Scolaris Content Display Scolaris Content Display

妊娠女性におけるマラリア予防のためのメフロキン

Esta versión no es la más reciente

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Briand 2009 BEN {published and unpublished data}

Briand V, Bottero J, Noël H, Masse V, Cordel H, Guerra J, et al. Intermittent treatment for the prevention of malaria during pregnancy in Benin: a randomized, open‐label equivalence trial comparing sulfadoxine‐pyrimethamine with mefloquine. Journal of Infectious Diseases 2009;200(6):991‐1001. [DOI: 10.1086/605474]CENTRAL

Denoeud‐Ndam 2014a BEN {published and unpublished data}

Denoeud‐Ndam L, Zannou DM, Fourcade C, Taron‐Brocard C, Porcher R, Atadokpede F, et al. Cotrimoxazole prophylaxis versus mefloquine intermittent preventive treatment to prevent malaria in HIV‐infected pregnant women: two randomized controlled trials. Journal of Acquired Immune Deficiency Syndromes 2014;65(2):198‐206. [DOI: 10.1097/QAI.0000000000000058]CENTRAL

Denoeud‐Ndam 2014b BEN {published and unpublished data}

Denoeud‐Ndam L, Zannou DM, Fourcade C, Taron‐Brocard C, Porcher R, Atadokpede F, et al. Cotrimoxazole prophylaxis versus mefloquine intermittent preventive treatment to prevent malaria in HIV‐infected pregnant women: two randomized controlled trials. Journal of Acquired Immune Deficiency Syndromes 2014;65(2):198‐206. [DOI: 10.1097/QAI.0000000000000058]CENTRAL

Gonzalez 2014a BEN GAB MOZ TAN {published and unpublished data}

González R, Mombo‐Ngoma G, Ouédraogo S, Kakolwa MA, Abdulla S, Accrombessi M, et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV‐negative women: a multicentre randomized controlled trial. PLoS Medicine 2014;11(9):e1001735. [DOI: 10.1371/journal.pmed.1001733]CENTRAL
Rupérez M, González R, Mombo‐Ngoma G, Kabanywanyi AM, Sevene E, Ouédraogo S, et al. Mortality, morbidity, and developmental outcomes in infants born to women who received either mefloquine or sulfadoxine‐pyrimethamine as intermittent preventive treatment of malaria in pregnancy: a cohort study. PLoS Medicine 2016;13(2):e1001964. [DOI: 10.1371/journal.pmed.1001964]CENTRAL

Gonzalez 2014b KEN MOZ TAN {published and unpublished data}

González R, Mombo‐Ngoma G, Ouédraogo S, Kakolwa MA, Abdulla S, Accrombessi M, et al. Intermittent preventive treatment of malaria in pregnancy with mefloquine in HIV‐infected women receiving cotrimoxazole prophylaxis: a multicenter randomized placebo‐controlled trial. PLoS Medicine 2014;11(9):e1001735. [DOI: 10.1371/journal.pmed.1001735]CENTRAL

Nosten 1994 THA {published data only}

Nosten F, ter Kuile F, Maelankiri L, Chongsuphajaisiddhi T, Nopdonrattakoon L, Tangkitchot S, et al. Mefloquine prophylaxis prevents malaria during pregnancy: a double‐ blind, placebo‐controlled study. Journal of Infectious Diseases 1994;169(3):595‐603. [DOI: 10.1093/infdis/169.3.595]CENTRAL

References to studies excluded from this review

Balocco 1992 {published data only}

Balocco R, Bonati M. Mefloquine prophylaxis against malaria for female travellers of childbearing age. Lancet 1992;340(8814):309‐10. CENTRAL

Briand 2015 {published data only}

Briand V, Escolano S, Journot V, Massougbodji A, Cot M, Tubert‐Bitter P. Mefloquine versus sulfadoxine‐pyrimethamine for intermittent preventive treatment in pregnancy: a joint analysis on efficacy and tolerability. American Journal of Tropical Medicine and Hygiene 2015;93(2):300‐4. CENTRAL

Denoeud‐Ndam 2012 {published data only}

Denoeud‐Ndam L, Clément MC, Briand V, Akakpo J, Agossou VK, Atadokpédé F, et al. Tolerability of mefloquine intermittent preventive treatment for malaria in HIV‐infected pregnant women in Benin. Journal of Acquired Immune Deficiency Syndromes 2012;61(1):64‐72. CENTRAL

Nosten 1990 THA {published data only}

Nosten F, Karbwang J, White NJ, Honeymoon, Na Bangchang K, Bunnag D, et al. Mefloquine antimalarial prophylaxis in pregnancy:dose finding and pharmacokinetic study. British Journal of Clinical Pharmacology 1990;30(1):79‐85. CENTRAL

