51
|
Das S, Kérah-Hinzoumbé C, Kebféné M, Srisutham S, Nagorngar TY, Saralamba N, Vongpromek R, Khomvarn T, Sibley CH, Guérin PJ, Imwong M, Dhorda M. Molecular surveillance for operationally relevant genetic polymorphisms in Plasmodium falciparum in Southern Chad, 2016–2017. Malar J 2022; 21:83. [PMID: 35279140 PMCID: PMC8917628 DOI: 10.1186/s12936-022-04095-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 02/19/2022] [Indexed: 11/17/2022] Open
Abstract
Background Resistance to anti-malarials is a serious threat to the efforts to control and eliminate malaria. Surveillance based on simple field protocols with centralized testing to detect molecular markers associated with anti-malarial drug resistance can be used to identify locations where further investigations are needed. Methods Dried blood spots were collected from 398 patients (age range 5–59 years, 99% male) with Plasmodium falciparum infections detected using rapid diagnostic tests over two rounds of sample collection conducted in 2016 and 2017 in Komé, South-West Chad. Specimens were genotyped using amplicon sequencing or qPCR for validated markers of anti-malarial resistance including partner drugs used in artemisinin-based combination therapy (ACT). Results No mutations in the pfk13 gene known to be associated with artemisinin resistance were found but a high proportion of parasites carried other mutations, specifically K189T (190/349, 54.4%, 95%CI 49.0–59.8%). Of 331 specimens successfully genotyped for pfmdr1 and pfcrt, 52% (95%CI 46.4–57.5%) carried the NFD-K haplotype, known to be associated with reduced susceptibility to lumefantrine. Only 20 of 336 (6.0%, 95%CI 3.7–9.0%) had parasites with the pfmdr1-N86Y polymorphism associated with increased treatment failures with amodiaquine. Nearly all parasites carried at least one mutation in pfdhfr and/or pfdhps genes but ‘sextuple’ mutations in pfdhfr—pfdhps including pfdhps -A581G were rare (8/336 overall, 2.4%, 95%CI 1.2–4.6%). Only one specimen containing parasites with pfmdr1 gene amplification was detected. Conclusions These results provide information on the likely high efficacy of artemisinin-based combinations commonly used in Chad, but suggest decreasing levels of sensitivity to lumefantrine and high levels of resistance to sulfadoxine-pyrimethamine used for seasonal malaria chemoprevention and intermittent preventive therapy in pregnancy. A majority of parasites had mutations in the pfk13 gene, none of which are known to be associated with artemisinin resistance. A therapeutic efficacy study needs to be conducted to confirm the efficacy of artemether-lumefantrine. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04095-9.
Collapse
|
52
|
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
|
53
|
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
|
54
|
Barrow A, Barrow S, Jobe A. Differentials in prevalence and correlates on uptake of tetanus toxoid and intermittent preventive treatment with sulfadoxine-pyrimethamine during pregnancy: A community-based cross-sectional study in The Gambia. SAGE Open Med 2022; 10:20503121211065908. [PMID: 35024140 PMCID: PMC8744190 DOI: 10.1177/20503121211065908] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 11/22/2021] [Indexed: 12/02/2022] Open
Abstract
Objectives: The study examined the differentials in prevalence and correlates on the
uptake of tetanus toxoid and intermittent preventive treatment of malaria
among pregnant women in The Gambia. Methods: The 2018 data from The Gambia Multiple Indicators Cluster Survey were
analyzed. Data from 6143 women of reproductive age who have given birth were
extracted for the analysis. Percentages and Chi-square tests were used. In
addition, a multivariable logistic regression model was used to calculate
the adjusted odds ratios (with a corresponding 95% confidence interval). The
level of significance was set at p < 0.05. Results: The prevalence of tetanus toxoid uptake among women in The Gambia was 88.2%,
while that of the adequate tetanus toxoid doses was 34.8%. The prevalence of
intermittent preventive treatment with sulfadoxine-pyrimethamine uptake
among maternal women in The Gambia was 98.6%, while that of the adequate
intermittent preventive treatment with sulfadoxine-pyrimethamine doses taken
was 34.3%. The identified statistically significant covariates of tetanus
toxoid immunization and intermittent preventive treatment with
sulfadoxine-pyrimethamine uptake includes women’s age, local government
areas, parity, use of radio, use of newspaper, and antenatal care
visits. Conclusion: The current utilization rate for adequate intermittent preventive treatment
with sulfadoxine-pyrimethamine and tetanus toxoid immunization during
pregnancy in The Gambia is very low and even below universal levels. The
country needs to strengthen more and effective mass media advocacy programs
that would target both rural and urban populace, and motivate maternal women
to ensure adequate vaccination against malaria and tetanus.
Collapse
Affiliation(s)
- Amadou Barrow
- Department of Public and Environmental Health, School of Medicine & Allied Health Sciences, University of The Gambia, Kanifing, The Gambia
| | - Sulayman Barrow
- Department of Public and Environmental Health, School of Medicine & Allied Health Sciences, University of The Gambia, Kanifing, The Gambia.,Public Health Unit, Brikama District Hospital, Ministry of Health, Brikama, The Gambia
| | - Amienatta Jobe
- Department of Obstetrics and Gynaecology, Edward Francis Small Teaching Hospital, Banjul, The Gambia
| |
Collapse
|
55
|
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
|
56
|
Ndiaye YD, Hartl DL, McGregor D, Badiane A, Fall FB, Daniels RF, Wirth DF, Ndiaye D, Volkman SK. Genetic surveillance for monitoring the impact of drug use on Plasmodium falciparum populations. Int J Parasitol Drugs Drug Resist 2021; 17:12-22. [PMID: 34333350 PMCID: PMC8342550 DOI: 10.1016/j.ijpddr.2021.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/24/2021] [Accepted: 07/07/2021] [Indexed: 11/23/2022]
Abstract
The use of antimalarial drugs is an effective strategy in the fight against malaria. However, selection of drug resistant parasites is a constant threat to the continued use of this approach. Antimalarial drugs are used not only to treat infections but also as part of population-level strategies to reduce malaria transmission toward elimination. While there is strong evidence that the ongoing use of antimalarial drugs increases the risk of the emergence and spread of drug-resistant parasites, it is less clear how population-level use of drug-based interventions like seasonal malaria chemoprevention (SMC) or mass drug administration (MDA) may contribute to drug resistance or loss of drug efficacy. Critical to sustained use of drug-based strategies for reducing the burden of malaria is the surveillance of population-level signals related to transmission reduction and resistance selection. Here we focus on Plasmodium falciparum and discuss the genetic signatures of a parasite population that are correlated with changes in transmission and related to drug pressure and resistance as a result of drug use. We review the evidence for MDA and SMC contributing to malaria burden reduction and drug resistance selection and examine the use and impact of these interventions in Senegal. Throughout we consider best strategies for ongoing surveillance of both population and resistance signals in the context of different parasite population parameters. Finally, we propose a roadmap for ongoing surveillance during population-level drug-based interventions to reduce the global malaria burden.
Collapse
Affiliation(s)
| | | | - David McGregor
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | | | - Fatou Ba Fall
- Programme National de Lutte Contre le Paludisme, Senegal.
| | - Rachel F Daniels
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA.
| | - Dyann F Wirth
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA.
| | | | - Sarah K Volkman
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Simmons University, Boston, MA, USA.
| |
Collapse
|
57
|
Kabuya JBB, Ippolito MM, Sikalima J, Tende C, Champo D, Mwakazanga D, Young AMP, Mulenga M, Chongwe G, Manyando C. Safety and efficacy of intermittent presumptive treatment with sulfadoxine-pyrimethamine using rapid diagnostic test screening and treatment with dihydroartemisinin-piperaquine at the first antenatal care visit (IPTp-SP+): study protocol for a randomized controlled trial. Trials 2021; 22:820. [PMID: 34801059 PMCID: PMC8605457 DOI: 10.1186/s13063-021-05745-0] [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: 04/21/2021] [Accepted: 10/20/2021] [Indexed: 11/23/2022] Open
Abstract
Background Intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) is recommended by the World Health Organization for the prevention of malaria in pregnancy (MIP)-associated adverse outcomes in high burden areas. However, the efficacy of IPTp-SP has decreased in step with increasing parasite drug resistance. Suitable alternative strategies are needed. Methods This is a protocol for a phase IIIb open-label, two-armed randomized controlled superiority trial to assess the safety and efficacy of a hybrid approach to IPTp combining screening and treatment with dihydroartemisinin-piperaquine (DP) to the current IPTp-SP regimen at the first antenatal care clinic visit. Pregnant women without HIV infection and without signs or symptoms of malaria will be randomized to either standard IPTp-SP or hybrid IPTp-SP plus screening and treatment (IPTp-SP+). In the IPTp-SP+ arm, participants who screen positive by rapid diagnostic test for P. falciparum will be treated with DP at the first antenatal visit while those who screen negative will receive SP per current guidelines. All participants will be administered SP on days 35 and 63 and will be actively followed biweekly up to day 63 and then monthly until delivery. Infants will be followed until 1 year after delivery. The primary endpoint is incident PCR-confirmed MIP at day 42. Secondary endpoints include incident MIP at other time points, placental malaria, congenital malaria, hemoglobin trends, birth outcomes, and incidence of adverse events in infants up to the first birthday. Discussion A hybrid approach to IPTp that combines screening and treatment with an artemisinin-based combination therapy at the first visit with standard IPTp-SP is hypothesized to confer added benefit over IPTp-SP alone in a high malaria transmission area with prevalent SP resistant parasites. Trial registration Pan African Clinical Trials Registry 201905721140808. Registered retrospectively on 11 May 2019 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-021-05745-0.
Collapse
Affiliation(s)
- Jean-Bertin Bukasa Kabuya
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia.
| | - Matthew M Ippolito
- Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jay Sikalima
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | - Clifford Tende
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | - Davies Champo
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | - David Mwakazanga
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | | | - Modest Mulenga
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | - Gershom Chongwe
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| | - Christine Manyando
- Department of Clinical Sciences, Tropical Diseases Research Centre, P.O. Box 71769, Copperbelt Province, Ndola, Zambia
| |
Collapse
|
58
|
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
|
59
|
Lingani M, Zango SH, Valéa I, Valia D, Sanou M, Samandoulougou SO, Robert A, Tinto H, Dramaix M, Donnen P. Magnitude of low birthweight in malaria endemic settings of Nanoro, rural Burkina Faso: a secondary data analysis. Sci Rep 2021; 11:21332. [PMID: 34716389 PMCID: PMC8556330 DOI: 10.1038/s41598-021-00881-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/15/2021] [Indexed: 12/02/2022] Open
Abstract
Low birthweight (LBW) is a worldwide problem that particularly affects developing countries. However, limited information is available on its magnitude in rural area of Burkina Faso. This study aimed to estimate the prevalence of low birthweight and to identify its associated factors in Nanoro health district. A secondary analysis of data collected during a cross-sectional survey was conducted to assess the prevalence of low birthweight in Nanoro health and demographic surveillance system area (HDSS). Maternal characteristics extracted from antenatal care books or by interview, completed by malaria diagnosis were examined through a multi-level logistic regression to estimate odd-ratios of association with low birthweight. Significance level was set at 5%. Of the 291 neonates examined, the prevalence of low birthweight was 12%. After adjustment for socio-demographic, obstetric and malaria prevention variables, being primigravid (OR = 8.84, [95% CI: 3.72-21.01]), or multigravid with history of stillbirth (OR = 5.03, [95% CI: 1.54-16.40]), as well as the lack of long-lasting insecticide treated bed net use by the mother the night preceding the admission for delivery (OR = 2.5, [95% CI: 1.1-5.9]) were significantly associated with neonate low birthweight. The number of antenatal visits however did not confer any direct benefit on birthweight status within this study area. The prevalence of low birthweight was high in the study area and represents an important public health problem in Burkina Faso. In light of these results, a redefinition of the content of the antenatal care package is needed.
Collapse
Affiliation(s)
- Moussa Lingani
- École de Santé Publique, Université Libre de Bruxelles, Route de Lennik 808, CP594, 1070, Bruxelles, Belgique.
