5,162
Views
0
CrossRef citations to date
0
Altmetric
Review

Antimalarial treatment in infants

, , , &

References

  • World Health Organization (WHO). World malaria report. WHO, Geneva, 2021.
  • D’Alessandro U, Ubben D, Hamed K, et al. Malaria in infants aged less than six months - is it an area of unmet medical need? Malar J. 2012;11(1):400.
  • Botwe AK, Oppong FB, Gyaase S, et al. Determinants of the varied profiles of Plasmodium falciparum infections among infants living in Kintampo, Ghana. Malar J. 2021;20(1):240.
  • Botwe AK, Owusu-Agyei S, Asghar M, et al. Profiles of Plasmodium falciparum infections detected by microscopy through the first year of life in Kintampo a high transmission area of Ghana. PLoS One. 2020;15(10):e0240814.
  • Ceesay SJ, Koivogui L, Nahum A, et al. Malaria prevalence among young infants in different transmission settings, Africa. Emerg Infect Dis. 2015;21(7):1114–1121.
  • Natama HM, Rovira-Vallbona E, Some MA, et al. Malaria incidence and prevalence during the first year of life in Nanoro, Burkina Faso: a birth-cohort study. Malar J. 2018;17(1):163.
  • World Health Organization (WHO). Guidelines for malaria. Geneva: WHO; 2021.
  • Wanzira H, Kakuru A, Arinaitwe E, et al. Longitudinal outcomes in a cohort of Ugandan children randomized to artemether-lumefantrine versus dihydroartemisinin-piperaquine for the treatment of malaria. Clin Infect Dis. 2014;59(4):509–516.
  • Afolabi BM, Rabiu KA, Sanyaolu NO. Malaria in the first 6 months of life in urban African infants with anemia. Am J Trop Med Hyg. 2001;65(6):822–827.
  • Binka F, Morris SS, Ross DA. Patterns of malaria morbidity and mortality in children in northern Ghana. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1994;88(4):381–385.
  • Klein Klouwenberg PM, Oyakhirome S, Schwarz NG, et al. Malaria and asymptomatic parasitaemia in Gabonese infants under the age of 3 months. Acta Trop. 2005;95(2):81–85.
  • Carneiro I, Roca-Feltrer A, Griffin JT, et al. Age-patterns of malaria vary with severity, transmission intensity and seasonality in sub-Saharan Africa: a systematic review and pooled analysis. PLoS One. 2010;5(2):e8988.
  • Danwang C, Bigna JJ, Nzalie RNT, et al. Epidemiology of clinical congenital and neonatal malaria in endemic settings: a systematic review and meta-analysis. Malar J. 2020;19(1):312.
  • Dobbs KR, Dent AE. Plasmodium malaria and antimalarial antibodies in the first year of life. Parasitology. 2016;143(2):129–138.
  • Bilal JA, Malik EE, Al-Nafeesah A, et al. Global prevalence of congenital malaria: a systematic review and meta-analysis. Eur J Obstet Gynecol Reprod Biol. 2020;252:534–542.
  • Menendez C, Mayor A. Congenital malaria: the least known consequence of malaria in pregnancy. Semin Fetal Neonatal Med. 2007;12(3):207–213.
  • Alonso S, Vidal M, Ruiz-Olalla G, et al. HIV infection and placental malaria reduce maternal transfer of multiple antimalarial antibodies in Mozambican women. J Infect. 2021;82(4):45–57.
  • Agbota G, Accrombessi M, Cottrell G, et al. Increased risk of malaria during the first year of life in small-for-gestational-age infants: a longitudinal study in Benin. J Infect Dis. 2019;219(10):1642–1651.
  • Sotimehin SA, Runsewe-Abiodun TI, Oladapo OT, et al. Possible risk factors for congenital malaria at a tertiary care hospital in Sagamu, Ogun State, South-West Nigeria. J Trop Pediatr. 2008;54(5):313–320.
  • Batchelor HK, Marriott JF. Paediatric pharmacokinetics: key considerations. Br J Clin Pharmacol. 2015;79(3):395–404.
  • Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacology–drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157–1167.
  • Lu H, Rosenbaum S. Developmental pharmacokinetics in pediatric populations. J Pediatr Pharmacol Ther. 2014;19(4):262–276.
  • White NJ. Pharmacokinetic and pharmacodynamic considerations in antimalarial dose optimization. Antimicrob Agents Chemother. 2013;57(12):5792–5807.
  • White NJ. Assessment of the pharmacodynamic properties of antimalarial drugs in vivo. Antimicrob Agents Chemother. 1997;41(7):1413–1422.
  • White NJ, van Vugt M, Ezzet F. Clinical pharmacokinetics and pharmacodynamics and pharmacodynamics of artemether-lumefantrine. Clin Pharmacokinet. 1999;37(2):105–125.
  • Simpson JA, Jamsen KM, Price RN, et al. Towards optimal design of anti-malarial pharmacokinetic studies. Malar J. 2009;8(1):189.