Phillips‐Howard 1998 {published data only}

Phillips‐Howard PA, Steffen R, Kerr L, Vanhauwere B, Schildknecht J, Fuchs E, et al. Safety of mefloquine and other antimalarial agents in the first trimester of pregnancy. Journal of Travel Medicine 1998;5(3):121‐6. CENTRAL

Schlagenhauf 2012 {published data only}

Schlagenhauf P, Blumentals WA, Suter P, Regep L, Vital‐Durand G, Schaerer MT, et al. Pregnancy and fetal outcomes after exposure to mefloquine in the pre‐ and periconception period and during pregnancy. Clinical Infectious Diseases 2012;54(11):e124‐31. CENTRAL

Smoak 1997 {published data only}

Smoak BL, Writer JV, Keep LW, Cowan J, Chantelois JL. The effects of inadvertent exposure of mefloquine chemoprophylaxis on pregnancy outcomes and infants of US Army servicewomen. Journal of Infectious Diseases 1992;176(3):831‐3. CENTRAL

Steketee 1996 MAL {published data only}

Steketee RW, Wirima JJ, Hightower AW, Slutsker L, Heymann DL, Breman JG. The effect of malaria and malaria prevention in pregnancy on offspring birthweight, prematurity, and intrauterine growth retardation in rural Malawi. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):33‐41. CENTRAL
Steketee RW, Wirima JJ, Slutsker L, Breman JG, Heymann DL. Comparability of treatment groups and risk factors for parasitemia at the first antenatal clinic visit in a study of malaria treatment and prevention in pregnancy in rural Malawi. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):17‐23. CENTRAL
Steketee RW, Wirima JJ, Slutsker L, Heymann DL, Breman JG. The problem of malaria and malaria control in pregnancy in sub‐Saharan Africa. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):2‐7. CENTRAL
Steketee RW, Wirima JJ, Slutsker L, Khoromana CO, Heymann DL, Breman JG. Malaria treatment and prevention in pregnancy: indications for use and adverse events associated with use of chloroquine or mefloquine. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):50‐6. CENTRAL
Steketee RW, Wirima JJ, Slutsker L, Roberts JM, Khoromana CO, Heymann DL, et al. Malaria parasite infection during pregnancy and at delivery in mother, placenta, and newborn: efficacy of chloroquine and mefloquine in rural Malawi. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):24‐32. CENTRAL
Steketee RW, Wirima JJ, Slutsker WL, Khoromana CO, Breman JG, Heymann DL. Objectives and methodology in a study of malaria treatment and prevention in pregnancy in rural Malawi: the Mangochi Malaria Research Project. American Journal of Tropical Medicine and Hygiene 1996;55(Suppl 1):8‐16. CENTRAL

Vanhauwere 1998 {published data only}

Vanhauwere B, Maradit H, Kerr L. Post‐marketing surveillance of prophylactic mefloquine (Lariam) use in pregnancy. American Journal of Tropical Medicine and Hygiene 1998;58(1):17‐21. CENTRAL

Akinyotu 2015 NIG {published data only (unpublished sought but not used)}

A comparative study of mefloquine and SP as prophylaxis against malaria in pregnant HIV‐infected patients. Ongoing studySeptember 2015.

Ataíde 2014

Ataíde R, Mayor A, Rogerson SJ. Malaria, primigravidae, and antibodies: knowledge gained and future perspectives. Trends in Parasitology 2014;30(2):85‐94.

Aubouy 2007

Aubouy A, Fievet N, Bertin G, Sagbo JC, Kossou H, Kinde‐Gazard D, et al. Dramatically decreased therapeutic efficacy of chloroquine and sulfadoxine‐pyrimethamine, but not mefloquine, in southern Benin. Tropical Medicine & International Health 2007;12(7):886‐94. [PUBMED: 17596256]

Ayisi 2003

Ayisi JG, van Eijk AM, ter Kuile FO, Kolczak MS, Otieno JA, Misore AO, et al. The effect of dual infection with HIV and malaria on pregnancy outcomes in western Kenya. AIDS 2003;17:585‐94.

Bardaji 2008

Bardaji A, Sigauque B, Bruni L, Romagosa C, Sanz S, Mabunda S, et al. Clinical malaria in African pregnant women. Malaria Journal 2008;7:27.

Bardaji 2012

Bardaji A, Bassat Q, Alonso PL, Menendez C. Intermittent preventive treatment of malaria in pregnant women and infants: making best use of the available evidence. Expert Opinion in Pharmacotherapy 2012;13(12):1719‐36.