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso.
| | - Serge H Zango
- Epidemiology and Biostatistics Research Division, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Clos Chapelle-aux-Champs 30, B1.30.13, 1200, Brussels, Belgique
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso
| | - Innocent Valéa
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso
| | - Daniel Valia
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso
| | - Maïmouna Sanou
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso
| | - Sékou O Samandoulougou
- Evaluation Platform on Obesity Prevention, Quebec Heart and Lung Institute Research Center, Quebec City, QC, G1V 4G5, Canada
| | - Annie Robert
- Epidemiology and Biostatistics Research Division, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Clos Chapelle-aux-Champs 30, B1.30.13, 1200, Brussels, Belgique
| | - Halidou Tinto
- Institut de Recherche en Sciences de la Santé/Direction Régionale du Centre Ouest (IRSS/DRCO), BP 218, 11, Nanoro, Burkina Faso
| | - Michèle Dramaix
- École de Santé Publique, Université Libre de Bruxelles, Route de Lennik 808, CP594, 1070, Bruxelles, Belgique
| | - Philippe Donnen
- École de Santé Publique, Université Libre de Bruxelles, Route de Lennik 808, CP594, 1070, Bruxelles, Belgique
| |
Collapse
|
60
|
Darteh EKM, Dickson KS, Ahinkorah BO, Owusu BA, Okyere J, Salihu T, Bio Bediako V, Budu E, Agbemavi W, Edjah JO, Seidu AA. Factors influencing the uptake of intermittent preventive treatment among pregnant women in sub-Saharan Africa: a multilevel analysis. Arch Public Health 2021; 79:182. [PMID: 34670628 PMCID: PMC8529836 DOI: 10.1186/s13690-021-00707-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/07/2021] [Indexed: 11/10/2022] Open
Abstract
Background Intermittent Preventive Treatment (IPT) of malaria in pregnancy is a full therapeutic course of antimalarial sulfadoxine-pyrimethamine (SP) medicine given to pregnant women in their second trimester at routine antenatal care visits, regardless of whether the recipient is infected with malaria. Given the negative consequences of malaria in pregnancy, studies on Intermittent Preventive Therapy with Sulfadoxine-Pyrimethamine (IPTp-SP) are important benchmarks for understanding the extent of malaria control and prevention during pregnancy. We, therefore, examined the factors associated with the uptake of IPTp-SP among pregnant women in sub-Saharan Africa. Methods We used data from the current versions of the Malaria Indicators Survey of 12 countries in sub-Saharan Africa. Women aged 15–49 years participated in the surveys. The analyses were carried out using Stata version 14.2. Descriptive (frequencies and percentages) and multilevel regression analyses were carried out. The results of the multilevel regression analysis were presented as adjusted odds ratios (aOR) with 95% confidence intervals (CIs). Results The average prevalence of uptake of IPTp-SP among pregnant women in the studied sub-Saharan African countries was 30.69%, with the highest and lowest prevalences in Ghana (59.64%) and Madagascar (10.08%), respectively. Women aged 40–44 compared to those aged 15–19 (aOR = 1.147, 95%CI = [1.02,1.30) had higher odds of receiving 3 or more doses of IPTp-SP. Women with a secondary/higher level of education compared to those with no formal education (aOR = 1.12, 95%CI = 1.04,1.20] also had higher odds of receiving 3 or more doses of IPTp-SP. Women who were exposed to malaria messages on the radio (aOR = 1.07, 95%CI = 1.02,1.12] and television (aOR = 1.13,95%CI = [1.05,1.21]) had higher odds of receiving 3 or more doses of IPTp-SP compared to those who were not exposed. Conclusion Our study indicates that the uptake of IPTp-SP is relatively low among the countries included in this study, with significant inter-country variations. Higher educational level, exposure to media, low parity, and higher age group were associated with higher odds of optimal IPTp-SP uptake. National policies, programs, guidance services such as information service and counselling and other interventions aimed at improving the coverage and uptake of IPTp-SP must be targeted at women with low level of education, non-exposure to media, high parity, and younger age group to attain the desired outcome. Supplementary Information The online version contains supplementary material available at 10.1186/s13690-021-00707-z.
Collapse
Affiliation(s)
| | | | - Bright Opoku Ahinkorah
- School of Public Health, Faculty of Health, University of Technology Sydney, Sydney, Australia
| | | | - Joshua Okyere
- Department of Population and Health, University of Cape Coast, Cape Coast, Ghana
| | - Tarif Salihu
- Department of Population and Health, University of Cape Coast, Cape Coast, Ghana
| | - Vincent Bio Bediako
- Department of Population and Health, University of Cape Coast, Cape Coast, Ghana
| | - Eugene Budu
- Department of Population and Health, University of Cape Coast, Cape Coast, Ghana
| | - Wonder Agbemavi
- Department of Population and Health, University of Cape Coast, Cape Coast, Ghana.
| | | | - Abdul-Aziz Seidu
- Department of Estate management, Takoradi Technical University, Takoradi, Ghana
| |
Collapse
|
61
|
Yamauchi M, Hirai M, Tachibana SI, Mori T, Mita T. Fitness of sulfadoxine-resistant Plasmodium berghei harboring a single mutation in dihydropteroate synthase (DHPS). Acta Trop 2021; 222:106049. [PMID: 34273314 DOI: 10.1016/j.actatropica.2021.106049] [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: 11/03/2020] [Revised: 05/30/2021] [Accepted: 07/05/2021] [Indexed: 12/24/2022]
Abstract
Genetic changes conferring drug resistance are generally believed to impose fitness costs to pathogens in the absence of the drug. However, the fitness of resistant parasites against sulfadoxine/pyrimethamine has been inconclusive in Plasmodium falciparum. This is because resistance is conferred by the complex combination of mutations in dihydropteroate synthase (dhps) and dihydrofolate reductase (dhfr), which makes it difficult to separately assess the extent and magnitude of the costs imposed by mutations in dhps and dhfr. To assess the fitness costs imposed by sulfadoxine resistance alone, we generated a transgenic rodent malaria parasite, P. berghei clone harboring an A394G mutation in dhps (PbDHPS-A394G), corresponding to the causative mutation for sulfadoxine resistance in P. falciparum (PfDHPS-A437G). A four-day suppressive test confirmed that the PbDHPS-A394G clone was resistant to sulfadoxine. PbDHPS-A394G and wild-type clones showed similar growth rates and gametocyte production. This observation was confirmed in competitive experiments in which PbDHPS-A394G and wild-type clones were co-infected into mice to directly assess the survival competition between them. In the mosquitoes, there were no significant differences in oocyst production between PbDHPS-A394G and wild-type. These results indicate that the PbDHPS-A394G mutation alters the parasites to sulfadoxine resistance but may not impose fitness disadvantages during the blood stages in mice and oocyst formation in mosquitoes. These results partly explain the persistence of the PfDHPS-A437G mutant in the natural parasite populations.
Collapse
|
62
|
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
|
63
|
Bakken L, Iversen PO. The impact of malaria during pregnancy on low birth weight in East-Africa: a topical review. Malar J 2021; 20:348. [PMID: 34429121 PMCID: PMC8386002 DOI: 10.1186/s12936-021-03883-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/16/2021] [Indexed: 11/10/2022] Open
Abstract
Background Globally, approximately 15% of all babies are born with low birth weight (< 2.5 kg) and ≥ 90% of them are born in low- and middle-income countries. Malaria infection in pregnancy remains a public health concern as it can affect both the mother and the newborn. Notably, it increases the risk of newborns with low birth weight. The World Health Organization (WHO) recommends intermittent preventive treatment with ≥ 3 doses of sulfadoxine-pyrimethamine (SP) during pregnancy in areas with moderate to high malaria transmission in Africa. The aim of this topical review is to give an overview of the impact of malaria infection during pregnancy on low birth weight, with focus on East Africa where malaria is endemic. Methods Eleven studies were selected according to a predefined set of criteria. Results Three studies showed a significant reduction in the prevalence of low birth weight with intermittent preventive treatment with SP, whereas four studies found no significant impact of such treatment on low birth weight. The number of SP doses and compliance to this treatment may in part explain these discrepancies. Pregnant women with frequent symptomatic malaria infection had significantly higher risk of placental malaria. Conclusion The WHO recommendation of ≥ 3 doses of intermittent preventive treatment with SP during pregnancy seem effective in preventing low birth weight, but treatment compliance is a challenge. Malaria prophylaxis is important during pregnancy, especially in endemic areas of malaria, such as East Africa.
Collapse
Affiliation(s)
- Line Bakken
- Department of Nutrition, University of Oslo, Oslo, Norway
| | - Per Ole Iversen
- Department of Nutrition, University of Oslo, Oslo, Norway. .,Department of Blood Transfusion and Haematology, Muhimbili University of Health and Allied Sciences, Dar-es-Salaam, Tanzania.
| |
Collapse
|
64
|
Evaluation of the usefulness of intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine in a context with increased resistance of Plasmodium falciparum in Kingasani Hospital, Kinshasa in the Democratic Republic of Congo. INFECTION GENETICS AND EVOLUTION 2021; 94:105009. [PMID: 34284138 DOI: 10.1016/j.meegid.2021.105009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Increasing resistance of Plasmodium falciparum to sulfadoxine-pyrimethamine (SP) threatens its usefulness for intermittent preventive treatment in pregnancy (IPTp-SP). The prophylactic effects of IPTp-SP on maternal malaria and adverse pregnancy outcomes were evaluated in Kingasani Hospital, Kinshasa in the Democratic Republic of Congo (DRC). METHODS Laboring women (n = 844) and respective newborns were investigated. Blood samples collected from women were tested for malaria using rapid diagnostic test (RDT), blood smears examination, and real-time PCR. The hemoglobin level was measured by HemoCue© analyzer. A PCR-RFLP method was applied for detecting N51I, C59R, and S108N mutations on dhfr along with A437G and K540E mutations on dhps in P. falciparum positive samples. Logistic regression models assessed relationships between IPTp-SP uptake and pregnancy outcomes. RESULTS P. falciparum malaria was detected at delivery in 10.8% of women and was statistically associated with fever during the pregnancy (OR = 2.9 [1.5; 6.3]; p = 0.004) and maternal anemia (OR = 3.9 [2.4; 6.3]; p < 0.001). One out of five parasites was a quintuple mutant encoding dhfr mutations 51I, 59R, and 108 N along with dhps mutations 437G and 540E. The molecular profile of parasites (i.e., 32.6% of parasites carrying dhps K540E) was suitable with continued use of SP for IPTp. IPTp-SP uptake was not associated with reduced maternal malaria, fever reported in pregnancy, or fetal deaths (p > 0.05). Conversely, three or more doses of SP were associated with reduced maternal anemia at delivery (OR = 0.4 [0.2; 0.9]; p = 0.024), shortened gestation (OR = 0.4 [0.2; 0.8]; p = 0.009), and low-birth weights (OR = 0.2 [0.1; 0.5]; p < 0.001). CONCLUSION IPTp-SP was not associated with reduced maternal malaria in our study, but evidence was found of a prophylactic effect against adverse pregnancy outcomes. To counteract further loss of clinical effects of IPTp-SP in the study population, alternative strategies able to improve its anti-malarial efficacy such as combination of SP with partner molecules should be implemented.
Collapse
|
65
|
Amimo F, Lambert B, Magit A, Sacarlal J, Hashizume M, Shibuya K. Plasmodium falciparum resistance to sulfadoxine-pyrimethamine in Africa: a systematic analysis of national trends. BMJ Glob Health 2021; 5:bmjgh-2020-003217. [PMID: 33214174 PMCID: PMC7678238 DOI: 10.1136/bmjgh-2020-003217] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/13/2020] [Accepted: 09/08/2020] [Indexed: 12/02/2022] Open
Abstract
Introduction The rising burden of drug resistance is a major challenge to the global fight against malaria. We estimated national Plasmodium falciparum resistance to sulfadoxine-pyrimethamine (SP) across Africa, from 2000 to 2020. Methods We assembled molecular, clinical and endemicity data covering malaria-endemic African countries up to December 2018. Subsequently, we reconstructed georeferenced patient data, using pfdhps540E and pfdhps581G to measure mid-level and high-level SP resistance. Gaussian process regression was applied to model spatiotemporal standardised prevalence. Results In eastern Africa, mid-level SP resistance increased by 64.0% (95% uncertainty interval, 30.7%–69.8%) in Tanzania, 55.4% (31.3%–65.2%) in Sudan, 45.7% (16.8%–54.3%) in Mozambique, 29.7% (10.0%–45.2%) in Kenya and 8.7% (1.4%–36.8%) in Malawi from 2000 to 2010. This was followed by a steady decline of 76.0% (39.6%–92.6%) in Sudan, 65.7% (25.5%–85.6%) in Kenya and 17.4% (2.6%–37.5%) in Tanzania from 2010 to 2020. In central Africa, the levels increased by 28.9% (7.2%–62.5%) in Equatorial Guinea and 85.3% (54.0%–95.9%) in the Congo from 2000 to 2020, while in the other countries remained largely unchanged. In western Africa, the levels have remained low from 2000 to 2020, except for Nigeria, with a reduction of 14.4% (0.7%–67.5%) and Mali, with an increase of 7.0% (0.8%–25.6%). High-level SP resistance increased by 5.5% (1.0%–20.0%) in Malawi, 4.7% (0.5%–25.4%) in Kenya and 2.0% (0.1%–39.2%) in Tanzania, from 2000 to 2020. Conclusion Under the WHO protocols, SP is no longer effective for intermittent preventive treatment in pregnancy and infancy in most of eastern Africa and parts of central Africa. Strengthening health systems capacity to monitor drug resistance at subnational levels across the endemicity spectrum is critical to achieve the global target to end the epidemic.
