other potential uses of the combination are prevention of malaria in children at particular risk from the adverse effects of clinical malaria, such as those with sickle cell disease, and during the final stages of a malaria elimination programme when vaccination could be combined with repeated rounds of mass drug administration. The combination of a pre-erythrocytic stage malaria vaccine with an effective chemopreventive regimen could make a valuable contribution to malaria control and elimination in a variety of clinical or epidemiological situations, and the potential of this approach to malaria control needs to be explored.
Collapse
Affiliation(s)
| | - Matthew Cairns
- London School of Hygiene and Tropical Medicine, London, UK
| | | | | | - Alassane Dicko
- Malaria Research and Training Centre, University of Science, Techniques and Technology of Bamako, Bamako, Mali
| | | | - Kamija S Phiri
- School of Public Health, Kamuzu University of Health Sciences, Blantyre, Malawi
| | | | | |
Collapse
|
50
|
Ter Kuile FO. Towards intermittent preventive therapy in pregnancy with dihydroartemisinin-piperaquine? Clin Pharmacol Ther 2021; 110:1432-1434. [PMID: 34389977 DOI: 10.1002/cpt.2394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/30/2021] [Indexed: 11/05/2022]
Abstract
Malaria is a major cause of adverse pregnancy outcomes in sub-Saharan Africa, but resistance to sulfadoxine-pyrimethamine, the only antimalarial recommended by the World Health Organisation for intermittent preventive therapy, is threatening the gains made in the last two decades. In this issue, Mlugu and colleagues present the results of a trial of dihydroartemisinin-piperaquine as an alternative to sulfadoxine-pyrimethamine. The results are impressive but raise the question why they differ so much from three previous trials.
Collapse
Affiliation(s)
- Feiko O Ter Kuile
- Liverpool School of Tropical Medicine, Pembroke Place, L3 5QA, Liverpool, United Kingdom
| |
Collapse
|