Brabin 1983

Brabin BJ. An analysis of malaria in pregnancy in Africa. Bulletin of the World Health Organization 1983;61(6):1005‐16.

Briand 2009

Briand V, Bottero J, Noel H, Masse V, Cordel H, Guerra J, et al. Intermittent treatment for the prevention of malaria during pregnancy in Benin: a randomized, open‐label equivalence trial comparing sulfadoxine‐pyrimethamine with mefloquine. Journal of Infectious Diseases 2009;200(6):991‐1001.

Carrara 2009

Carrara VI, Zwang J, Ashley EA, Price RN, Stepniewska K, Barends M, et al. Changes in the treatment responses to artesunate‐mefloquine on the northwestern border of Thailand during 13 years of continuous deployment. PLoS ONE 2009;4(2):e4551.

CDC 2016

Centers for Disease Control and Prevention. Yellow Book ‐ Malaria, Chapter 3. Travelers' Health. Available from wwwnc.cdc.gov/travel/yellowbook/2016/infectious‐diseases‐related‐to‐travel/malaria (accessed 13 February 2018).

CDC 2017

Centers for Disease Control and Prevention. CDC Malaria Maps. Available from https://www.cdc.gov/malaria/travelers/about_maps.html (accessed 13 February 2018).

Desai 2007

Desai M, ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, et al. Epidemiology and burden of malaria in pregnancy. Lancet Infectious Diseases 2007;7(2):93‐104.

Desai 2018

Desai M, Hill J, Fernandes S, Walker P, Pell C, Gutman J, et al. Prevention of malaria in pregnancy. Lancet Infect Diseases 2018 Jan 30 [Epub ahead of print]. [DOI: 10.1016/S1473‐3099(18)30064‐1]

FDA 2004

US Food, Drug Administration. Lariam‐brand of mefloquine hydrochloride. 2004. Available from www.accessdata.fda.gov/drugsatfda_docs/label/2008/019591s024s025lbl.pdf (accessed 1 December 2014).

Gamble 2006

Gamble C, Ekwaru JP, ter Kuile FO. Insecticide‐reated nets for preventing malaria in pregnancy. Cochrane Database of Systematic Reviews 2006, Issue 2. [DOI: 10.1002/14651858.CD003755.pub2]

Gamble 2007

Gamble C, Ekwaru PJ, Garner P, ter Kuile FO. Insecticide‐treated nets for the prevention of malaria in pregnancy: a systematic review of randomised controlled trials. PLoS Medicine 2007;4(3):e107.

Gonzalez 2012

Gonzalez R, Ataide R, Naniche D, Menéndez C, Mayor A. HIV and malaria interactions: where do we stand?. Expert Review of Anti‐infective Therapy 2012;10(2):153‐65.

Gonzalez 2014

Gonzalez R, Hellgren U, Greenwood B, Menendez C. Mefloquine safety and tolerability in pregnancy: a systematic literature review. Malaria Journal 2014;13(1):75.

González 2013

González R, Hellgren U, Greenwood B, Menéndez C. Mefloquine safety and tolerability in pregnancy: a systematic literature review. Malaria Journal 2014;13:75. [DOI: 10.1186/1475‐2875‐13‐75]

GRADEpro GDT 2015 [Computer program]

McMaster University (developed by Evidence Prime). GRADEpro GDT. www.gradeworkinggroup.org/. Version accessed 6 August 2017. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.

Guyatt 2004

Guyatt HL, Snow RW. Impact of malaria during pregnancy on low birth weight in sub‐Saharan Africa. Clinical Microbiology Reviews 2004;17(4):760‐9.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC, editor(s). Chapter 8: Assessing risk of bias in included studies. In: Higgins JP, Green S, editor(s). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 (updated March 2011). The Cochrane Collaboration, 2011. Available from handbook.cochrane.org. The Cochrane Collaboration.

Iriemenam 2012

Iriemenam NC, Shah M, Gatei W, van Eijk AM, Ayis J, Kariuki S, et al. Temporal trends of sulphadoxine‐pyrimethamine (SP) drug‐resistance molecular markers in Plasmodium falciparum parasites from pregnant women in western Kenya. Malaria Journal 2012;11:134.