Collapse
Affiliation(s)
- Floriano Amimo
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan .,Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Ben Lambert
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Anthony Magit
- Human Research Protection Program, University of California San Diego School of Medicine, University of California System, San Diego, California, USA
| | - Jahit Sacarlal
- Faculty of Medicine, Eduardo Mondlane University, Maputo, Mozambique
| | - Masahiro Hashizume
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kenji Shibuya
- Department of Global Health Policy, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Institute for Population Health, King's College London, London, UK
| |
Collapse
|
66
|
Mills M, Holley SL, Coly P, DeJoy S. Malaria in Pregnancy: Considerations for Health Care Providers in Nonendemic Countries. J Midwifery Womens Health 2021; 66:343-350. [PMID: 34166575 DOI: 10.1111/jmwh.13258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 11/29/2022]
Abstract
Malaria is a common infection world-wide, which carries significant risk of morbidity and mortality. Health care providers in the United States may lack experience in recognizing and treating this disease. The pathophysiology of malaria differs during pregnancy, resulting in increased risk for serious morbidity and mortality for the woman and her fetus. Screening for risk factors, especially immigration from and travel to endemic countries, is critical. Symptoms of malaria can mimic influenza-type illnesses, causing delay in diagnosis. Consultation with an infectious disease specialist and hospitalization may be required for appropriate testing and treatment. Chemoprophylaxis and counseling regarding methods to reduce risk are important components of prevention. The US Centers for Disease Control and Prevention and the World Health Organization have established protocols for treatment and are helpful resources for clinicians. A team approach to care based on the woman's stage of illness and recovery, can involve midwives, physicians, specialists and others.
Collapse
Affiliation(s)
- Michelle Mills
- Department of Midwifery, Baystate OB/GYN Group, Inc., Springfield, Massachusetts
| | - Sharon L Holley
- Program Director, Nurse-Midwifery Specialty Master's Program, University of Alabama School of Nursing, Birmingham, Alabama
| | - Paul Coly
- Division of Developmental Services, Department of Human Services, Windsor, Connecticut
| | - Susan DeJoy
- Division of Midwifery and Community Health, Department of Obstetrics and Gynecology, Baystate Medical Center, Springfield, Massachusetts
| |
Collapse
|
67
|
Cunningham CH, Hennelly CM, Lin JT, Ubalee R, Boyce RM, Mulogo EM, Hathaway N, Thwai KL, Phanzu F, Kalonji A, Mwandagalirwa K, Tshefu A, Juliano JJ, Parr JB. A novel CRISPR-based malaria diagnostic capable of Plasmodium detection, species differentiation, and drug-resistance genotyping. EBioMedicine 2021; 68:103415. [PMID: 34139428 PMCID: PMC8213918 DOI: 10.1016/j.ebiom.2021.103415] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND CRISPR-based diagnostics are a new class of highly sensitive and specific assays with multiple applications in infectious disease diagnosis. SHERLOCK, or Specific High-Sensitivity Enzymatic Reporter UnLOCKing, is one such CRISPR-based diagnostic that combines recombinase polymerase pre-amplification, CRISPR-RNA base-pairing, and LwCas13a activity for nucleic acid detection. METHODS We developed SHERLOCK assays capable of detecting all Plasmodium species known to cause human malaria and species-specific detection of P. vivax and P. falciparum, the species responsible for the majority of malaria cases worldwide. We further tested these assays using a diverse panel of clinical samples from the Democratic Republic of the Congo, Uganda, and Thailand and pools of Anopheles mosquitoes from Thailand. In addition, we developed a prototype SHERLOCK assay capable of detecting the dihydropteroate synthetase (dhps) single nucleotide variant A581G associated with P. falciparum sulfadoxine resistance. FINDINGS The suite of Plasmodium assays achieved analytical sensitivities ranging from 2•5-18•8 parasites per reaction when tested against laboratory strain genomic DNA. When compared to real-time PCR, the P. falciparum assay achieved 94% sensitivity and 94% specificity during testing of 123 clinical samples. Compared to amplicon-based deep sequencing, the dhps SHERLOCK assay achieved 73% sensitivity and 100% specificity when applied to a panel of 43 clinical samples, with false-negative calls only at lower parasite densities. INTERPRETATION These novel SHERLOCK assays demonstrate the versatility of CRISPR-based diagnostics and their potential as a new generation of molecular tools for malaria diagnosis and surveillance. FUNDING National Institutes of Health (T32GM007092, R21AI148579, K24AI134990, R01AI121558, UL1TR002489, P30CA016086).
Collapse
Affiliation(s)
- Clark H Cunningham
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | | | - Jessica T Lin
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ratawan Ubalee
- Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Ross M Boyce
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Mbarara University of Science and Technology, Mbarara, Uganda
| | - Edgar M Mulogo
- Mbarara University of Science and Technology, Mbarara, Uganda
| | - Nicholas Hathaway
- University of Massachusetts School of Medicine, Worcester, MA, United States
| | - Kyaw L Thwai
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Fernandine Phanzu
- SANRU ASBL (Global Fund), Kinshasa, Democratic Republic of the Congo
| | - Albert Kalonji
- SANRU ASBL (Global Fund), Kinshasa, Democratic Republic of the Congo
| | | | - Antoinette Tshefu
- Kinshasa School of Public Health, Kinshasa, Democratic Republic of the Congo
| | - Jonathan J Juliano
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jonathan B Parr
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
68
|
Faye SLB, Lugand MM. Participatory research for the development of information, education and communication tools to promote intermittent preventive treatment of malaria in pregnancy in the Democratic Republic of the Congo, Nigeria and Mozambique. Malar J 2021; 20:223. [PMID: 34011371 PMCID: PMC8136127 DOI: 10.1186/s12936-021-03765-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To improve the coverage of intermittent preventive treatment of malaria in pregnancy (IPTp) in Africa, Medicines for Malaria Venture (MMV) developed, tested and validated a new packaging of sulfadoxine-pyrimethamine (SP), as well as specific communications tools designed to improve knowledge of IPTp and the motivation of women to adhere to it, particularly if it is distributed by community health workers (CHW). METHODS This article describes and analyses the results of an empirical research carried out in the Democratic Republic of the Congo (DRC), Nigeria and Mozambique, to evaluate the perception and social acceptability of SP for healthcare providers, CHW and pregnant women, and to assess the ability of the new SP packaging and the communications tools to change their perception of SP and improve their attitudes towards IPTp. RESULTS The results indicate that SP's new individual packaging was perceived by pregnant women and healthcare providers as a "hygienic" and "safe", with a specific identity. The graphics used in IPTp communications tools were modified according to the respondents' feedback to make them more culturally and socially sensitive, and then validated. However, although the new blister packaging and IPTp communications tools generated greater confidence and motivation, SP side effects as well as preconceived ideas, particularly regarding its efficacy, remain a challenge that must be addressed to improve IPTp acceptance and compliance by healthcare providers and pregnant women. CONCLUSION This participatory approach to social research based on ongoing feedback to the graphic designer provided more empirical evidence to improve and adapt the textual and visual content of communication tools (SP blister packaging, leaflet, user guide) to local contexts and user preferences. Tested and validated in different socio-cultural and socio-political contexts, these tools provide a good basis for the promotion of IPTp in Africa.
Collapse
Affiliation(s)
- Sylvain Landry Birane Faye
- Laboratoire de Sociologie, Anthropologie, Psychologie (LASAP), Department of Sociology, Cheikh Anta DIOP University (UCAD), Dakar, Senegal.
| | - Maud Majeres Lugand
- Social Research Manager, Access and Product Management, Medicines for Malaria Venture, Geneva, Switzerland
| |
Collapse
|
69
|
Rouamba T, Samadoulougou S, Ouédraogo M, Hien H, Tinto H, Kirakoya-Samadoulougou F. Asymptomatic malaria and anaemia among pregnant women during high and low malaria transmission seasons in Burkina Faso: household-based cross-sectional surveys in Burkina Faso, 2013 and 2017. Malar J 2021; 20:211. [PMID: 33933072 PMCID: PMC8088076 DOI: 10.1186/s12936-021-03703-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/15/2021] [Indexed: 11/29/2022] Open
Abstract
Background Malaria in endemic countries is often asymptomatic during pregnancy, but it has substantial consequences for both the mother and her unborn baby. During pregnancy, anaemia is an important consequence of malaria infection. In Burkina Faso, the intensity of malaria varies according to the season, albeit the prevalence of malaria and anaemia as well as their risk factors, during high and low malaria transmission seasons is underexplored at the household level. Methods Data of 1751 pregnant women from October 2013 to March 2014 and 1931 pregnant women from April 2017 to June 2017 were drawn from two cross-sectional household surveys conducted in 24 health districts of Burkina Faso. Pregnant women were tested for malaria in their household after consenting. Asymptomatic carriage was defined as a positive result from malaria rapid diagnostic tests in the absence of clinical symptoms of malaria. Anaemia was defined as haemoglobin level less than 11 g/dL in the first and third trimester and less than 10.5 g/dL in the second trimester of pregnancy. Results Prevalence of asymptomatic malaria in pregnancy was estimated at 23.9% (95% CI 20.2–28.0) during the high transmission season (October–November) in 2013. During the low transmission season, it was 12.7% (95% CI 10.9–14.7) between December and March in 2013–2014 and halved (6.4%; 95% CI 5.3–7.6) between April and June 2017. Anaemia prevalence was estimated at 59.4% (95% CI 54.8–63.8) during the high transmission season in 2013. During the low transmission season, it was 50.6% (95% CI 47.7–53.4) between December and March 2013–2014 and 65.0% (95% CI 62.8–67.2) between April and June, 2017. Conclusion This study revealed that the prevalence of malaria asymptomatic carriage and anaemia among pregnant women at the community level remain high throughout the year. Thus, more efforts are needed to increase prevention measures such as IPTp–SP coverage in order to reduce anaemia and contribute to preventing low birth weight and poor pregnancy outcomes.
Collapse
Affiliation(s)
- Toussaint Rouamba
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de La Santé, Centre National de La Recherche Scientifique Et Technologique, 42, Avenue Kumda-Yoore, BP 218 Ouagadougou CMS 11, Ouagadougou, Burkina Faso. .,Centre de Recherche en Epidémiologie, Biostatistique Et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Bruxelles, Belgium.
| | - Sékou Samadoulougou
- Evaluation Platform On Obesity Prevention, Quebec Heart and Lung Institute, Quebec, G1V 4G5, Canada.,Centre for Research On Planning and Development (CRAD), Laval University, Quebec, G1V 0A6, Canada
| | - Mady Ouédraogo
- Centre de Recherche en Epidémiologie, Biostatistique Et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Bruxelles, Belgium.,Institut National de La Statistique Et de La Démographie [INSD], 01 BP 374, Ouagadougou 01, Ouagadougou, Burkina Faso
| | - Hervé Hien
- Intitut National de Santé Publique (INSP), Centre Muraz, Bobo-Dioulasso, Burkina Faso.,Institut de Recherche en Sciences de La Santé, Centre National de La Recherche Scientifique Et Technologique, 42, Avenue Kumda-Yoore, BP 218 Ouagadougou CMS 11, Ouagadougou, Burkina Faso
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de La Santé, Centre National de La Recherche Scientifique Et Technologique, 42, Avenue Kumda-Yoore, BP 218 Ouagadougou CMS 11, Ouagadougou, Burkina Faso
| | - Fati Kirakoya-Samadoulougou
- Centre de Recherche en Epidémiologie, Biostatistique Et Recherche Clinique, Ecole de Santé Publique, Université Libre de Bruxelles (ULB), Route de Lennik, 808, 1070, Bruxelles, Belgium
| |
Collapse
|
70
|
Park JJH, Ford N, Xavier D, Ashorn P, Grais RF, Bhutta ZA, Goossens H, Thorlund K, Socias ME, Mills EJ. Randomised trials at the level of the individual. Lancet Glob Health 2021; 9:e691-e700. [PMID: 33865474 DOI: 10.1016/s2214-109x(20)30540-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/31/2022]
Abstract
In global health research, short-term, small-scale clinical trials with fixed, two-arm trial designs that generally do not allow for major changes throughout the trial are the most common study design. Building on the introductory paper of this Series, this paper discusses data-driven approaches to clinical trial research across several adaptive trial designs, as well as the master protocol framework that can help to harmonise clinical trial research efforts in global health research. We provide a general framework for more efficient trial research, and we discuss the importance of considering different study designs in the planning stage with statistical simulations. We conclude this second Series paper by discussing the methodological and operational complexity of adaptive trial designs and master protocols and the current funding challenges that could limit uptake of these approaches in global health research.