  • Verrest L, Wilthagen EA, Beijnen JH, et al. Influence of malnutrition on the pharmacokinetics of drugs used in the treatment of poverty-related diseases: a systematic review. Clin Pharmacokinet. 2021;60(9):1149–1169.
  • Dondorp AM, Fanello CI, Hendriksen IC, et al. Artesunate versus quinine in the treatment of severe falciparum malaria in African children (AQUAMAT): an open-label, randomised trial. Lancet. 2010;376(9753):1647–1657.
  • Dondorp A, Nosten F, Stepniewska K, et al. South east asian quinine artesunate malaria trial g: artesunate versus quinine for treatment of severe falciparum malaria: a randomised trial. Lancet. 2005;366(9487):717–725.
  • Cao XT, Bethell DB, Pham TP, et al. Comparison of artemisinin suppositories, intramuscular artesunate and intravenous quinine for the treatment of severe childhood malaria. Trans R Soc Trop Med Hyg. 1997;91(3):335–342.
  • Sinclair D, Donegan S, Isba R, et al. Artesunate versus quinine for treating severe malaria. Cochrane Database Syst Rev. 2012;2012(6):CD005967
  • Zaloumis SG, Tarning J, Krishna S, et al. Population pharmacokinetics of intravenous artesunate: a pooled analysis of individual data from patients with severe malaria. CPT Pharmacometrics Syst Pharmacol. 2014;3(11):e145.
  • Hendriksen IC, Mtove G, Kent A, et al. Population pharmacokinetics of intramuscular artesunate in African children with severe malaria: implications for a practical dosing regimen. Clin Pharmacol Ther. 2013;93(5):443–450.
  • Kitabi E, Bensman TJ, Earp JC, et al. Effect of body weight and age on the pharmacokinetics of dihydroartemisinin: food and drug administration basis for dose determination of artesunate for injection in pediatric patients with severe malaria. Clin Infect Dis. 2021;73(5):903–906.
  • Kremsner PG, Adegnika AA, Hounkpatin AB, et al. Intramuscular artesunate for severe malaria in African children: a multicenter randomized controlled trial. PLoS Med. 2016;13(1):e1001938.
  • Kremsner PG, Taylor T, Issifou S, et al. A simplified intravenous artesunate regimen for severe malaria. J Infect Dis. 2012;205(2):312–319.
  • Fanello C, Onyamboko M, Lee SJ, et al. Post-treatment haemolysis in African children with hyperparasitaemic falciparum malaria; a randomized comparison of artesunate and quinine. BMC Infect Dis. 2017;17(1):575.
  • Rolling T, Agbenyega T, Issifou S, et al. Delayed hemolysis after treatment with parenteral artesunate in African children with severe malaria–a double-center prospective study. J Infect Dis. 2014;209(12):1921–1928.
  • Krishna S, Planche T, Agbenyega T, et al. Bioavailability and preliminary clinical efficacy of intrarectal artesunate in Ghanaian children with moderate malaria. Antimicrob Agents Chemother. 2001;45(2):509–516.
  • Okebe J, Eisenhut M. Pre-referral rectal artesunate for severe malaria. Cochrane Database Syst Rev. 2014;(5):CD009964.
  • Gomes MF, Faiz MA, Gyapong JO, et al. Pre-referral rectal artesunate to prevent death and disability in severe malaria: a placebo-controlled trial. Lancet. 2009;373(9663):557–566.
  • Grobusch MP. Early rectal artesunate administration: a life-saver in remote areas? Future Microbiol. 2009;4(4):397–400.
  • Esu EB, Effa EE, Opie ON, et al. Artemether for severe malaria. Cochrane Database Syst Rev. 2019;6: CD010678 doi:10.1002/14651858.CD010678.pub3
  • Bobossi-Serengbe G, Gody JC, Fioboy R, et al. [Comparison of the effectiveness of artemether and quinine for treatment of severe malaria in children, Bangui, Central African Republic]. Bull Soc Pathol Exot. 2015;108(2):107–111.
  • Huda SN, Shahab T, Ali SM, et al. A comparative clinical trial of artemether and quinine in children with severe malaria. Indian Pediatr. 2003;40(10):939–945.
  • Minta D, Sissoko M, Sidibe I, et al. Efficacy and safety of artemether in the treatment of severe end complicated malaria in Mali. Mali Med. 2005;20(1–2):28–32.
  • Murphy SA, Mberu E, Muhia D, et al. The disposition of intramuscular artemether in children with cerebral malaria; a preliminary study. Trans R Soc Trop Med Hyg. 1997;91(3):331–334.
  • Mithwani S, Aarons L, Kokwaro GO, et al. Population pharmacokinetics of artemether and dihydroartemisinin following single intramuscular dosing of artemether in African children with severe falciparum malaria. Br J Clin Pharmacol. 2004;57(2):146–152.
  • Aceng JR, Byarugaba JS, Tumwine JK. Rectal artemether versus intravenous quinine for the treatment of cerebral malaria in children in Uganda: randomised clinical trial. BMJ. 2005;330(7487):334.