Kayentao 2013

Kayentao A, Garner P, van Eijk AM, Naidoo I, Roper C, Mulokozi A, et al. Intermittent preventive therapy for malaria during pregnancy using 2 vs 3 or more doses of sulfadoxine‐pyrimethamine and risk of low birth weight in Africa: systematic review and meta‐analysis. JAMA 2013;309(6):594–604. [DOI: 10.1001/jama.2012.216231]

Lee 2017

Lee SJ, ter Kuile FO, Price RN, Luxemburger C, Nosten F. Adverse effects of mefloquine for the treatment of uncomplicated malaria in Thailand: a pooled analysis of 19,850 individual patients. PLoS ONE 2017;12(2):e0168780. [DOI: 10.1371/journal.pone.0168780]

MacArthur 2001

MacArthur J, Stennies GM, Macheso A, Kolczak MS, Green MD, Ali D, et al. Efficacy of mefloquine and sulfadoxine‐pyrimethamine for the treatment of uncomplicated Plasmodium falciparum infection in Machinga District, Malawi, 1998. American Journal of Tropical Medicine and Hygiene 2001;65(6):679‐84.

Menendez 2010

Menendez C, Bardaji A, Sigauque B, Sanz S, Aponte JJ, Mabunda S, et al. Malaria prevention with IPTp during pregnancy reduces neonatal mortality. PLoS ONE 2010;5(2):9438.

Menéndez 2008

Menéndez C, Bardají A, Sigauque B, Romagosa C, Sanz S, Serra‐Casas E, et al. A randomized placebo‐controlled trial of intermittent preventive treatment in pregnant women in the context of insecticide treated nets delivered through the antenatal clinic. PLoS ONE 2008;3(4):e1934.

Menéndez 2011

Menéndez C, Serra‐Casas E, Scahill MD, Sanz S, Nhabomba A, Bardají A, et al. HIV and placental infection modulate the appearance of drug‐resistant Plasmodium falciparum in pregnant women who receive intermittent preventive treatment. Clinical Infectious Diseases 2011;52(1):41‐8.

Mockenhaupt 2008

Mockenhaupt FP, Bedu‐Addo G, Eggelte TA, Hommerich L, Holmberg V, von Oertzen, C, et al. Rapid increase in the prevalence of sulfadoxine‐pyrimethamine resistance among Plasmodium falciparum isolated from pregnant women in Ghana. Journal of Infectious Diseases 2008;198(10):1545‐9.

Nosten 1999

Nosten F, Vincenti M, Simpson J, Yei P, Thwai KL, de Vries A, et al. The effects of mefloquine treatment in pregnancy. Clinical Infectious Diseases 1999;28(4):808‐15.

Nosten 2000

Nosten F, van Vugt M, Price R, Luxemburger C, Thway KL, Brockman A, et al. Effects of artesunate‐mefloquine combination on incidence of Plasmodium falciparum malaria and mefloquine resistance in western Thailand: a prospective study. Lancet 2000;356(9226):297‐302.

Oduola 1987

Oduola AM, Milhous WK, Salako LA, Walker O, Desjardins RE. Reduced in‐vitro susceptibility to mefloquine in West African isolates of Plasmodium falciparum. Lancet 1987;2(8571):1304‐5.

Pekyi 2016

PREGACT Study Group, Pekyi D, Ampromfi AA, Tinto H, Traoré‐Coulibaly M, Tahita MC, et al. Four artemisinin‐based treatments in African pregnant women with malaria. New England Journal of Medicine 2016;374(10):913‐27. [DOI: 10.1056/NEJMoa1508606]

Phillips‐Howard 1995

Phillips‐Howard PA, ter Kuile FO. CNS adverse events associated with antimalarial agents. Fact or fiction?. Drug Experience 1995;12(6):370‐83.

Radeva‐Petrova 2014

Radeva‐Petrova D, Kayentao K, ter Kuile FO, Sinclair D, Garner P. Drugs for preventing malaria in pregnant women in endemic areas: any drug regimen versus placebo or no treatment. Cochrane Database of Systematic Reviews 2014, Issue 10. [DOI: 10.1002/14651858.CD000169.pub3]

RevMan 2014 [Computer program]

Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Schlagenhauf 2010

Schlagenhauf P, Adamcova M, Regep L, Schaerer MT, Rhein HG. The position of mefloquine as a 21st century malaria chemoprophylaxis. Malaria Journal 2010;9:357.

Schwarz 2008

Schwarz NG, Adegnika AA, Breitling LP, Gabor J, Agnandji ST, Newman RD, et al. Placental malaria increases malaria risk in the first 30 months of life. Clinical Infectious Diseases 2008;47(8):1017‐25.

Sevene 2010

Sevene E, Gonzalez R, Menendez C. Current knowledge and challenges of antimalarial drugs for treatment and prevention in pregnancy. Expert Opinion on Pharmacotherapy 2010;11(8):1277‐93.