Collapse
Affiliation(s)
- Jay J H Park
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nathan Ford
- Centre for Infectious Disease Epidemiology and Research, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Denis Xavier
- Department of Pharmacology and Divison of Clinical Research, St John's Medical College, Bangalore, India
| | - Per Ashorn
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Zulfiqar A Bhutta
- Centre for Global Child Health, Hospital for Sick Children, Toronto, ON, Canada; Institute of Global Health and Development, and Centre of Excellence in Women and Child Health, Aga Khan University, Karachi, Pakistan
| | - Herman Goossens
- Laboratory of Medical Microbiology, University of Antwerp, Antwerp, Belgium
| | - Kristian Thorlund
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Maria Eugenia Socias
- Fundación Huésped, Buenos Aires, Argentina; British Columbia Centre for Substance Use, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edward J Mills
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada; School of Public Health, University of Rwanda, Kigali, Rwanda; Cytel, Vancouver, BC, Canada.
| |
Collapse
|
71
|
Mlugu EM, Minzi O, Kamuhabwa AAR, Aklillu E. Effectiveness of Intermittent Preventive Treatment With Dihydroartemisinin-Piperaqunine Against Malaria in Pregnancy in Tanzania: A Randomized Controlled Trial. Clin Pharmacol Ther 2021; 110:1478-1489. [PMID: 33891721 DOI: 10.1002/cpt.2273] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/30/2021] [Indexed: 11/10/2022]
Abstract
Intermittent preventive treatment in pregnancy with sulfadoxine-pyrimethamine (IPTp-SP) to prevent malaria and adverse birth outcomes is threatened by Plasmodium falciparum resistance to sulfadoxine-pyrimethamine. We investigated the effectiveness of intermittent preventive treatment in pregnancy with monthly dihydroartemisinin-piperaquine (IPTp-DHP) as an alternative option to IPTp-SP. A total of 956 malaria-free (malaria rapid diagnostic test (MRDT) negative) pregnant women from moderate malaria transmission areas in Tanzania were enrolled and randomized to receive monthly IPTp-DHP (n = 478) or IPTp-SP (n = 478) and followed for maternal and birth outcomes. The primary outcome was the prevalence of histopathologically confirmed placental malaria (active or past infection). Secondary outcomes were overall malaria at delivery, symptomatic-malaria, parasitemia during pregnancy, and adverse birth outcomes as a composite of spontaneous-abortion, premature birth, stillbirth, and low birth weight (LBW) fetal anemia. Outcome differences between treatment groups were expressed as the protective efficacy (PE), defined as 1-prevalence ratios or 1-incidence rate ratio. The prevalence of histopathologically confirmed placental malaria was significantly lower in IPTp-DHP (2.5%, 12/478) than IPTp-SP (8.2%, 39/478); PE = 69% (95% confidence interval (CI): 42-84, P < 0.001). The prevalence of maternal malaria at delivery was significantly lower in IPTp-DHP (8.2%) than IPTp-SP (18.2%, P < 0.001). The incidence per person-years at risk for symptomatic-malaria (0.02 vs. 0.12, P = 0.002) and parasitemia during pregnancy (0.28 vs. 0.67, P < 0.001) were significantly lower in the IPTp-DHP group than in the IPTp-SP group. The prevalence of any adverse birth outcomes (composite) was not significantly (P = 0.06) different between IPTp-DHP (17.9%) and IPTp-SP (23.8%). However, the prevalence of LBW (4.6% vs. 9.6%, P = 0.003) was significantly lower in IPTp-DHP compared with IPTp-SP. We report superior protective efficacy of monthly IPTp-DHP against malaria in pregnancy and LBW than IPTp-SP.
Collapse
Affiliation(s)
- Eulambius M Mlugu
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden.,Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Omary Minzi
- Department of Clinical Pharmacy and Pharmacology, 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
- Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
72
|
Pessanha de Carvalho L, Kreidenweiss A, Held J. Drug Repurposing: A Review of Old and New Antibiotics for the Treatment of Malaria: Identifying Antibiotics with a Fast Onset of Antiplasmodial Action. Molecules 2021; 26:2304. [PMID: 33921170 PMCID: PMC8071546 DOI: 10.3390/molecules26082304] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria is one of the most life-threatening infectious diseases and constitutes a major health problem, especially in Africa. Although artemisinin combination therapies remain efficacious to treat malaria, the emergence of resistant parasites emphasizes the urgent need of new alternative chemotherapies. One strategy is the repurposing of existing drugs. Herein, we reviewed the antimalarial effects of marketed antibiotics, and described in detail the fast-acting antibiotics that showed activity in nanomolar concentrations. Antibiotics have been used for prophylaxis and treatment of malaria for many years and are of particular interest because they might exert a different mode of action than current antimalarials, and can be used simultaneously to treat concomitant bacterial infections.
Collapse
Affiliation(s)
- Lais Pessanha de Carvalho
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
- Centre de Recherches Medicales de Lambaréné (CERMEL), Lambaréné BP 242, Gabon
| | - Jana Held
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
- Centre de Recherches Medicales de Lambaréné (CERMEL), Lambaréné BP 242, Gabon
| |
Collapse
|
73
|
Slater L, Betson M, Ashraf S, Sargison N, Chaudhry U. Current methods for the detection of antimalarial drug resistance in Plasmodium parasites infecting humans. Acta Trop 2021; 216:105828. [PMID: 33465353 DOI: 10.1016/j.actatropica.2021.105828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 12/19/2022]
Abstract
Malaria is the world's deadliest parasitic disease. Great progress has been made in the fight against malaria over the past two decades, but this has recently begun to plateau, in part due to the global development of antimalarial drug resistance. The ability to track drug resistance is necessary to achieve progress in treatment, disease surveillance and epidemiology, which has prompted the development of advanced diagnostic methods. These new methods provide unprecedented access to information that can help to guide public health policies. Development of new technologies increases the potential for high throughput and reduced costs of diagnostic tests; improving the accessibility of tools to investigate the forces driving disease dynamics and, ultimately, clinical outcomes for malaria patients and public health. This literature review provides a summary of the methods currently available for the detection of antimalarial drug resistance from the examination of patients' blood samples. While no single method is perfect for every application, many of the newly developed methods give promise for more reliable and efficient characterisation of Plasmodium resistance in a range of settings. By exploiting the strengths of the tools available, we can develop a deeper understanding of the evolutionary and spatiotemporal dynamics of this disease. This will translate into more effective disease control, better-informed policy, and more timely and successful treatment for malaria patients.
Collapse
|
74
|
Sibley CH. Prevalence of Antimalarial Resistance Mediators. J Infect Dis 2021; 223:927-929. [PMID: 33146721 DOI: 10.1093/infdis/jiaa688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Carol Hopkins Sibley
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| |
Collapse
|
75
|
Chua CLL, Hasang W, Rogerson SJ, Teo A. Poor Birth Outcomes in Malaria in Pregnancy: Recent Insights Into Mechanisms and Prevention Approaches. Front Immunol 2021; 12:621382. [PMID: 33790894 PMCID: PMC8005559 DOI: 10.3389/fimmu.2021.621382] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
Pregnant women in malaria-endemic regions are susceptible to malaria in pregnancy, which has adverse consequences on birth outcomes, including having small for gestational age and preterm babies. These babies are likely to have low birthweights, which predisposes to infant mortality and lifelong morbidities. During malaria in pregnancy, Plasmodium falciparum-infected erythrocytes express a unique variant surface antigen, VAR2CSA, that mediates sequestration in the placenta. This process may initiate a range of host responses that contribute to placental inflammation and dysregulated placental development, which affects placental vasculogenesis, angiogenesis and nutrient transport. Collectively, these result in the impairment of placental functions, affecting fetal development. In this review, we provide an overview of malaria in pregnancy and the different pathological pathways leading to malaria in pregnancy-associated low birthweight. We also discuss current prevention and management strategies for malaria in pregnancy, and some potential therapeutic interventions that may improve birth outcomes. Lastly, we outline some priorities for future research that could bring us one step closer to reducing this health burden.
Collapse
Affiliation(s)
| | - Wina Hasang
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen J Rogerson
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Teo
- Department of Medicine at Royal Melbourne Hospital, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
76
|
Ippolito MM, Moser KA, Kabuya JBB, Cunningham C, Juliano JJ. Antimalarial Drug Resistance and Implications for the WHO Global Technical Strategy. CURR EPIDEMIOL REP 2021; 8:46-62. [PMID: 33747712 PMCID: PMC7955901 DOI: 10.1007/s40471-021-00266-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Five years have passed since the World Health Organization released its Global Technical Strategy for Malaria (GTS). In that time, progress against malaria has plateaued. This review focuses on the implications of antimalarial drug resistance for the GTS and how interim progress in parasite genomics and antimalarial pharmacology offer a bulwark against it. RECENT FINDINGS For the first time, drug resistance-conferring genes have been identified and validated before their global expansion in malaria parasite populations. More efficient methods for their detection and elaboration have been developed, although low-density infections and polyclonality remain a nuisance to be solved. Clinical trials of alternative regimens for multidrug-resistant malaria have delivered promising results. New agents continue down the development pipeline, while a nascent infrastructure in sub-Saharan Africa for conducting phase I trials and trials of transmission-blocking agents has come to fruition after years of preparation. SUMMARY These and other developments can help inform the GTS as the world looks ahead to the next two decades of its implementation. To remain ahead of the threat that drug resistance poses, wider application of genomic-based surveillance and optimization of existing and forthcoming antimalarial drugs are essential.
Collapse
Affiliation(s)
- Matthew M. Ippolito
- Divisions of Clinical Pharmacology and Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
- The Johns Hopkins Malaria Research Institute, Johns Hopkins University School of Public Health, Baltimore, MD USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Kara A. Moser
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, NC USA
| | | | - Clark Cunningham
- School of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Jonathan J. Juliano
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of North Carolina, CB#7030, 130 Mason Farm Rd, Chapel Hill, NC 27599 USA
- Curriculum in Genetics and Molecular Biology, 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
|
77
|
Hansson H, Minja DTR, Moeller SL, Lusingu JPA, Bygbjerg IC, Yde AM, Jensen RW, Nag S, Msemo OA, Theander TG, Alifrangis M, Schmiegelow C. Reduced birth weight caused by sextuple drug resistant Plasmodium falciparum infection in early 2nd trimester. J Infect Dis 2021; 224:1605-1613. [PMID: 33684211 DOI: 10.1093/infdis/jiab117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/26/2021] [Indexed: 11/12/2022] Open
Abstract
Mutations in the Plasmodium falciparum genes Pfdhfr and Pfdhps, particularly the sextuple mutant haplotype threatens the antimalarial effectiveness of sulfadoxine-pyrimethamine as intermittent preventive treatment during pregnancy (IPTp). To explore the impact of sextuple mutant haplotype infections on outcome measures after provision of IPTp-SP, we monitored birth outcomes in women followed from prior to conception or from the first trimester until delivery. Women infected with sextuple haplotypes in early 2 nd trimester specifically, delivered newborns with a lower birth weight (-267g, 95% CI -454; -59, p=0·01) compared to women who did not have malaria during pregnancy and women infected with less SP resistant haplotypes (-461g, 95% CI -877; -44, p=0·03). Thus, sextuple haplotype infections seems to impact the effectiveness of SP for IPTp and directly impact birth outcome by lowering birth weight. Close monitoring and targeted malaria control during early pregnancy is therefore crucial to improve birth outcomes.
Collapse
Affiliation(s)
- Helle Hansson
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Daniel T R Minja
- National Institute for Medical Research, Tanga Research Centre, Bombo Area, Tanga, Tanzania
| | - Sofie L Moeller
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark.,Global Health Section, Department of Public Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - John P A Lusingu
- National Institute for Medical Research, Tanga Research Centre, Bombo Area, Tanga, Tanzania
| | - Ib C Bygbjerg
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark.,Global Health Section, Department of Public Health, University of Copenhagen, Øster Farimagsgade, Copenhagen K, Denmark
| | - Anna-Mathilde Yde
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Rasmus W Jensen
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Sidsel Nag
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Omari A Msemo
- National Institute for Medical Research, Tanga Research Centre, Bombo Area, Tanga, Tanzania
| | - Thor G Theander
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Michael Alifrangis
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| | - Christentze Schmiegelow
- Department of Immunology and Microbiology, University of Copenhagen, Denmark and Department of Infectious Diseases, Copenhagen University Hospital, Blegdamsvej, Copenhagen N, Denmark
| |
Collapse
|
78
|
Gamain B, Chêne A, Viebig NK, Tuikue Ndam N, Nielsen MA. Progress and Insights Toward an Effective Placental Malaria Vaccine. Front Immunol 2021; 12:634508. [PMID: 33717176 PMCID: PMC7947914 DOI: 10.3389/fimmu.2021.634508] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/06/2021] [Indexed: 12/03/2022] Open
Abstract
In areas where Plasmodium falciparum transmission is endemic, clinical immunity against malaria is progressively acquired during childhood and adults are usually protected against the severe clinical consequences of the disease. Nevertheless, pregnant women, notably during their first pregnancies, are susceptible to placental malaria and the associated serious clinical outcomes. Placental malaria is characterized by the massive accumulation of P. falciparum infected erythrocytes and monocytes in the placental intervillous spaces leading to maternal anaemia, hypertension, stillbirth and low birth weight due to premature delivery, and foetal growth retardation. Remarkably, the prevalence of placental malaria sharply decreases with successive pregnancies. This protection is associated with the development of antibodies directed towards the surface of P. falciparum-infected erythrocytes from placental origin. Placental sequestration is mediated by the interaction between VAR2CSA, a member of the P. falciparum erythrocyte membrane protein 1 family expressed on the infected erythrocytes surface, and the placental receptor chondroitin sulfate A. VAR2CSA stands today as the leading candidate for a placental malaria vaccine. We recently reported the safety and immunogenicity of two VAR2CSA-derived placental malaria vaccines (PRIMVAC and PAMVAC), spanning the chondroitin sulfate A-binding region of VAR2CSA, in both malaria-naïve and P. falciparum-exposed non-pregnant women in two distinct Phase I clinical trials (ClinicalTrials.gov, NCT02658253 and NCT02647489). This review discusses recent advances in placental malaria vaccine development, with a focus on the recent clinical data, and discusses the next clinical steps to undertake in order to better comprehend vaccine-induced immunity and accelerate vaccine development.