  • Krishna S, White NJ. Pharmacokinetics of quinine, chloroquine and amodiaquine. Clinical implications. Clin Pharmacokinet. 1996;30(4):263–299.
  • Schapira A, Solomon T, Julien M, et al. Comparison of intramuscular and intravenous quinine for the treatment of severe and complicated malaria in children. Trans R Soc Trop Med Hyg. 1993;87(3):299–302.
  • Achan J, Byarugaba J, Barennes H, et al. Rectal versus intravenous quinine for the treatment of childhood cerebral malaria in Kampala, Uganda: a randomized, double-blind clinical trial. Clin Infect Dis. 2007;45(11):1446–1452.
  • Assimadi JK, Gbadoe AD, Agbodjan-Djossou O, et al. [Diluted injectable quinine in the intramuscular and intrarectal route: comparative efficacity and tolerance in malaria treatment for children]. Med Trop (Mars). 2002;62(2):158–162.
  • Eisenhut M, Omari A, MacLehose HG. Intrarectal quinine for treating Plasmodium falciparum malaria: a systematic review. Malar J. 2005;4(1):24.
  • Hendriksen IC, Maiga D, Lemnge MM, et al. Population pharmacokinetic and pharmacodynamic properties of intramuscular quinine in Tanzanian children with severe Falciparum malaria. Antimicrob Agents Chemother. 2013;57(2):775–783.
  • Saeheng T, Na-Bangchang K. Clinical pharmacokinetics of quinine and its relationship with treatment outcomes in children, pregnant women, and elderly patients, with uncomplicated and complicated malaria: a systematic review. Malar J. 2022;21(1):41.
  • Shann FA, Stace J, Edstein M. Pharmacokinetics of quinine in children. J Pediatr. 1985;106(3):506–510.
  • Krishna S, Nagaraja NV, Planche T, et al. Population pharmacokinetics of intramuscular quinine in children with severe malaria. Antimicrob Agents Chemother. 2001;45(6):1803–1809.
  • Achan J, Talisuna AO, Erhart A, et al. Quinine, an old anti-malarial drug in a modern world: role in the treatment of malaria. Malar J. 2011;10(1):144.
  • World Health Organization (WHO). Report on antimalarial drug efficacy, resistance and response In.; 2020.
  • WHO. Methods and techniques for assessing exposure to antimalarial drugs in clinical field studies. Informal consultation organized by the World Health Organization, with the technical support of the worldwide antimalarial resistance network. Bangkok; 2010.
  • World Health Organization (WHO). Guidelines for the treatment of malaria. third edition ed. Geneva: World Health Organization; 2015.
  • Guidi M, Mercier T, Aouri M, et al. Population pharmacokinetics and pharmacodynamics of the artesunate-mefloquine fixed dose combination for the treatment of uncomplicated falciparum malaria in African children. Malar J. 2019;18(1):139.
  • Djimde AA, Tekete M, Abdulla S, et al. Pharmacokinetic and pharmacodynamic characteristics of a new pediatric formulation of artemether-lumefantrine in African children with uncomplicated Plasmodium falciparum malaria. Antimicrob Agents Chemother. 2011;55(9):3994–3999.
  • Plucinski MM, Dimbu PR, Macaia AP, et al. Efficacy of artemether-lumefantrine, artesunate-amodiaquine, and dihydroartemisinin-piperaquine for treatment of uncomplicated Plasmodium falciparum malaria in Angola, 2015. Malar J. 2017;16(1):62.
  • Ishengoma DS, Mandara CI, Francis F, et al. Efficacy and safety of artemether-lumefantrine for the treatment of uncomplicated malaria and prevalence of Pfk13 and Pfmdr1 polymorphisms after a decade of using artemisinin-based combination therapy in mainland Tanzania. Malar J. 2019;18(1):88.
  • Zongo I, Compaore YD, Nikiema F, et al. Efficacy of artemether-lumefantrine and artesunate-amodiaquine as first line therapy of uncomplicated malaria in Burkina Faso, 11 years after policy change. Pan Afr Med J. 2020;35:68.
  • Yeka A, Wallender E, Mulebeke R, et al. Comparative efficacy of artemether-lumefantrine and dihydroartemisinin-piperaquine for the treatment of uncomplicated malaria in Ugandan children. J Infect Dis. 2019;219(7):1112–1120.
  • Beavogui AH, Camara A, Delamou A, et al. Efficacy and safety of artesunate-amodiaquine and artemether-lumefantrine and prevalence of molecular markers associated with resistance, Guinea: an open-label two-arm randomised controlled trial. Malar J. 2020;19(1):223.
  • Tiono AB, Tinto H, Alao MJ, et al. Increased systemic exposures of artemether and dihydroartemisinin in infants under 5 kg with uncomplicated Plasmodium falciparum malaria treated with artemether-lumefantrine (Coartem(R)). Malar J. 2015;14(1):157.
  • Bassat Q, Gonzalez R, Machevo S, et al. Similar efficacy and safety of artemether-lumefantrine (Coartem(R)) in African infants and children with uncomplicated falciparum malaria across different body weight ranges. Malar J. 2011;10(1):369.