Steffen 1993

Steffen R, Fuchs E, Schildknecht J, Naef U, Funk M, Schlagenhauf P, et al. Mefloquine compared with other malaria chemoprophylactic regimens in tourists visiting east Africa. Lancet 1993;341(8856):1299‐303.

Steketee 1996

Steketee RW, Wirima JJ, Slutsker L, Roberts JM, Khoromana CO, Heymann DL, et al. Malaria parasite infection during pregnancy and at delivery in mother, placenta, and newborn: efficacy of chloroquine and mefloquine in rural Malawi. American Journal of Tropical Medicine and Hygiene 1996;55(1 Suppl):24‐32.

Steketee 2001

Steketee RW, Nahlen BL, Parise ME, Menendez C. The burden of malaria in pregnancy in malaria‐endemic areas. American Journal of Tropical Medicine and Hygiene 2001;64:28‐35.

ter Kuile 1995

ter Kuile FO, Nosten F, Luxemburger C, Kyle D, Teja‐Isavatharm P, Phaipun L, et al. Mefloquine treatment of acute falciparummalaria: a prospective study of non‐serious adverse effects in 3673 patients. Bulletin of the World Health Organization 1995;73(5):631‐42.

ter Kuile 2004

ter Kuile FO, Parise ME, Verhoeff FH, Udhayakumar V, Newman RD, van Eijk AM, et al. The burden of co‐infection with human immunodeficiency virus type 1 and malaria in pregnant women in sub‐Saharan Africa. American Journal of Tropical Medicine and Hygiene 2004;72(Suppl 2):41‐54.

ter Kuile 2007

ter Kuile FO, van Eijk AM, Filler SJ. Effect of sulfadoxine‐pyrimethamine resistance on the efficacy of intermittent preventive therapy for malaria control during pregnancy: a systematic review. Journal of the American Medical Association 2007;297(23):2603‐16.

van Eijk 2002

van Eijk AM, Ayisi JG, Ter Kuile FO, Misore AO, Otieno JA, Kolczak MS, et al. Malaria and human immunodeficiency virus infection as risk factors for anemia in infants in Kisumu, western Kenya. American Journal of Tropical Medicine and Hygiene 2002;67:44‐53.

van Eijk 2003

van Eijk AM, Ayisi JG, ter Kuile FO, Misore AO, Otieno JA, Rosen DH, et al. HIV increases the risk of malaria in women of all gravidities in Kisumu, Kenya. AIDS 2003;17(4):595‐603.

Walker 2013

Walker PGT, Griffin JT, Cairns M, Rogerson SJ, van Eijk AM, ter Kuile F, et al. A model of parity‐dependent immunity to placental malaria. Nature Communications 2013;4:1609.

White 2005

White NJ. Intermittent presumptive treatment for malaria. PLoS Medicine 2005;2(1):e3.

WHO 2004

World Health Organization. A strategic framework for malaria prevention and control during pregnancy in the African Region. World Health Organization Regional Office for Africa2004; Vol. AFR/MAL/04/01.

WHO 2012a

World Health Organization. World malaria report. World Health Organization2012; Vol. ISBN 978 92 4 156453 3.

WHO 2012b

World Health Organization. Intermittent preventive treatment of malaria in pregnancy using sulfadoxine‐pyrimethamine (IPTp‐SP). Updated WHO Policy RecommendationOctober 2012.

WHO 2013

World Health Organization. Policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using sulfadoxine‐pyrimethamine (IPTp‐SP). WHO/HTM/GMP/2014.4. Available from www.who.int/malaria/publications/atoz/iptp‐sp‐updated‐policy‐brief‐24jan2014.pdf?ua=1 (accessed 1 December 2014).

WHO 2015

World Health Organization. Guidelines for the Treatment of Malaria, 3rd edition. Available from www.who.int/malaria/publications/atoz/9789241549127/en/ (accessed 13 February 2018).

WHO MPAC 2013

World Health Organization. Malaria Policy Advisory Committee and Secretariat. Malaria Policy Advisory Committee to the WHO: conclusions and recommendations of September 2013 meeting. Malaria Journal 2013;12:346. [PUBMED: 24359206]

References to other published versions of this review

González 2015

González R, Boerma RS, Sinclair D, Aponte JJ, ter Kuile FO, Menéndez C. Mefloquine for preventing malaria in pregnant women. Cochrane Database of Systematic Reviews 2015, Issue 1. [DOI: 10.1002/14651858.CD011444]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Briand 2009 BEN

Methods

Trial design: open‐label, randomized, 2‐arm trial of 2 doses of IPTp

Follow‐up: the second IPTp dose was administered from 30 weeks of gestation and at least 1 month after administration of the first dose. Women were visited at home, at delivery, and until 6 weeks after the end of pregnancy.