Collapse
Affiliation(s)
- Benoît Gamain
- Université de Paris, Inserm, Biologie Intégrée du Globule Rouge, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France
| | - Arnaud Chêne
- Université de Paris, Inserm, Biologie Intégrée du Globule Rouge, Paris, France.,Institut National de la Transfusion Sanguine, Paris, France
| | - Nicola K Viebig
- European Vaccine Initiative, UniversitätsKlinikum Heidelberg, Heidelberg, Germany
| | | | - Morten A Nielsen
- Centre for Medical Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
79
|
Taylor SM, Levitt B, Freedman B, Madanitsa M, Thwai KL, Kalilani-Phiri L, Khairallah C, Mwapasa V, Ter Kuile FO, Meshnick SR. Interactions Between Antenatal Sulfadoxine-Pyrimethamine, Drug-Resistant Plasmodium falciparum Parasites, and Delivery Outcomes in Malawi. J Infect Dis 2021; 222:661-669. [PMID: 32221555 DOI: 10.1093/infdis/jiaa145] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/25/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Sulfadoxine-pyrimethamine (SP) is used as intermittent preventive therapy in pregnancy (IPTp) for malaria in sub-Saharan Africa. The resistance marker dhps A581G has been associated with reduced IPTp-SP efficacy and enhanced morbidity in SP recipients. METHODS We measured SP-resistance allele frequencies in Malawian women participating in a trial comparing IPTp with SP against intermittent screening by rapid diagnostic tests (ISTp). We genotyped polymerase chain reaction-detected parasites using deep sequencing of SP-resistance alleles. RESULTS Among 125 placental infections, A581G-bearing parasites were associated with reduced birth weight (mean difference [MD], 252 g; 95% confidence interval [CI], 46-457; P = .017). Relative to ISTp, IPTp-SP was associated with higher birth weights in women with wild-type parasites (MD, 116 g; 95% CI, -40 to 272; P = .142) and lower birth weights in women with A581G-bearing parasites (MD, 192 g; 95% CI, -264 to 648; P = .385) (Pinteraction = .033). Similar associations were noted on gestational age (Pinteraction = .075). Amongst only IPTp-SP recipients, relative to women who last received SP > 4 weeks before delivery, recent SP receipt was associated with lower birth weight in women with wild-type parasites (MD, 118 g; 95% CI, -376 to 139; P = .361) and higher birth weight in women with A581G-bearing parasites (MD, 783 g; 95% CI, -20 to 1586; P = .054) (Pinteraction = .005). CONCLUSIONS The effectiveness in birth weight of IPTp-SP is compromised by A581G-bearing parasites, but there was no evidence that the adverse effects of these parasites are exacerbated by antenatal SP. ISRCTN REGISTRY www.isrctn.com/ISRCTN69800930.
Collapse
Affiliation(s)
- Steve M Taylor
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina, USA.,Duke Global Health Institute, Durham, North Carolina, USA.,Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brandt Levitt
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Betsy Freedman
- Division of Infectious Diseases, Duke University Medical Center, Durham, North Carolina, USA
| | - Mwayiwawo Madanitsa
- Department of Public Health, School of Public Health and Family Medicine, College of Medicine, Blantyre, Malawi.,Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kyaw-Lay Thwai
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Linda Kalilani-Phiri
- Department of Public Health, School of Public Health and Family Medicine, College of Medicine, Blantyre, Malawi
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Victor Mwapasa
- Department of Public Health, School of Public Health and Family Medicine, College of Medicine, Blantyre, Malawi
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
80
|
Al Khaja KAJ, Sequeira RP. Drug treatment and prevention of malaria in pregnancy: a critical review of the guidelines. Malar J 2021; 20:62. [PMID: 33485330 PMCID: PMC7825227 DOI: 10.1186/s12936-020-03565-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022] Open
Abstract
Background Malaria caused by Plasmodium falciparum in pregnancy can result in adverse maternal and fetal sequelae. This review evaluated the adherence of the national guidelines drawn from World Health Organization (WHO) regions, Africa, Eastern Mediterranean, Southeast Asia, and Western Pacific, to the WHO recommendations on drug treatment and prevention of chloroquine-resistant falciparum malaria in pregnant women. Methods Thirty-five updated national guidelines and the President’s Malaria Initiative (PMI), available in English language, were reviewed. The primary outcome measures were the first-line anti-malarial treatment protocols adopted by national guidelines for uncomplicated and complicated falciparum malaria infections in early (first) and late (second and third) trimesters of pregnancy. The strategy of intermittent preventive treatment of malaria in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) was also addressed. Results This review evaluated the treatment and prevention of falciparum malaria in pregnancy in 35 national guidelines/PMI-Malaria Operational Plans (MOP) reports out of 95 malaria-endemic countries. Of the 35 national guidelines, 10 (28.6%) recommend oral quinine plus clindamycin as first-line treatment for uncomplicated malaria in the first trimester. As the first-line option, artemether–lumefantrine, an artemisinin-based combination therapy, is adopted by 26 (74.3%) of the guidelines for treating uncomplicated or complicated malaria in the second and third trimesters. Intravenous artesunate is approved by 18 (51.4%) and 31 (88.6%) guidelines for treating complicated malaria during early and late pregnancy, respectively. Of the 23 national guidelines that recommend IPTp-SP strategy, 8 (34.8%) are not explicit about directly observed therapy requirements, and three-quarters, 17 (73.9%), do not specify contra-indication of SP in human immunodeficiency virus (HIV)-infected pregnant women receiving cotrimoxazole prophylaxis. Most of the guidelines (18/23; 78.3%) state the recommended folic acid dose. Conclusion Several national guidelines and PMI reports require update revisions to harmonize with international guidelines and emergent trends in managing falciparum malaria in pregnancy. National guidelines and those of donor agencies should comply with those of WHO guideline recommendations although local conditions and delayed guideline updates may call for deviations from WHO evidence-based guidelines.
Collapse
Affiliation(s)
- Khalid A J Al Khaja
- Department of Pharmacology & Therapeutics, College of Medicine & Medical Sciences, Arabian Gulf University, P.O. Box 22979, Manama, Kingdom of Bahrain.
| | - Reginald P Sequeira
- Department of Pharmacology & Therapeutics, College of Medicine & Medical Sciences, Arabian Gulf University, P.O. Box 22979, Manama, Kingdom of Bahrain
| |
Collapse
|
81
|
Kwambai TK, Dhabangi A, Idro R, Opoka R, Watson V, Kariuki S, Kuya NA, Onyango ED, Otieno K, Samuels AM, Desai MR, Boele van Hensbroek M, Wang D, John CC, Robberstad B, Phiri KS, Ter Kuile FO. Malaria Chemoprevention in the Postdischarge Management of Severe Anemia. N Engl J Med 2020; 383:2242-2254. [PMID: 33264546 PMCID: PMC9115866 DOI: 10.1056/nejmoa2002820] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Children who have been hospitalized with severe anemia in areas of Africa in which malaria is endemic have a high risk of readmission and death within 6 months after discharge. No prevention strategy specifically addresses this period. METHODS We conducted a multicenter, two-group, randomized, placebo-controlled trial in nine hospitals in Kenya and Uganda to determine whether 3 months of malaria chemoprevention could reduce morbidity and mortality after hospital discharge in children younger than 5 years of age who had been admitted with severe anemia. All children received standard in-hospital care for severe anemia and a 3-day course of artemether-lumefantrine at discharge. Two weeks after discharge, children were randomly assigned to receive dihydroartemisinin-piperaquine (chemoprevention group) or placebo, administered as 3-day courses at 2, 6, and 10 weeks after discharge. Children were followed for 26 weeks after discharge. The primary outcome was one or more hospital readmissions for any reason or death from the time of randomization to 6 months after discharge. Conditional risk-set modeling for recurrent events was used to calculate hazard ratios with the use of the Prentice-Williams-Peterson total-time approach. RESULTS From May 2016 through May 2018, a total of 1049 children underwent randomization; 524 were assigned to the chemoprevention group and 525 to the placebo group. From week 3 through week 26, a total of 184 events of readmission or death occurred in the chemoprevention group and 316 occurred in the placebo group (hazard ratio, 0.65; 95% confidence interval [CI], 0.54 to 0.78; P<0.001). The lower incidence of readmission or death in the chemoprevention group than in the placebo group was restricted to the intervention period (week 3 through week 14) (hazard ratio, 0.30; 95% CI, 0.22 to 0.42) and was not sustained after that time (week 15 through week 26) (hazard ratio, 1.13; 95% CI, 0.87 to 1.47). No serious adverse events were attributed to dihydroartemisinin-piperaquine. CONCLUSIONS In areas with intense malaria transmission, 3 months of postdischarge malaria chemoprevention with monthly dihydroartemisinin-piperaquine in children who had recently received treatment for severe anemia prevented more deaths or readmissions for any reason after discharge than placebo. (Funded by the Research Council of Norway and the Centers for Disease Control and Prevention; ClinicalTrials.gov number, NCT02671175.).
Collapse
Affiliation(s)
- Titus K Kwambai
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Aggrey Dhabangi
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Richard Idro
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Robert Opoka
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Victoria Watson
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Simon Kariuki
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Nickline A Kuya
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Eric D Onyango
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Kephas Otieno
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Aaron M Samuels
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Meghna R Desai
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Michael Boele van Hensbroek
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Duolao Wang
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Chandy C John
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Bjarne Robberstad
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Kamija S Phiri
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| | - Feiko O Ter Kuile
- From the Centre for Global Health Research, Kenya Medical Research Institute (T.K.K., S.K., N.A.K., E.D.O., K.O., F.O.K.), and the Kisumu County Department of Health, Kenya Ministry of Health (T.K.K.) and the Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention (CDC) (A.M.S., M.R.D.) - all in Kisumu; the Department of Clinical Sciences, Liverpool School of Tropical Medicine (T.K.K., V.W., D.W., F.O.K.), and the Department of Biostatistics, University of Liverpool (V.W.), Liverpool, United Kingdom; Makerere University College of Health Sciences, Kampala, Uganda (A.D., R.I., R.O.); the Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, CDC, Atlanta (A.M.S., M.R.D.); Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam (M.B.H.); the Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis (C.C.J.); the Section for Ethics and Health Economics and the Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway (B.R.); and the School of Public Health and Family Medicine, College of Medicine, University of Malawi, Blantyre (K.S.P.)
| |
Collapse
|
82
|
Turkiewicz A, Manko E, Sutherland CJ, Diez Benavente E, Campino S, Clark TG. Genetic diversity of the Plasmodium falciparum GTP-cyclohydrolase 1, dihydrofolate reductase and dihydropteroate synthetase genes reveals new insights into sulfadoxine-pyrimethamine antimalarial drug resistance. PLoS Genet 2020; 16:e1009268. [PMID: 33382691 PMCID: PMC7774857 DOI: 10.1371/journal.pgen.1009268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Plasmodium falciparum parasites resistant to antimalarial treatments have hindered malaria disease control. Sulfadoxine-pyrimethamine (SP) was used globally as a first-line treatment for malaria after wide-spread resistance to chloroquine emerged and, although replaced by artemisinin combinations, is currently used as intermittent preventive treatment of malaria in pregnancy and in young children as part of seasonal malaria chemoprophylaxis in sub-Saharan Africa. The emergence of SP-resistant parasites has been predominantly driven by cumulative build-up of mutations in the dihydrofolate reductase (pfdhfr) and dihydropteroate synthetase (pfdhps) genes, but additional amplifications in the folate pathway rate-limiting pfgch1 gene and promoter, have recently been described. However, the genetic make-up and prevalence of those amplifications is not fully understood. We analyse the whole genome sequence data of 4,134 P. falciparum isolates across 29 malaria endemic countries, and reveal that the pfgch1 gene and promoter amplifications have at least ten different forms, occurring collectively in 23% and 34% in Southeast Asian and African isolates, respectively. Amplifications are more likely to be present in isolates with a greater accumulation of pfdhfr and pfdhps substitutions (median of 1 additional mutations; P<0.00001), and there was evidence that the frequency of pfgch1 variants may be increasing in some African populations, presumably under the pressure of SP for chemoprophylaxis and anti-folate containing antibiotics used for the treatment of bacterial infections. The selection of P. falciparum with pfgch1 amplifications may enhance the fitness of parasites with pfdhfr and pfdhps substitutions, potentially threatening the efficacy of this regimen for prevention of malaria in vulnerable groups. Our work describes new pfgch1 amplifications that can be used to inform the surveillance of SP drug resistance, its prophylactic use, and future experimental work to understand functional mechanisms.