  • Kloprogge F, Workman L, Borrmann S, et al. Artemether-lumefantrine dosing for malaria treatment in young children and pregnant women: a pharmacokinetic-pharmacodynamic meta-analysis. PLoS Med. 2018;15(6):e1002579.
  • Tchaparian E, Sambol NC, Arinaitwe E, et al. Population pharmacokinetics and pharmacodynamics of lumefantrine in young Ugandan children treated with artemether-lumefantrine for uncomplicated malaria. J Infect Dis. 2016;214(8):1243–1251. Study on lumefantrine pharmacokinetics and dosing in infants.
  • Onyamboko MA, Fanello CI, Wongsaen K, et al. Randomized comparison of the efficacies and tolerabilities of three artemisinin-based combination treatments for children with acute Plasmodium falciparum malaria in the Democratic Republic Of The Congo. Antimicrob Agents Chemother. 2014;58(9):5528–5536.
  • Worldwide antimalarial resistance network ALDISG: the effect of dose on the antimalarial efficacy of artemether-lumefantrine: a systematic review and pooled analysis of individual patient data. Lancet Infect Dis. 2015;15(6):692–702.
  • Ngasala BE, Malmberg M, Carlsson AM, et al. Efficacy and effectiveness of artemether-lumefantrine after initial and repeated treatment in children <5 years of age with acute uncomplicated Plasmodium falciparum malaria in rural Tanzania: a randomized trial. Clin Infect Dis. 2011;52(7):873–882.
  • Garner P. WorldWide antimalarial resistance network DPSG: the effect of dosing regimens on the antimalarial efficacy of dihydroartemisinin-piperaquine: a pooled analysis of individual patient data. PLoS Med. 2013;10(12):e1001564. discussion e1001564.
  • Byakika-Kibwika P, Ssenyonga R, Lamorde M, et al. Piperaquine concentration and malaria treatment outcomes in Ugandan children treated for severe malaria with intravenous Artesunate or quinine plus dihydroartemisinin-piperaquine. BMC Infect Dis. 2019;19(1):1025.
  • Zongo I, Some FA, Somda SA, et al. Efficacy and day 7 plasma piperaquine concentrations in African children treated for uncomplicated malaria with dihydroartemisinin-piperaquine. PLoS One. 2014;9(8):e103200.
  • Sambol NC, Yan L, Creek DJ, et al. Population pharmacokinetics of piperaquine in young Ugandan children treated with dihydroartemisinin-piperaquine for uncomplicated malaria. Clin Pharmacol Ther. 2015;98(1):87–95.
  • Hoglund RM, Workman L, Edstein MD, et al. Population pharmacokinetic properties of piperaquine in falciparum malaria: an individual participant data meta-analysis. PLoS Med. 2017;14(1):e1002212.
  • Zani B, Gathu M, Donegan S, et al. Dihydroartemisinin-piperaquine for treating uncomplicated Plasmodium falciparum malaria. Cochrane Database Syst Rev. 2014;(1):CD010927.
  • Terlouw DJ, Nahlen BL, Courval JM, et al. Sulfadoxine-pyrimethamine in treatment of malaria in Western Kenya: increasing resistance and underdosing. Antimicrob Agents Chemother. 2003;47(9):2929–2932.
  • Takechi M, Matsuo M, Ziba C, et al. Therapeutic efficacy of sulphadoxine/pyrimethamine and susceptibility in vitro of P. falciparum isolates to sulphadoxine-pyremethamine and other antimalarial drugs in Malawian children. Trop Med Int Health. 2001;6(6):429–434.
  • Dorsey G, Njama D, Kamya MR, et al. Sulfadoxine/pyrimethamine alone or with amodiaquine or artesunate for treatment of uncomplicated malaria: a longitudinal randomised trial. Lancet. 2002;360(9350):2031–2038.
  • Atroosh WM, Al-Mekhlafi HM, Snounou G, et al. Sustained efficacy of artesunate-sulfadoxine-pyrimethamine against Plasmodium falciparum in Yemen and a renewed call for an adjunct single dose primaquine to clear gametocytes. Malar J. 2016;15(1):295.
  • Obonyo CO, Ochieng F, Taylor WR, et al. Artesunate plus sulfadoxine-pyrimethamine for uncomplicated malaria in Kenyan children: a randomized, double-blind, placebo-controlled trial. Trans R Soc Trop Med Hyg. 2003;97(5):585–591.
  • Abacassamo F, Enosse S, Aponte JJ, et al. Efficacy of chloroquine, amodiaquine, sulphadoxine-pyrimethamine and combination therapy with artesunate in Mozambican children with non-complicated malaria. Trop Med Int Health. 2004;9(2):200–208.
  • Warsame M, Hassan AM, Barrette A, et al. Treatment of uncomplicated malaria with artesunate plus sulfadoxine-pyrimethamine is failing in Somalia: evidence from therapeutic efficacy studies and Pfdhfr and Pfdhps mutant alleles. Trop Med Int Health. 2015;20(4):510–517.