Adverse event (AE) monitoring: AEs were recorded via an open‐labelled questionnaire during visits at home occurring within 1 week after each IPTp intake.

Participants

Numbers of participants randomized: 802 (IPTp‐mefloquine), 799 (IPTp‐sulfadoxine‐pyrimethamine)

Inclusion criteria: HIV‐uninfected women of all gravidities at 16 to 28 weeks of gestation who had no history of a neurological or psychiatric disorder and who had not previously used sulfadoxine‐pyrimethamine or mefloquine nor reported having adverse reactions to medications containing sulfa.

Exclusion criteria: pregnant women not meeting inclusion criteria.

Interventions

  • Two doses of IPTp with sulfadoxine‐pyrimethamine (1500 mg of sulfadoxine and 75 mg of pyrimethamine per dose)

  • Two doses of IPTp with mefloquine (15 mg/kg per dose; Mepha)

Outcomes

  • Maternal peripheral parasitaemia at delivery

  • Placental malaria (presence of asexual stage parasites in blood smear)

  • Maternal anaemia at delivery (defined by haemoglobin < 10 g/dL)

  • Mean haemoglobin at delivery

  • Clinical malaria episodes during pregnancy

  • Cord blood parasitaemia

  • Mean birth weight

  • Low birth weight rates

  • Prematurity rates

  • Spontaneous abortion (expulsion of a foetus at < 28 weeks of gestation) rates

  • Stillbirth rates (delivery of a dead child at < 28 weeks of gestation)

  • Congenital malformation rates

  • Maternal mortality

  • Neonatal mortality

  • Frequency of adverse events: vomiting, headache, weakness, and dizziness

Notes

Country: Benin

Setting: antenatal care clinics from Ouidah,a semi‐rural town

Transmission: perennial with seasonal peaks

Resistance: in 2005, rates of sulfadoxine‐pyrimethamine and mefloquine resistance in vivo in children < 5 years of age were estimated to be 50% and 2.5% by day 28 of treatment, respectively.

Dates: 2005 to 2008

Funding: Fonds de Solidarité Prioritaire (French Ministry of Foreign Affairs; project no. 2006–22); Institut de Recherche pour le Développement;
Fondation pour la Recherche Médicale (grant FDM20060907976 to V.B.); Fondation de France; and Fondation Mérieux

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quote: "Randomization of subjects was stratified according to maternity clinic and gravidity".

Allocation concealment (selection bias)

High risk

Allocation was not concealed.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No blinding was reported, and safety outcomes are likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Efficacy

Low risk

No blinding of outcome assessment was reported, but the review authors judge that the efficacy outcome measurement is not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Safety

High risk

No blinding of outcome assessment was reported; thus the review authors judge that the safety outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data were balanced in numbers across intervention groups, and similar reasons for missing data were reported across groups.

Selective reporting (reporting bias)

Low risk

The study protocol is not available, but it is clear that published reports describe all expected outcomes, including those that were prespecified.

Other bias

Low risk

The study appears to be free of other sources of bias.

Denoeud‐Ndam 2014a BEN

Methods

Trial design: randomized, open‐label trial of 3 doses of IPTp

Follow‐up: 3 scheduled IPTp administrations with at least a 1‐month interval between them. IPTp‐mefloquine administration and provision of cotrimoxazole. Clinical and adherence information, complete blood count, CD4 count, malaria screening, and treatment of malaria.

At delivery: blood smears from placenta and umbilical cords and evaluation of newborns. Infant evaluation at 6 weeks, 4 months, and 2 months after weaning

Adverse event (AE) monitoring: self‐reporting of all AEs. All adverse events were recorded at each visit. In addition, direct observation of early adverse reactions to mefloquine within 30 minutes after supervised intake was noted and later reactions were collected by phone the same day/evening or on the next day. Medical examination was performed 2 weeks after cotrimoxazole initiation to search for cutaneous reactions. An independent data and safety monitoring board reviewed all SAEs.

Participants

Numbers of participants randomized: 146 (cotrimoxazole), 146 (cotrimoxazole+mefloquine)

Inclusion criteria: HIV‐infected pregnant women of all gravidities aged > 18 years, living permanently in the study area, between 16 and 28 weeks of gestation; last dosage of IPTp taken 1 month before enrolment; women requiring antimalarial treatment enrolled at least 2 weeks after completion of treatment

Exclusion criteria: history of neuropsychiatric disorder; severe kidney or liver disease; serious adverse reaction to mefloquine, sulfa drugs, or quinine

Interventions

IPTp with mefloquine plus cotrimoxazole

  • 15 mg/kg single dose (250 mg tablet, Lariam, Roche), 3 doses 1 month apart

  • Daily dose of 800 mg sulfamethoxazole and 160 mg trimethoprim

Cotrimoxazole

  • Daily dose of 800 mg sulfamethoxazole and 160 mg trimethoprim

All study participants were given LLITNs and daily supplementation with 100 mg ferrous sulphate and 5 mg folic acid.