Collapse
Affiliation(s)
- Anna Turkiewicz
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emilia Manko
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Colin J. Sutherland
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ernest Diez Benavente
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Susana Campino
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taane G. Clark
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
83
|
Tola M, Ajibola O, Idowu ET, Omidiji O, Awolola ST, Amambua-Ngwa A. Molecular detection of drug resistant polymorphisms in Plasmodium falciparum isolates from Southwest, Nigeria. BMC Res Notes 2020; 13:497. [PMID: 33109270 PMCID: PMC7588951 DOI: 10.1186/s13104-020-05334-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/14/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Nigeria bears 25% of global malaria burden despite concerted efforts towards its control and elimination. The emergence of drug resistance to first line drugs, artemisinin combination therapies (ACTs), indicates an urgent need for continuous molecular surveillance of drug resistance especially in high burden countries where drug interventions are heavily relied on. This study describes mutations in Plasmodium falciparum genes associated with drug resistance in malaria; Pfk13, Pfmdr1, PfATPase6 and Pfcrt in isolates obtained from 83 symptomatic malaria patients collected in August 2014, aged 1-61 years old from South-west Nigeria. RESULTS Two Pfmdr1, N86 and Y184 variants were present at a prevalence of 56% and 13.25% of isolates respectively. There was one synonymous (S679S) and two non-synonymous (M699V, S769M) mutations in the PATPase6 gene, while Pfcrt genotype (CVIET), had a prevalence of 45%. The Pfk13 C580Y mutant allele was suspected by allelic discrimination in two samples with mixed genotypes although this could not be validated with independent isolation or additional methods. Our findings call for robust molecular surveillance of antimalarial drug resistance markers in west Africa especially with increased use of antimalarial drugs as prophylaxis for Covid-19.
Collapse
Affiliation(s)
- Monday Tola
- Public Health Division, Nigerian Institute of Medical Research, Lagos, Nigeria.,Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| | - Olumide Ajibola
- First Technical University, Ibadan, Oyo State, Nigeria.,Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | | | - Olusesan Omidiji
- Department of Cell Biology and Genetics, University of Lagos, Lagos, Nigeria
| | | | - Alfred Amambua-Ngwa
- Medical Research Council Unit The Gambia At London, School of Hygiene and Tropical Medicine, Banjul, The Gambia.
| |
Collapse
|
84
|
Saito M, Briand V, Min AM, McGready R. Deleterious effects of malaria in pregnancy on the developing fetus: a review on prevention and treatment with antimalarial drugs. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:761-774. [PMID: 32946830 DOI: 10.1016/s2352-4642(20)30099-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 03/09/2020] [Accepted: 03/25/2020] [Indexed: 12/29/2022]
Abstract
All malaria infections are harmful to both the pregnant mother and the developing fetus. One in ten maternal deaths in malaria endemic countries are estimated to result from Plasmodium falciparum infection. Malaria is associated with a 3-4 times increased risk of miscarriage and a substantially increased risk of stillbirth. Current treatment and prevention strategies reduce, but do not eliminate, malaria's damaging effects on pregnancy outcomes. Reviewing evidence generated from meta-analyses, systematic reviews, and observational data, the first paper in this Series aims to summarise the adverse effects of malaria in pregnancy on the fetus and how the current drug treatment and prevention strategies can alleviate these effects. Although evidence supports the safety and treatment efficacy of artemisinin-based combination therapies in the first trimester, these therapies have not been recommended by WHO for the treatment of malaria at this stage of pregnancy. Intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine is contraindicated in the first trimester and provides imperfect chemoprevention because of inadequate dosing, poor (few and late) antenatal clinic attendance, increasing antimalarial drug resistance, and decreasing naturally acquired maternal immunity due to the decreased incidence of malaria. Alternative strategies to prevent malaria in pregnancy are needed. The prevention of all malaria infections by providing sustained exposure to effective concentrations of antimalarial drugs is key to reducing the adverse effects of malaria in pregnancy.
Collapse
Affiliation(s)
- Makoto Saito
- Division of Infectious Diseases, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Valérie Briand
- Infectious Diseases in Lower Income Countries, Research Institute for Sustainable Development, French National Institute of Health and Medical Research, University of Bordeaux, Bordeaux, France
| | - Aung Myat Min
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
85
|
Huijben S, Macete E, Mombo-Ngoma G, Ramharter M, Kariuki S, Desai M, Shi YP, Mwangoka G, Massougbodji A, Cot M, Ndam NT, Uberegui E, Gupta H, Cisteró P, Aponte JJ, González R, Menéndez C, Mayor A. Counter-Selection of Antimalarial Resistance Polymorphisms by Intermittent Preventive Treatment in Pregnancy. J Infect Dis 2020; 221:293-303. [PMID: 31677349 DOI: 10.1093/infdis/jiz451] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 09/19/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Innovative approaches are needed to limit antimalarial resistance evolution. Understanding the role of intermittent preventive treatment in pregnancy (IPTp) on the selection for resistance and the impact such selection has on pregnancy outcomes can guide future interventions. METHODS Plasmodium falciparum isolates (n = 914) from 2 randomized clinical trials were screened for pfmdr1 copy number variation and pfcrt, pfmdr1, pfdhfr, and pfdhps resistance markers. The trials were conducted between 2010 and 2013 in Benin, Gabon, Kenya, and Mozambique to establish the efficacy of IPTp-mefloquine (MQ) compared with IPTp-sulphadoxine-pyrimethamine (SP) in human immunodeficiency virus (HIV)-uninfected and to IPTp-placebo in HIV-infected women. RESULTS In HIV-uninfected women, the prevalence of pfcrt mutants, pfdhfr/pfdhps quintuple mutants, and pfmdr1 copy number was similar between women receiving IPT-SP and IPTp-MQ. However, prevalence of pfmdr1 polymorphism 86Y was lower in the IPTp-MQ group than in the IPTp-SP group, and within the IPTp-MQ group it was lower at delivery compared with recruitment. No effect of IPTp-MQ on resistance markers was observed among HIV-infected women. The carriage of resistance markers was not associated with pregnancy outcomes. CONCLUSIONS Selection of wild-type pfmdr1 polymorphism N86 by IPTp-MQ highlights the strong selective pressure IPTp can exert and the opportunity for using negative cross-resistance in drug choice for clinical treatment and IPTp.
Collapse
Affiliation(s)
- Silvie Huijben
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | - Eusebio Macete
- Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Ghyslain Mombo-Ngoma
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universität Tübingen, und Deutsches Zentrum für Infektionsforschung, Tübingen, Germany.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and Department of Medicine I, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - 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
| | - Ya Ping Shi
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Achille Massougbodji
- Unité d'Enseignement et de Recherche de Parasitologie Mycologie, Faculté des Sciences de la Santé, Cotonou, Bénin
| | - Michel Cot
- Université de Paris, MERIT, IRD, Paris, France
| | | | | | - Himanshu Gupta
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - Pau Cisteró
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
| | - John J Aponte
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Raquel González
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Clara Menéndez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain.,Centro de Investigação em Saúde da Manhiça, Manhiça, Mozambique
| |
Collapse
|
86
|
Walker PGT, Cairns M, Slater H, Gutman J, Kayentao K, Williams JE, Coulibaly SO, Khairallah C, Taylor S, Meshnick SR, Hill J, Mwapasa V, Kalilani-Phiri L, Bojang K, Kariuki S, Tagbor H, Griffin JT, Madanitsa M, Ghani ACH, Desai M, Ter Kuile FO. Modelling the incremental benefit of introducing malaria screening strategies to antenatal care in Africa. Nat Commun 2020; 11:3799. [PMID: 32732892 PMCID: PMC7393377 DOI: 10.1038/s41467-020-17528-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/29/2020] [Indexed: 12/01/2022] Open
Abstract
Plasmodium falciparum in pregnancy is a major cause of adverse pregnancy outcomes. We combine performance estimates of standard rapid diagnostic tests (RDT) from trials of intermittent screening and treatment in pregnancy (ISTp) with modelling to assess whether screening at antenatal visits improves upon current intermittent preventative therapy with sulphadoxine-pyrimethamine (IPTp-SP). We estimate that RDTs in primigravidae at first antenatal visit are substantially more sensitive than in non-pregnant adults (OR = 17.2, 95% Cr.I. 13.8-21.6), and that sensitivity declines in subsequent visits and with gravidity, likely driven by declining susceptibility to placental infection. Monthly ISTp with standard RDTs, even with highly effective drugs, is not superior to monthly IPTp-SP. However, a hybrid strategy, recently adopted in Tanzania, combining testing and treatment at first visit with IPTp-SP may offer benefit, especially in areas with high-grade SP resistance. Screening and treatment in the first trimester, when IPTp-SP is contraindicated, could substantially improve pregnancy outcomes.
Collapse
Affiliation(s)
- Patrick G T Walker
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK.
| | - Matt Cairns
- London School of Hygiene and Tropical Medicine, London, UK
| | - Hannah Slater
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
- PATH, Seattle, WA, USA
| | - Julie Gutman
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Kassoum Kayentao
- Malaria Research and Training Centre, Department of Epidemiology of Parasitic Diseases, Faculty of Medicine, Pharmacy, and Dentistry, University of Sciences, Techniques, and Technologies of Bamako, Bamako, Mali
| | | | - Sheick O Coulibaly
- Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Steve Taylor
- Global Health Institute, Duke University, Durham, NC, USA
| | | | - Jenny Hill
- Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | | | - Kalifa Bojang
- Medical Research Council, London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Simon Kariuki
- Kenya Medical Research Institute/Centre for Global Health Research, Kisumu, Kenya
| | - Harry Tagbor
- University of Health and Allied Sciences, Ho, Ghana
| | - Jamie T Griffin
- School of Mathematical Sciences, Queen Mary University of London, Mile End Road, London, UK
| | | | - Azra C H Ghani
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, UK
| | - Meghna Desai
- Malaria Branch, Division of Parasitic Diseases and Malaria, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Feiko O Ter Kuile
- Faculty of Health Sciences, University of Ouagadougou, Ouagadougou, Burkina Faso
| |
Collapse
|
87
|
Quan H, Igbasi U, Oyibo W, Omilabu S, Chen SB, Shen HM, Okolie C, Chen JH, Zhou XN. High multiple mutations of Plasmodium falciparum-resistant genotypes to sulphadoxine-pyrimethamine in Lagos, Nigeria. Infect Dis Poverty 2020; 9:91. [PMID: 32653033 PMCID: PMC7353807 DOI: 10.1186/s40249-020-00712-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/01/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Plasmodium falciparum-resistance to sulphadoxine-pyrimethamine (SP) has been largely reported among pregnant women. However, the profile of resistance markers to SP dihydrofolate reductase (dhfr) and dihydropteroate synthase (dhps) in the general population are varied and not frequently monitored. Currently, SP is used as partner drug for artemisinin combination therapy (SP-artesunate) in some sub-Saharan African countries or as a prophylactic drug in intermittent preventive treatment of malaria during pregnancy and infants and in seasonal malaria chemoprevention (SMC). Profiling of P. falciparum-resistant genotypes to SP is dynamic and critical in providing data that would be useful for malaria control programmes. This study assessed the profile of dhfr and dhps genes genotypes among individuals with malaria in Lagos, Nigeria. METHODS Molecular markers of SP resistance were identified by nested PCR and sequenced among malaria positive dried blood spots (DBS) that were collected from individuals attending health facilities from January 2013 to February 2014 and during community surveys from October 2010 to September 2011 across different Local Government Areas of Lagos State, Nigeria. RESULTS A total of 242 and 167 samples were sequenced for dhfr and dhps, respectively. Sequence analysis of dhfr showed that 95.5% (231/242), 96.3% (233/242) and 96.7% (234/242) of the samples had N51I, C59R and S108N mutant alleles, respectively. The prevalence of dhps mutation at codons A437G, A613S, S436A, A581G, I431V and K540E were 95.8% (160/167), 41.9% (70/167), 41.3% (69/167), 31.1% (52/167), 25.1% (42/167), and 1.2% (2/167) respectively. The prevalence of triple mutations (CIRNI) in dhfr was 93.8% and 44.3% for the single dhps haplotype mutation (SGKAA). Partial SP-resistance due to quadruple dhfr-dhps haplotype mutations (CIRNI-SGKAA) and octuple haplotype mutations (CIRNI-VAGKGS) with rate of 42.6% and 22.0%, respectively has been reported. CONCLUSIONS There was increased prevalence in dhfr triple haplotype mutations when compared with previous reports in the same environment but aligned with high prevalence in other locations in Nigeria and other countries in Africa. Also, high prevalence of dhfr and dhps mutant alleles occurred in the study areas in Lagos, Nigeria five to eight years after the introduction of artemisinin combination therapy underscores the need for continuous monitoring.