  • Priotto G, Kabakyenga J, Pinoges L, et al. Artesunate and sulfadoxine-pyrimethamine combinations for the treatment of uncomplicated Plasmodium falciparum malaria in Uganda: a randomized, double-blind, placebo-controlled trial. Trans R Soc Trop Med Hyg. 2003;97(3):325–330.
  • Kayentao K, Doumbo OK, Penali LK, et al. Pyronaridine-artesunate granules versus artemether-lumefantrine crushed tablets in children with Plasmodium falciparum malaria: a randomized controlled trial. Malar J. 2012;11(1):364.
  • Tona Lutete G, Mombo-Ngoma G, Assi SB, et al. Pyronaridine-artesunate real-world safety, tolerability, and effectiveness in malaria patients in 5 African countries: a single-arm, open-label, cohort event monitoring study. PLoS Med. 2021;18(6):e1003669.
  • Sagara I, Beavogui AH, Zongo I, et al. Safety and efficacy of re-treatments with pyronaridine-artesunate in African patients with malaria: a substudy of the WANECAM randomised trial. Lancet Infect Dis. 2016;16(2):189–198.
  • Simpson JA, Price R, ter Kuile F, et al. Population pharmacokinetics of mefloquine in patients with acute falciparum malaria. Clin Pharmacol Ther. 1999;66(5):472–484.
  • Ramharter M, Kurth FM, Belard S, et al. Pharmacokinetics of two paediatric artesunate mefloquine drug formulations in the treatment of uncomplicated falciparum malaria in Gabon. J Antimicrob Chemother. 2007;60(5):1091–1096.
  • Schlagenhauf P, Adamcova M, Regep L, et al. Use of mefloquine in children - a review of dosage, pharmacokinetics and tolerability data. Malar J. 2011;10(1):292.
  • Fryauff DJ, Owusu-Agyei S, Utz G, et al. Mefloquine treatment for uncomplicated falciparum malaria in young children 6-24 months of age in northern Ghana. Am J Trop Med Hyg. 2007;76(2):224–231.
  • Slutsker LM, Khoromana CO, Payne D, et al. Mefloquine therapy for Plasmodium falciparum malaria in children under 5 years of age in Malawi: in vivo/in vitro efficacy and correlation of drug concentration with parasitological outcome. Bull World Health Organ. 1990;68(1):53–59.
  • Oyakhirome S, Potschke M, Schwarz NG, et al. Artesunate–amodiaquine combination therapy for falciparum malaria in young Gabonese children. Malar J. 2007;6(1):29.
  • Nhama A, Bassat Q, Enosse S, et al. In vivo efficacy of artemether-lumefantrine and artesunate-amodiaquine for the treatment of uncomplicated falciparum malaria in children: a multisite, open-label, two-cohort, clinical trial in Mozambique. Malar J. 2014;13(1):309.
  • Grandesso F, Bachy C, Donam I, et al. Efficacy of chloroquine, sulfadoxine-pyrimethamine and amodiaquine for treatment of uncomplicated Plasmodium falciparum malaria among children under five in Bongor and Koumra, Chad. Trans R Soc Trop Med Hyg. 2006;100(5):419–426.
  • Anyorigiya TA, Castel S, Mauff K, et al. Pharmacokinetic profile of amodiaquine and its active metabolite desethylamodiaquine in Ghanaian patients with uncomplicated falciparum malaria. Malar J. 2021;20(1):18.
  • Hietala SF, Bhattarai A, Msellem M, et al. Population pharmacokinetics of amodiaquine and desethylamodiaquine in pediatric patients with uncomplicated falciparum malaria. J Pharmacokinet Pharmacodyn. 2007;34(5):669–686.
  • Zwang J, Olliaro P, Barennes H, et al. Efficacy of artesunate-amodiaquine for treating uncomplicated falciparum malaria in sub-Saharan Africa: a multi-centre analysis. Malar J. 2009;8(1):203.
  • Valecha N, Phyo AP, Mayxay M, et al. An open-label, randomised study of dihydroartemisinin-piperaquine versus artesunate-mefloquine for falciparum malaria in Asia. PLoS One. 2010;5(7):e11880.
  • Marwa K, Kapesa A, Baraka V, et al. Therapeutic efficacy of artemether-lumefantrine, artesunate-amodiaquine and dihydroartemisinin-piperaquine in the treatment of uncomplicated Plasmodium falciparum malaria in Sub-Saharan Africa: a systematic review and meta-analysis. PLoS One. 2022;17(3):e0264339.
  • Kamya MR, Yeka A, Bukirwa H, et al. Artemether-lumefantrine versus dihydroartemisinin-piperaquine for treatment of malaria: a randomized trial. PLoS Clin Trials. 2007;2(5):e20.
  • Bassat Q, Mulenga M, Tinto H, et al. Dihydroartemisinin-piperaquine and artemether-lumefantrine for treating uncomplicated malaria in African children: a randomised, non-inferiority trial. PLoS One. 2009;4(11):e7871.