The first dose was given at ≥ 16 weeks of gestation.

All women were observed for 30 minutes following IPTp administration. Women vomiting within the first 30 minutes were given a second full IPTp dose.

Asymptomatic women and women with low parasitaemia (< 1000 parasites/µL) were treated by the IPTp‐mefloquine dose in the mefloquine groups. Otherwise, women received artemether‐lumefantrine or oral quinine. Those with severe malaria were treated with intravenous quinine.

Outcomes

  • Maternal peripheral parasitaemia at delivery (PCR)

  • Placental parasitaemia at delivery (blood smear and PCR)

  • Mean maternal haemoglobin at delivery

  • Maternal anaemia (< 9.5 g/dL) at delivery

  • Cord blood parasitaemia at delivery

  • Mean birth weight

  • Low birth weight (< 2500 g)

  • Prematurity

  • Serious adverse events (SAEs) during pregnancy

  • Spontaneous abortions (< 28 weeks)

  • Stillbirths (≥ 28 weeks of gestation)

  • Congenital malformations (< 28 weeks of gestation)

  • Early neonatal mortality (< 7 days)

  • Neonatal mortality

  • Infant deaths after 7 days

  • Vomiting

  • Dizziness

  • Headache

  • Fatigue/weakness

Notes

Country: Benin

Setting: 5 urban hospitals with PMTCT programmes

Malaria transmission: intense and perennial transmission, with peaks during rainy seasons

Resistance: increasing risk of resistance to sulfa drugs. Parasite resistance to cotrimoxazole

Dates: 2009 to 2012

Funding: Sidaction Grant AI19‐3‐01528

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quote: "Randomization was stratified according to the study site and the number of previous pregnancies".

Allocation concealment (selection bias)

Low risk

Quote: "The study coordination center retained the master list and assigned treatment by phone".

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial blinded only the microscopist who evaluated blood smears.

Blinding of outcome assessment (detection bias)
Efficacy

Low risk

No blinding of outcome assessment was reported, but the review authors judge that the efficacy outcome measurement is not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Safety

High risk

No blinding of outcome assessment was reported; thus the review authors judge that the safety outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data were balanced in numbers across groups.

Selective reporting (reporting bias)

Low risk

Protocol was not available, but published report describes all expected outcomes including those prespecified.

Other bias

Low risk

The study appears to be free of other sources of bias.

Denoeud‐Ndam 2014b BEN

Methods

Trial design: randomized, open‐label trial of 3 doses of IPTp

Follow‐up: 3 scheduled IPTp administrations with at least a 1‐month interval between them. IPTp‐mefloquine administration and provision of cotrimoxazole. Clinical and adherence information, complete blood count, CD4 count, malaria screening, and treatment of malaria.

At delivery: blood smears from placenta and umbilical cords and evaluation of newborns. Infant evaluation at 6 weeks, 4 months, and 2 months after weaning

Adverse event (AE) monitoring: self‐reporting of all AEs. All adverse events were recorded at each visit. In addition, direct observation of early adverse reactions to mefloquine within 30 minutes after supervised intake was noted and later reactions were collected by phone the same day/evening or on the next day. Medical examination was performed 2 weeks after cotrimoxazole initiation to search for cutaneous reactions. An independent data and safety monitoring board reviewed all SAEs.

Participants

Numbers of participants randomized: 72 (cotrimoxazole), 68 (mefloquine)

Inclusion criteria: HIV‐infected pregnant women of all gravidities aged > 18 years, living permanently in the study area, between 16 and 28 weeks of gestation, last dosage of IPTp taken 1 month before enrolment, women requiring antimalarial treatment enrolled at least 2 weeks after completion of treatment

Exclusion criteria: history of neuropsychiatric disorder; severe kidney or liver disease; serious adverse reaction to mefloquine, sulfa drugs, or quinine

Interventions

IPTp with mefloquine

  • 15 mg/kg single dose (250 mg tablet, Lariam, Roche)

  • Three doses 1 month apart

Cotrimoxazole

  • Daily dose of 800 mg sulfamethoxazole and 160 mg trimethoprim

All study participants were given LLITNs and daily supplementation with 100 mg ferrous sulphate and 5 mg folic acid.

The first dose was given at ≥ 16 weeks of gestation.