Collapse
Affiliation(s)
- Hong Quan
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Center for Tropical Diseases, National Centre for International Research on Tropical Diseases, Ministry of Science and Technology, Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025 People’s Republic of China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention⁃Shenzhen Center for Disease Control and Prevention Joint Laboratory for Imported Tropical Disease Control, Shanghai, 200025 People’s Republic of China
| | - Uche Igbasi
- Center for Infectious Diseases Research, Microbiology Department, Nigerian Institute of Medical Research, 6 Edmund Crescent, Yaba, Lagos, Nigeria
| | - Wellington Oyibo
- ANDI Center of Excellence for Malaria Diagnosis, Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Sunday Omilabu
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Shen-Bo Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Center for Tropical Diseases, National Centre for International Research on Tropical Diseases, Ministry of Science and Technology, Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025 People’s Republic of China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention⁃Shenzhen Center for Disease Control and Prevention Joint Laboratory for Imported Tropical Disease Control, Shanghai, 200025 People’s Republic of China
| | - Hai-Mo Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Center for Tropical Diseases, National Centre for International Research on Tropical Diseases, Ministry of Science and Technology, Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025 People’s Republic of China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention⁃Shenzhen Center for Disease Control and Prevention Joint Laboratory for Imported Tropical Disease Control, Shanghai, 200025 People’s Republic of China
| | - Chukwuma Okolie
- Department of Surveying and Geoinformatics, Faculty of Engineering, University of Lagos, Lagos, Nigeria
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Center for Tropical Diseases, National Centre for International Research on Tropical Diseases, Ministry of Science and Technology, Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025 People’s Republic of China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention⁃Shenzhen Center for Disease Control and Prevention Joint Laboratory for Imported Tropical Disease Control, Shanghai, 200025 People’s Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Chinese Center for Tropical Diseases Research, WHO Collaborating Center for Tropical Diseases, National Centre for International Research on Tropical Diseases, Ministry of Science and Technology, Key Laboratory of Parasite and Vector Biology, Ministry of Health, Shanghai, 200025 People’s Republic of China
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention⁃Shenzhen Center for Disease Control and Prevention Joint Laboratory for Imported Tropical Disease Control, Shanghai, 200025 People’s Republic of China
| |
Collapse
|
88
|
Kublin JG, Murphy SC, Maenza J, Seilie AM, Jain JP, Berger D, Spera D, Zhao R, Soon RL, Czartoski JL, Potochnic MA, Duke E, Chang M, Vaughan A, Kappe SHI, Leong FJ, Pertel P, Prince WT. Safety, pharmacokinetics and causal prophylactic efficacy of KAF156 in a Plasmodium falciparum human infection study. Clin Infect Dis 2020; 73:e2407-e2414. [PMID: 32644127 DOI: 10.1093/cid/ciaa952] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/07/2020] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND KAF156 is a novel antimalarial drug that is active against both liver- and blood- stage Plasmodium parasites, including drug-resistant strains. Here, we investigated the causal prophylactic efficacy of KAF156 in a controlled human malaria infection (CHMI) model. METHODS In Part 1, healthy, malaria-naïve participants received 800 mg KAF156 or placebo three hr before CHMI with Pf-infected mosquitoes. In Part 2, KAF156 was administered as single doses of 800, 300, 100, 50, or 20 mg 21 hr post-CHMI. All participants received atovaquone/proguanil treatment if blood-stage infection was detected or on day 29. For each cohort, 7-14 subjects were enrolled to KAF156 treatment and up to four subjects to placebo. RESULTS KAF156 at all dose levels was safe and well tolerated. Two serious adverse events were reported - both resolved without sequelae and neither was considered related to KAF156. In Part 1, all participants treated with KAF156 and none of those randomized to placebo were protected against malaria infection. In Part 2, all participants treated with placebo or 20 mg KAF156 developed malaria infection. In contrast, 50 mg KAF156 protected 3/14 participants from infection, and doses of 800, 300, and 100 mg KAF156 protected all subjects against infection. An exposure-response analysis suggested that a 24-hr post-dose concentration of KAF156 of 21·5 ng/mL (90% CI 17.66 to 25.32 ng/mL) would ensure a 95% chance of protection from malaria parasite infection. CONCLUSIONS KAF156 was safe and well tolerated and demonstrated high levels of pre- and post-CHMI protective efficacy.
Collapse
Affiliation(s)
- James G Kublin
- Seattle Malaria Clinical Trials Center, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Sean C Murphy
- Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Janine Maenza
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Annette M Seilie
- Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Jay Prakash Jain
- Novartis Institutes for BioMedical Research, Emeryville, California, USA.,Novartis Healthcare Pvt Ltd, Salarpuria-Sattva Knowledge City Raidurg, Rangareddy District Madhapur/ Hyderabad, Rangareddy, India
| | - David Berger
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Danielle Spera
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Rong Zhao
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Rachel L Soon
- Novartis Pharmaceuticals, Health Plaza, East Hanover, NJ
| | - Julie L Czartoski
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | | | - Elizabeth Duke
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ming Chang
- Departments of Laboratory Medicine and Microbiology and the Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Ashley Vaughan
- Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - F Joel Leong
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Peter Pertel
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - William T Prince
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| |
Collapse
|
89
|
Overall, anti-malarial, and non-malarial effect of intermittent preventive treatment during pregnancy with sulfadoxine-pyrimethamine on birthweight: a mediation analysis. Lancet Glob Health 2020; 8:e942-e953. [PMID: 32562650 PMCID: PMC7303957 DOI: 10.1016/s2214-109x(20)30119-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Trials of intermittent preventive treatment (IPTp) of malaria in pregnant women that compared dihydroartemisinin-piperaquine with the standard of care, sulfadoxine-pyrimethamine, showed dihydroartemisinin-piperaquine was superior at preventing malaria infection, but not at improving birthweight. We aimed to assess whether sulfadoxine-pyrimethamine shows greater non-malarial benefits for birth outcomes than does dihydroartemisinin-piperaquine, and whether dihydroartemisinin-piperaquine shows greater antimalarial benefits for birth outcomes than does sulfadoxine-pyrimethamine. METHODS We defined treatment as random assignment to sulfadoxine-pyrimethamine or dihydroartemisinin-piperaquine before pooling individual participant-level data from 1617 HIV-uninfected pregnant women in Kenya (one trial; n=806) and Uganda (two trials; n=811). We quantified the relative effect of treatment on birthweight (primary outcome) attributed to preventing placental malaria infection (mediator). We estimated antimalarial (indirect) and non-malarial (direct) effects of IPTp on birth outcomes using causal mediation analyses, accounting for confounders. We used two-stage individual participant data meta-analyses to calculate pooled-effect sizes. FINDINGS Overall, birthweight was higher among neonates of women randomly assigned to sulfadoxine-pyrimethamine compared with women assigned to dihydroartemisinin-piperaquine (mean difference 69 g, 95% CI 26 to 112), despite placental malaria infection being lower in the dihydroartemisinin-piperaquine group (relative risk [RR] 0·64, 95% CI 0·39 to 1·04). Mediation analyses showed sulfadoxine-pyrimethamine conferred a greater non-malarial effect than did dihydroartemisinin-piperaquine (mean difference 87 g, 95% CI 43 to 131), whereas dihydroartemisinin-piperaquine conferred a slightly larger antimalarial effect than did sulfadoxine-pyrimethamine (8 g, -9 to 26), although more frequent dosing increased the antimalarial effect (31 g, 3 to 60). INTERPRETATION IPTp with sulfadoxine-pyrimethamine appears to have potent non-malarial effects on birthweight. Further research is needed to evaluate monthly dihydroartemisinin-piperaquine with sulfadoxine-pyrimethamine (or another compound with non-malarial effects) to achieve greater protection against malarial and non-malarial causes of low birthweight. FUNDING Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bill & Melinda Gates Foundation, and Worldwide Antimalarial Resistance Network.
Collapse
|
90
|
Saito M, Mansoor R, Kennon K, Anvikar AR, Ashley EA, Chandramohan D, Cohee LM, D'Alessandro U, Genton B, Gilder ME, Juma E, Kalilani-Phiri L, Kuepfer I, Laufer MK, Lwin KM, Meshnick SR, Mosha D, Muehlenbachs A, Mwapasa V, Mwebaza N, Nambozi M, Ndiaye JLA, Nosten F, Nyunt M, Ogutu B, Parikh S, Paw MK, Phyo AP, Pimanpanarak M, Piola P, Rijken MJ, Sriprawat K, Tagbor HK, Tarning J, Tinto H, Valéa I, Valecha N, White NJ, Wiladphaingern J, Stepniewska K, McGready R, Guérin PJ. Pregnancy outcomes and risk of placental malaria after artemisinin-based and quinine-based treatment for uncomplicated falciparum malaria in pregnancy: a WorldWide Antimalarial Resistance Network systematic review and individual patient data meta-analysis. BMC Med 2020; 18:138. [PMID: 32482173 PMCID: PMC7263905 DOI: 10.1186/s12916-020-01592-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Malaria in pregnancy, including asymptomatic infection, has a detrimental impact on foetal development. Individual patient data (IPD) meta-analysis was conducted to compare the association between antimalarial treatments and adverse pregnancy outcomes, including placental malaria, accompanied with the gestational age at diagnosis of uncomplicated falciparum malaria infection. METHODS A systematic review and one-stage IPD meta-analysis of studies assessing the efficacy of artemisinin-based and quinine-based treatments for patent microscopic uncomplicated falciparum malaria infection (hereinafter uncomplicated falciparum malaria) in pregnancy was conducted. The risks of stillbirth (pregnancy loss at ≥ 28.0 weeks of gestation), moderate to late preterm birth (PTB, live birth between 32.0 and < 37.0 weeks), small for gestational age (SGA, birthweight of < 10th percentile), and placental malaria (defined as deposition of malaria pigment in the placenta with or without parasites) after different treatments of uncomplicated falciparum malaria were assessed by mixed-effects logistic regression, using artemether-lumefantrine, the most used antimalarial, as the reference standard. Registration PROSPERO: CRD42018104013. RESULTS Of the 22 eligible studies (n = 5015), IPD from16 studies were shared, representing 95.0% (n = 4765) of the women enrolled in literature. Malaria treatment in this pooled analysis mostly occurred in the second (68.4%, 3064/4501) or third trimester (31.6%, 1421/4501), with gestational age confirmed by ultrasound in 91.5% (4120/4503). Quinine (n = 184) and five commonly used artemisinin-based combination therapies (ACTs) were included: artemether-lumefantrine (n = 1087), artesunate-amodiaquine (n = 775), artesunate-mefloquine (n = 965), and dihydroartemisinin-piperaquine (n = 837). The overall pooled proportion of stillbirth was 1.1% (84/4361), PTB 10.0% (619/4131), SGA 32.3% (1007/3707), and placental malaria 80.1% (2543/3035), and there were no significant differences of considered outcomes by ACT. Higher parasitaemia before treatment was associated with a higher risk of SGA (adjusted odds ratio [aOR] 1.14 per 10-fold increase, 95% confidence interval [CI] 1.03 to 1.26, p = 0.009) and deposition of malaria pigment in the placenta (aOR 1.67 per 10-fold increase, 95% CI 1.42 to 1.96, p < 0.001). CONCLUSIONS The risks of stillbirth, PTB, SGA, and placental malaria were not different between the commonly used ACTs. The risk of SGA was high among pregnant women infected with falciparum malaria despite treatment with highly effective drugs. Reduction of malaria-associated adverse birth outcomes requires effective prevention in pregnant women.
Collapse
Affiliation(s)
- Makoto Saito
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.
- Infectious Diseases Data Observatory (IDDO), Oxford, UK.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Rashid Mansoor
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kalynn Kennon
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Elizabeth A Ashley
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Lao-Oxford-Mahosot Hospital-Wellcome Trust Research Unit, Vientiane, Lao PDR
| | | | - Lauren M Cohee
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Umberto D'Alessandro
- Medical Research Council Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Blaise Genton
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- University Center of General Medicine and Public Health, Lausanne, Switzerland
| | - Mary Ellen Gilder
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | | | - Linda Kalilani-Phiri
- Department of Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Irene Kuepfer
- London School of Hygiene and Tropical Medicine, London, UK
| | - Miriam K Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Khin Maung Lwin
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | | | - Victor Mwapasa
- Department of Medicine, University of Malawi College of Medicine, Blantyre, Malawi
| | - Norah Mwebaza
- Infectious Disease Research Collaboration, Makerere University, Kampala, Uganda
| | - Michael Nambozi
- Department of Clinical Sciences, Tropical Diseases Research Centre, Ndola, Zambia
| | | | - François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Myaing Nyunt
- Duke Global Health Institute, Duke University, Durham, NC, USA
| | | | - Sunil Parikh
- Yale School of Public Health, New Haven, CT, USA
| | - Moo Kho Paw
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Aung Pyae Phyo
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
- Myanmar-Oxford Clinical Research Unit, Yangon, Myanmar
| | - Mupawjay Pimanpanarak
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | | | - Marcus J Rijken
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
- Department of Obstetrics and Gynecology, Division of Woman and Baby, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kanlaya Sriprawat
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Harry K Tagbor
- School of Medicine, University of Health and Allied Sciences, Ho, Ghana
| | - Joel Tarning
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - Innocent Valéa
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, Nanoro, Burkina Faso
| | - Neena Valecha
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Nicholas J White
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jacher Wiladphaingern
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Kasia Stepniewska
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK
- Infectious Diseases Data Observatory (IDDO), Oxford, UK
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rose McGready
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak, Thailand
| | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network (WWARN), Oxford, UK.