  • Artemisinin-based combinations study G F, Beeson JG. A head-to-head comparison of four artemisinin-based combinations for treating uncomplicated malaria in African children: a randomized trial. PLoS Med. 2011;8(11):e1001119.
  • Arinaitwe E, Sandison TG, Wanzira H, et al., Artemether-lumefantrine versus dihydroartemisinin-piperaquine for falciparum malaria: a longitudinal, randomized trial in young Ugandan children. Clin Infect Dis. 2009;49(11): 1629–1637.
  • Sinclair D, Zani B, Donegan S, et al. Artemisinin-based combination therapy for treating uncomplicated malaria. Cochrane Database Syst Rev. 2009;(3):CD007483.
  • Jittamala P, Pukrittayakamee S, Ashley EA, et al. Pharmacokinetic interactions between primaquine and pyronaridine-artesunate in healthy adult Thai subjects. Antimicrob Agents Chemother. 2015;59(1):505–513.
  • Kurth F, Belard S, Adegnika AA, et al. Do paediatric drug formulations of artemisinin combination therapies improve the treatment of children with malaria? A systematic review and meta-analysis. Lancet Infect Dis. 2010;10(2):125–132. • Summary of current knowledge on the benefit of a paediatric ACT formula.
  • Sowunmi A, Akano K, Ayede AI, et al. Therapeutic efficacy and effects of artesunate-amodiaquine and artemether-lumefantrine on malaria-associated anaemia in Nigerian children aged two years and under. Infect Dis Poverty. 2016;5(1):70.
  • Gargano N, Madrid L, Valentini G, et al. Efficacy and tolerability outcomes of a phase II, randomized, open-label, multicenter study of a new water-dispersible pediatric formulation of dihydroartemisinin-piperaquine for the treatment of uncomplicated Plasmodium falciparum malaria in African Infants. Antimicrob Agents Chemother. 2018;62(1). DOI:10.1128/AAC.00596-17.
  • Frey SG, Chelo D, Kinkela MN, et al. Artesunate-mefloquine combination therapy in acute Plasmodium falciparum malaria in young children: a field study regarding neurological and neuropsychiatric safety. Malar J. 2010;9(1):291.
  • Tietche F, Chelo D, Mina Ntoto NK, et al. Tolerability and efficacy of a pediatric granule formulation of artesunate-mefloquine in young children from Cameroon with uncomplicated falciparum malaria. Am J Trop Med Hyg. 2010;82(6):1034–1040.
  • Luxemburger C, Price RN, Nosten F, et al. Mefloquine in infants and young children. Ann Trop Paediatr. 1996;16(4):281–286.
  • Creek D, Bigira V, Arinaitwe E, et al. Increased risk of early vomiting among infants and young children treated with dihydroartemisinin-piperaquine compared with artemether-lumefantrine for uncomplicated malaria. Am J Trop Med Hyg. 2010;83(4):873–875. • Study on tolerability of A-L and DHA-P in young infants specifically.
  • Falade CO, Ogundele AO, Yusuf BO, et al. High efficacy of two artemisinin-based combinations (artemether-lumefantrine and artesunate plus amodiaquine) for acute uncomplicated malaria in Ibadan, Nigeria. Trop Med Int Health. 2008;13(5):635–643.
  • Falade CO, Dada-Adegbola HO, Ogunkunle OO, et al. Evaluation of the comparative efficacy and safety of artemether-lumefantrine, artesunate-amodiaquine and artesunate-amodiaquine-chlorpheniramine (Artemoclo) for the treatment of acute uncomplicated malaria in Nigerian children. Med Princ Pract. 2014;23(3):204–211.
  • Djalle D, Njuimo SP, Manirakiza A, et al. Efficacy and safety of artemether + lumefantrine, artesunate + sulphamethoxypyrazine-pyrimethamine and artesunate + amodiaquine and sulphadoxine-pyrimethamine + amodiaquine in the treatment of uncomplicated falciparum malaria in Bangui, Central African Republic: a randomized trial. Malar J. 2014;13:9.
  • Ashley EA, Lwin KM, McGready R, et al. An open label randomized comparison of mefloquine-artesunate as separate tablets vs. a new co-formulated combination for the treatment of uncomplicated multidrug-resistant falciparum malaria in Thailand. Trop Med Int Health. 2006;11(11):1653–1660.
  • Belard S, Ramharter M, Kurth F. Paediatric formulations of artemisinin-based combination therapies for treating uncomplicated malaria in children. Cochrane Database Syst Rev. 2020;12:CD009568.
  • Karunajeewa H, Lim C, Hung TY, et al. Safety evaluation of fixed combination piperaquine plus dihydroartemisinin (Artekin) in Cambodian children and adults with malaria. Br J Clin Pharmacol. 2004;57(1):93–99.
  • Bassat Q, Ogutu B, Djimde A, et al. Tailoring a pediatric formulation of artemether-lumefantrine for treatment of Plasmodium falciparum malaria. Antimicrob Agents Chemother. 2015;59(8):4366–4374.