- Infectious Diseases Data Observatory (IDDO), Oxford, UK.
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
91
|
Effect of Drug Pressure on Promoting the Emergence of Antimalarial-Resistant Parasites among Pregnant Women in Ghana. Antimicrob Agents Chemother 2020; 64:AAC.02029-19. [PMID: 32179528 DOI: 10.1128/aac.02029-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/08/2020] [Indexed: 11/20/2022] Open
Abstract
The continuous spread of antimalarial drug resistance is a threat to current chemotherapy efficacy. Therefore, characterizing the genetic diversity of drug resistance markers is needed to follow treatment effectiveness and further update control strategies. Here, we genotyped Plasmodium falciparum resistance gene markers associated with sulfadoxine-pyrimethamine (SP) and artemisinin-based combination therapy (ACT) in isolates from pregnant women in Ghana. The prevalence of the septuple IRN I- A/FG K GS/T pfdhfr/pfdhps haplotypes, including the pfdhps A581G and A613S/T mutations, was high at delivery among post-SP treatment isolates (18.2%) compared to those of first antenatal care (before initiation of intermittent preventive treatment of malaria in pregnancy with sulfadoxine-pyrimethamine [IPTp-SP]; 6.1%; P = 0.03). Regarding the pfk13 marker gene, two nonsynonymous mutations (N458D and A481C) were detected at positions previously related to artemisinin resistance in isolates from Southeast Asia. These mutations were predicted in silico to alter the stability of the pfk13 propeller-encoding domain. Overall, these findings highlight the need for intensified monitoring and surveillance of additional mutations associated with increased SP resistance as well as emergence of resistance against artemisinin derivatives.
Collapse
|
92
|
Geographical Accessibility to Glucose-6-Phosphate Dioxygenase Deficiency Point-of-Care Testing for Antenatal Care in Ghana. Diagnostics (Basel) 2020; 10:diagnostics10040229. [PMID: 32316233 PMCID: PMC7235997 DOI: 10.3390/diagnostics10040229] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 02/01/2023] Open
Abstract
Background: Glucose-6-Phosphate Dehydrogenase (G6PD) deficiency screening test is essential for malaria treatment, control, and elimination programs. G6PD deficient individuals are at high risk of severe hemolysis when given anti-malarial drugs such as primaquine, quinine, other sulphonamide-containing medicines, and chloroquine, which has recently been shown to be potent for the treatment of coronavirus disease (COVID-19). We evaluated the geographical accessibility to POC testing for G6PD deficiency in Ghana, a malaria-endemic country. Methods: We obtained the geographic information of 100 randomly sampled clinics previously included in a cross-sectional survey. We also obtained the geolocated data of all public hospitals providing G6PD deficiency testing services in the region. Using ArcGIS 10.5, we quantified geographical access to G6PD deficiency screening test and identified clinics as well as visualize locations with poor access for targeted improvement. The travel time was estimated using an assumed speed of 20 km per hour. Findings: Of the 100 clinics, 58% were Community-based Health Planning and Services facilities, and 42% were sub-district health centers. The majority (92%) were Ghana Health Service facilities, and the remaining 8% were Christian Health Association of Ghana facilities. Access to G6PD deficiency screening test was varied across the districts, and G6PD deficiency screening test was available in all eight public hospitals. This implies that the health facility-to-population ratio for G6PD deficiency testing service was approximately 1:159,210 (8/1,273,677) population. The spatial analysis quantified the current mean distance to a G6PD deficiency testing service from all locations in the region to be 34 ± 14 km, and travel time (68 ± 27 min). The estimated mean distance from a clinic to a district hospital for G6PD deficiency testing services was 15 ± 11 km, and travel time (46 ± 33 min). Conclusion: Access to POC testing for G6PD deficiency in Ghana was poor. Given the challenges associated with G6PD deficiency, it would be essential to improve access to G6PD deficiency POC testing to facilitate administration of sulphadoxine-pyrimethamine to pregnant women, full implementation of the malaria control program in Ghana, and treatment of COVID-19 patients with chloroquine in malaria-endemic countries. To enable the World Health Organization include appropriate G6PD POC diagnostic tests in its list of essential in-vitro diagnostics for use in resource-limited settings, we recommend a wider evaluation of available POC diagnostic tests for G6PD deficiency, particularly in malaria-endemic countries.
Collapse
|
93
|
Fowkes FJI, Davidson E, Moore KA, McGready R, Simpson JA. The invisible burden of malaria-attributable stillbirths. Lancet 2020; 395:268. [PMID: 31982063 DOI: 10.1016/s0140-6736(19)33011-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/08/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Freya J I Fowkes
- Burnet Institute, Melbourne, VIC 3004, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, VIC, Australia.
| | - Eliza Davidson
- Burnet Institute, Melbourne, VIC 3004, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, VIC, Australia
| | - Kerryn A Moore
- Burnet Institute, Melbourne, VIC 3004, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, VIC, Australia
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, VIC, Australia
| |
Collapse
|
94
|
No evidence of P. falciparum K13 artemisinin conferring mutations over a 24-year analysis in Coastal Kenya, but a near complete reversion to chloroquine wild type parasites. Antimicrob Agents Chemother 2019:AAC.01067-19. [PMID: 31591113 PMCID: PMC6879256 DOI: 10.1128/aac.01067-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antimalarial drug resistance is a substantial impediment to malaria control. The spread of resistance has been described using genetic markers, which are important epidemiological tools. We carried out a temporal analysis of changes in allele frequencies of 12 drug resistance markers over 2 decades of changing antimalarial drug policy in Kenya. Antimalarial drug resistance is a substantial impediment to malaria control. The spread of resistance has been described using genetic markers, which are important epidemiological tools. We carried out a temporal analysis of changes in allele frequencies of 12 drug resistance markers over 2 decades of changing antimalarial drug policy in Kenya. We did not detect any of the validated kelch 13 (k13) artemisinin resistance markers; nonetheless, a single k13 allele, K189T, was maintained at a stable high frequency (>10%) over time. There was a distinct shift from chloroquine-resistant transporter (crt)-76, multidrug-resistant gene 1 (mdr1)-86 and mdr1-1246 chloroquine (CQ) resistance alleles to a 99% prevalence of CQ-sensitive alleles in the population, following the withdrawal of CQ from routine use. In contrast, the dihydropteroate synthetase (dhps) double mutant (437G and 540E) associated with sulfadoxine-pyrimethamine (SP) resistance was maintained at a high frequency (>75%), after a change from SP to artemisinin combination therapies (ACTs). The novel cysteine desulfurase (nfs) K65 allele, implicated in resistance to lumefantrine in a West African study, showed a gradual significant decline in allele frequency pre- and post-ACT introduction (from 38% to 20%), suggesting evidence of directional selection in Kenya, potentially not due to lumefantrine. The high frequency of CQ-sensitive parasites circulating in the population suggests that the reintroduction of CQ in combination therapy for the treatment of malaria can be considered in the future. However, the risk of a reemergence of CQ-resistant parasites circulating below detectable levels or being reintroduced from other regions remains.
Collapse
|
95
|
Ishengoma DS, Saidi Q, Sibley CH, Roper C, Alifrangis M. Deployment and utilization of next-generation sequencing of Plasmodium falciparum to guide anti-malarial drug policy decisions in sub-Saharan Africa: opportunities and challenges. Malar J 2019; 18:267. [PMID: 31477109 PMCID: PMC6719357 DOI: 10.1186/s12936-019-2853-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/22/2019] [Indexed: 01/13/2023] Open
Abstract
Parasite resistance against anti-malarial drugs is a major threat to the ongoing malaria control and elimination strategies. This is especially true since resistance to the currently recommended artemisinins and partner drugs has been confirmed in South East Asia (SEA) and new anti-malarial compounds are not expected to be available in the near future. Spread from SEA or independent emergence of artemisinin resistance in sub-Saharan Africa (SSA) could reverse the achievements in malaria control that have been attained in the past two decades and derail the ongoing elimination strategies. The current surveillance of clinical efficacy and resistance to anti-malarial drugs is based on efficacy trials to assess the clinical performance of anti-malarials, in vivo/ex vivo assessment of parasite susceptibility to anti-malarials and prevalence of known molecular markers of drug resistance. Whereas clinical efficacy trials are restricted by cost and the complex logistics of patient follow-up, molecular detection of genetic mutations associated with resistance or reduced susceptibility to anti-malarials is by contrast a simple and powerful tool for early detection and monitoring of the prevalence of resistant parasites at population level. This provides needed information before clinical failure emerges, allowing policy makers to anticipate problems and respond. The various methods previously used in detection of molecular markers of drug resistance share some limitations: low-throughput, and high costs per sample and demanding infrastructure. However, recent technological advances including next-generation sequencing (NGS) methodologies promise greatly increased throughput and reduced costs, essentially providing unprecedented potential to address different research and operational questions of relevance for drug policy. This review assesses the potential role of NGS to provide comprehensive information that could guide drug policies in malaria endemic countries and looks at the foreseeable challenges facing the establishment of NGS approaches for routine surveillance of parasite resistance to anti-malarials in SSA.
Collapse
Affiliation(s)
- Deus S Ishengoma
- National Institute for Medical Research, Tanga Centre, Tanga, Tanzania.
| | - Queen Saidi
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Carol H Sibley
- Department of Genome Sciences, University of Washington, Seattle, USA
| | - Cally Roper
- London School of Hygiene & Tropical Medicine, London, UK
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
96
|
Kakuru A, Staedke SG, Dorsey G, Rogerson S, Chandramohan D. Impact of Plasmodium falciparum malaria and intermittent preventive treatment of malaria in pregnancy on the risk of malaria in infants: a systematic review. Malar J 2019; 18:304. [PMID: 31481075 PMCID: PMC6724246 DOI: 10.1186/s12936-019-2943-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 08/28/2019] [Indexed: 09/16/2023] Open
Abstract
Background Studies of the association between malaria in pregnancy (MiP) and malaria during infancy have provided mixed results. A systematic review was conducted to evaluate available evidence on the impact of Plasmodium falciparum malaria infection during pregnancy, and intermittent preventive treatment of malaria during pregnancy (IPTp), on the risk of clinical malaria or parasitaemia during infancy. Methods MEDLINE, EMBASE, Global Health, and Malaria in Pregnancy Library databases were searched from inception to 22 May 2018 for articles published in English that reported on associations between MiP and malaria risk in infancy. Search terms included malaria, Plasmodium falciparum, pregnancy, placenta, maternal, prenatal, foetal, newborn, infant, child or offspring or preschool. Randomized controlled trials and prospective cohort studies, which followed infants for at least 6 months, were included if any of the following outcomes were reported: incidence of clinical malaria, prevalence of parasitaemia, and time to first episode of parasitaemia or clinical malaria. Substantial heterogeneity between studies precluded meta-analysis. Thus, a narrative synthesis of included studies was conducted. Results The search yielded 14 published studies, 10 prospective cohort studies and four randomized trials; all were conducted in sub-Saharan Africa. Infants born to mothers with parasitaemia during pregnancy were at higher risk of malaria in three of four studies that assessed this association. Placental malaria detected by microscopy or histology was associated with a higher risk of malaria during infancy in nine of 12 studies, but only one study adjusted for malaria transmission intensity. No statistically significant associations between the use of IPTp or different IPTp regimens and the risk of malaria during infancy were identified. Conclusion Evidence of an association between MiP and IPTp and risk of malaria in infancy is limited and of variable quality. Most studies did not adequately adjust for malaria transmission intensity shared by mothers and their infants. Further research is needed to confirm or exclude an association between MiP and malaria in infancy. Randomized trials evaluating highly effective interventions aimed at preventing MiP, such as IPTp with dihydroartemisinin–piperaquine, may help to establish a causal association between MiP and malaria in infancy.
Collapse
Affiliation(s)
- Abel Kakuru
- Infectious Diseases Research Collaboration, P.O Box 7475, Kampala, Uganda. .,London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Sarah G Staedke
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Grant Dorsey
- University of California San Francisco, San Francisco, CA, USA
| | - Stephen Rogerson
- Department of Medicine at the Doherty Institute, University of Melbourne, 792 Elizabeth Street, Melbourne, VIC, 3000, Australia
| | - Daniel Chandramohan
- London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
97
|
Divala TH, Cohee LM, Laufer MK. The remarkable tenacity of sulfadoxine-pyrimethamine. THE LANCET. INFECTIOUS DISEASES 2019; 19:460-461. [PMID: 30922819 PMCID: PMC10321323 DOI: 10.1016/s1473-3099(18)30796-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Titus H Divala
- University of Malawi College of Medicine, Blantyre, Malawi
| | - Lauren M Cohee
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Miriam K Laufer
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|