  • Juma EA, Obonyo CO, Akhwale WS, et al. A randomized, open-label, comparative efficacy trial of artemether-lumefantrine suspension versus artemether-lumefantrine tablets for treatment of uncomplicated Plasmodium falciparum malaria in children in western Kenya. Malar J. 2008;7(1):262.
  • Bouyou-Akotet MK, Ramharter M, Ngoungou EB, et al. Efficacy and safety of a new pediatric artesunate-mefloquine drug formulation for the treatment of uncomplicated falciparum malaria in Gabon. Wien Klin Wochenschr. 2010;122(5–6):173–178.
  • Abdulla S, Sagara I. Dispersible formulation of artemether/lumefantrine: specifically developed for infants and young children. Malar J. 2009;8(1):S7.
  • Banek K, Webb EL, Smith SJ, et al. Adherence to treatment with artemether-lumefantrine or amodiaquine-artesunate for uncomplicated malaria in children in Sierra Leone: a randomized trial. Malar J. 2018;17(1):222.
  • Ogutu BR, Onyango KO, Koskei N, et al. Efficacy and safety of artemether-lumefantrine and dihydroartemisinin-piperaquine in the treatment of uncomplicated Plasmodium falciparum malaria in Kenyan children aged less than five years: results of an open-label, randomized, single-centre study. Malar J. 2014;13(1):33.
  • Wirima JJ, Khormana CO, Macheso AF, et al. In vivo efficacy of quinine treatment for Plasmodium falciparum malaria in Malawian children. Ann Trop Med Parasitol. 1990;84(3):223–227.
  • Roche J, Guerra-Neira A, Raso J, et al. Surveillance of in vivo resistance of Plasmodium falciparum to antimalarial drugs from 1992 to 1999 in Malabo (Equatorial Guinea). Am J Trop Med Hyg. 2003;68(5):598–601.
  • Obonyo CO, Juma EA. Clindamycin plus quinine for treating uncomplicated falciparum malaria: a systematic review and meta-analysis. Malar J. 2012;11(1):2.
  • Ramharter M, Oyakhirome S, Klein Klouwenberg P, et al. Artesunate-clindamycin versus quinine-clindamycin in the treatment of Plasmodium falciparum malaria: a randomized controlled trial. Clin Infect Dis. 2005;40(12):1777–1784.
  • Song T, Chen J, Huang L, et al. Should we abandon quinine plus antibiotic for treating uncomplicated falciparum malaria? A systematic review and meta-analysis of randomized controlled trials. Parasitol Res. 2016;115(3):903–912.
  • Kotepui M, Kotepui KU, Milanez GD, et al. Global prevalence and mortality of severe Plasmodium malariae infection: a systematic review and meta-analysis. Malar J. 2020;19(1):274.
  • Rahimi BA, Thakkinstian A, White NJ, et al. Severe vivax malaria: a systematic review and meta-analysis of clinical studies since 1900. Malar J. 2014;13(1):481.
  • Kotepui M, Kotepui KU, Milanez GD, et al. Severity and mortality of severe Plasmodium ovale infection: a systematic review and meta-analysis. PLoS One. 2020;15(6):e0235014.
  • Kenangalem E, Karyana M, Burdarm L, et al. Plasmodium vivax infection: a major determinant of severe anaemia in infancy. Malar J. 2016;15(1):321.
  • Douglas NM, Anstey NM, Buffet PA, et al. The anaemia of Plasmodium vivax malaria. Malar J. 2012;11(1):135.
  • Langford S, Douglas NM, Lampah DA, et al. Plasmodium malariae infection associated with a high burden of anemia: a hospital-based surveillance study. PLoS Negl Trop Dis. 2015;9(12):e0004195.
  • Naing C, Racloz V, Whittaker MA, et al. Efficacy and safety of dihydroartemisinin-piperaquine for treatment of Plasmodium vivax malaria in endemic countries: meta-analysis of randomized controlled studies. PLoS One. 2013;8(12):e78819.
  • Gogtay N, Kannan S, Thatte UM, et al. Artemisinin-based combination therapy for treating uncomplicated Plasmodium vivax malaria. Cochrane Database Syst Rev. 2013;(10):CD008492.
  • Awab GR, Pukrittayakamee S, Imwong M, et al. Dihydroartemisinin-piperaquine versus chloroquine to treat vivax malaria in Afghanistan: an open randomized, non-inferiority, trial. Malar J. 2010;9(1):105.
  • Karunajeewa HA, Mueller I, Senn M, et al. A trial of combination antimalarial therapies in children from Papua New Guinea. N Engl J Med. 2008;359(24):2545–2557.
  • Commons RJ, Simpson JA, Thriemer K, et al. The effect of chloroquine dose and primaquine on Plasmodium vivax recurrence: a worldwide antimalarial resistance network systematic review and individual patient pooled meta-analysis. Lancet Infect Dis. 2018;18(9):1025–1034.
  • Obua C, Hellgren U, Ntale M, et al. Population pharmacokinetics of chloroquine and sulfadoxine and treatment response in children with malaria: suggestions for an improved dose regimen. Br J Clin Pharmacol. 2008;65(4):493–501.
  • Ursing J, Eksborg S, Rombo L, et al. Chloroquine is grossly under dosed in young children with malaria: implications for drug resistance. PLoS One. 2014;9(1):e86801.
  • Zhao Q, Tensfeldt TG, Chandra R, et al. Population pharmacokinetics of azithromycin and chloroquine in healthy adults and paediatric malaria subjects following oral administration of fixed-dose azithromycin and chloroquine combination tablets. Malar J. 2014;13(1):36.
  • Ursing J, Kofoed PE, Rodrigues A, et al. Similar efficacy and tolerability of double-dose chloroquine and artemether-lumefantrine for treatment of Plasmodium falciparum infection in Guinea-Bissau: a randomized trial. J Infect Dis. 2011;203(1):109–116.
  • Goncalves BP, Pett H, Tiono AB, et al. Age, weight, and CYP2D6 genotype are major determinants of primaquine pharmacokinetics in African children. Antimicrob Agents Chemother. 2017;61(5). DOI:10.1128/AAC.02590-16
  • Vieira M, Matos Lopes TR, Mello A, et al. Doses of primaquine administered to children with Plasmodium vivax according to an age-based dose regimen. Pathog Glob Health. 2020;114(7):388–392.
  • Taylor WR, Hoglund RM, Peerawaranun P, et al. Development of weight and age-based dosing of daily primaquine for radical cure of vivax malaria. Malar J. 2021;20(1):366.
  • Setyadi A, Arguni E, Kenangalem E, et al. Safety of primaquine in infants with Plasmodium vivax malaria in Papua, Indonesia. Malar J. 2019;18(1):111.
  • Taylor WRJ, Thriemer K, von Seidlein L, et al. Short-course primaquine for the radical cure of Plasmodium vivax malaria: a multicentre, randomised, placebo-controlled non-inferiority trial. Lancet. 2019;394(10202):929–938.
  • Woon SA, Manning L, Moore BR. Antimalarials for children with Plasmodium vivax infection: current status, challenges, and research priorities. Parasitol Int. 2022;87:102512.
  • Llanos-Cuentas A, Manrrique P, Rosas-Aguirre A, et al. Tafenoquine for the treatment of Plasmodium vivax malaria. Expert Opin Pharmacother. 2022;23(7):1–10.
  • Velez ID, Hien TT, Green JA, et al. Tafenoquine exposure assessment, safety, and relapse prevention efficacy in children with Plasmodium vivax malaria: open-label, single-arm, non-comparative, multicentre, pharmacokinetic bridging, phase 2 trial. Lancet Child Adolesc Health. 2022;6(2):86–95.
  • Lesko CR, Arguin PM, Newman RD. Congenital malaria in the United States: a review of cases from 1966 to 2005. Arch Pediatr Adolesc Med. 2007;161(11):1062–1067.
  • Uneke, Uneke CJ. Congenital malaria: an overview. Tanzania Journal of Health Research. 2011;13(3). doi: 10.4314/thrb.v13i3.67615
  • Alao MJ, Gbadoe AD, Meremikwu M, et al. Plasmodium falciparum malaria in infants under 5 kg: retrospective surveillance of hospital records in five sub-saharan African countries. J Trop Pediatr. 2013;59(2):154–159.
  • Poespoprodjo JR, Hasanuddin A, Fobia W, et al. Severe congenital malaria acquired in utero. Am J Trop Med Hyg. 2010;82(4):563–565.
  • Prior AR, Prata F, Mouzinho A, et al. Congenital malaria in a European country. BMJ Case Rep. 2012;2012(dec11 1):bcr2012007310–bcr2012007310.
  • Kajoba D, Ivan Egesa W, Jean Petit H, et al. Congenital malaria in a 2-day-old neonate: a case report and literature review. Case Rep Infect Dis. 2021;2021:9960006.
  • Olupot-Olupot P, Eregu EIE, Naizuli K, et al. Neonatal and congenital malaria: a case series in malaria endemic eastern Uganda. Malar J. 2018;17(1):171.
  • Quinn. Congenital malaria: a report of four cases and a review. 1982.
  • Opare DA. Congenital malaria in newborn twins. Ghana Med J. 2010;44(2):76–78.
  • KAF/lumefantrine. Medicine for malaria venture, 2018, available at: https://www.mmv.org/newsroom/interviews/kaflumefantrine accessed 27 July 2022
  • Efficacy and safety of KAF156 in combination with LUM-SDF in adults and children with uncomplicated Plasmodium falciparum malaria, ClinicalTrials.gov, Novartis Pharmaceuticals, 2022, Available at: https://clinicaltrials.gov/ct2/show/NCT03167242, accessed 2022 Jun 27
  • Pharmacokinetics, Safety. Tolerability and efficacy of a new artemether-lumefantrine dispersible tablet in infants and neonates;5 kg body weight with acute uncomplicated Plasmodium falciparum malaria (CALINA), available at https://clinicaltrials.gov/ct2/show/NCT04300309, accessed 2022 sep 12