All women were observed for 30 minutes following IPTp administration. Women vomiting within the first 30 minutes were given a second full IPTp dose.

Asymptomatic women and women with low parasitaemia (< 1000 parasites/µL) in the mefloquine groups were treated by the IPTp‐mefloquine dose. Otherwise, women received artemether‐lumefantrine or oral quinine. Thos with severe malaria were treated with intravenous quinine.

Outcomes

  • Maternal peripheral parasitaemia at delivery (PCR)

  • Placental parasitaemia at delivery (blood smear and PCR)

  • Mean maternal haemoglobin at delivery

  • Maternal anaemia (< 9.5 g/dL) at delivery

  • Cord blood parasitaemia at delivery

  • Mean birth weight

  • Low birth weight (< 2500 g)

  • Prematurity

  • Serious adverse events (SAEs) during pregnancy

  • Spontaneous abortions (< 28 weeks)

  • Stillbirths (≥ 28 weeks of gestation)

  • Congenital malformations (< 28 weeks of gestation)

  • Early neonatal mortality (< 7 days)

  • Neonatal mortality

  • Infant deaths after 7 days

  • Vomiting

  • Dizziness

  • Headache

  • Fatigue/weakness

Notes

Country: Benin

Setting: 5 urban hospitals with PMTCT programmes

Malaria transmission: intense and perennial transmission, with peaks during rainy seasons

Resistance: increasing risk of resistance to sulfa drugs. Parasite resistance to cotrimoxazole

Dates: 2009 to 2012

Funding: Sidaction Grant AI19‐3‐01528

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

Quote: "Randomization was stratified according to the study site and the number of previous pregnancies".

Allocation concealment (selection bias)

Low risk

Quote: "The study coordination center retained the master list and assigned treatment by phone".

Blinding of participants and personnel (performance bias)
All outcomes

High risk

The trial blinded only the microscopist who evaluated blood smears.

Blinding of outcome assessment (detection bias)
Efficacy

Low risk

No blinding of outcome assessment was reported, but the review authors judge that the efficacy outcome measurement is not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Safety

High risk

No blinding of outcome assessment was reported; thus the review authors judge that the safety outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

Missing outcome data were balanced in numbers across groups.

Selective reporting (reporting bias)

Low risk

Protocol was not available, but published report describes all expected outcomes including those prespecified.

Other bias

Low risk

The study appears to be free of other sources of bias.

Gonzalez 2014a BEN GAB MOZ TAN

Methods

Trial design: open‐label, randomized, 3‐arm trial of 2 doses of IPTp

Follow‐up: at each scheduled and unscheduled visit, a standardized symptom questionnaire was completed, as were blood smears for malaria parasites, and haemoglobin if symptoms and/or signs were suggestive of malaria. At delivery, blood samples were collected for haematological and parasitological evaluation. Weighting of newborns and gestational age at birth were recorded. Malaria parasite was determined 6 weeks after the end of pregnancy.

Adverse event monitoring: home visits by field workers were done 2 days after IPTp administration to assess drug tolerability.

Solicited and unsolicited adverse events (AEs) were assessed. The former were assessed by directed questioning regarding malaria‐related signs and symptoms during unscheduled visits, whereas the latter were assessed through open questioning during scheduled visits.

Participants

Numbers of participants randomized: 1578 (sulfadoxine‐pyrimethamine), 1580 (mefloquine full dose), 1591 (mefloquine split)

Inclusion criteria: HIV‐uninfected women of all gravidities attending the antenatal care clinic for the first time, did not receive IPTp during current pregnancy, permanent residence in the study area, gestational age of ≤ 28 weeks

Exclusion criteria: HIV‐positive; history of allergy to sulfa drugs or mefloquine; history of severe renal, hepatic, psychiatric, or neurological disease; mefloquine or halofantrine treatment in the preceding 4 weeks; participating in other intervention studies

Interventions

IPTp with sulfadoxine‐pyrimethamine, 3 tablets

  • 500 mg/25 mg

  • Two doses 1 month apart

IPTp with mefloquine

  • 15 mg/kg given once as a full dose (250‐mg tablets)

  • Two doses 1 month apart

IPTp with mefloquine (split dose)

  • 15 mg/kg given as a split dose over 2 days (250‐mg tablets)

  • Two doses 1 month apart

All study participants were given LLITNs.

The first dose was given at > 13 weeks of gestation.

All women were observed for 60 minutes following IPT administration. Women vomiting within the first 30 minutes were given a second full IPT dose, and those vomiting 30 to 60 minutes after drug intake were given a half replacement dose.

Uncomplicated malaria episodes were treated with oral quinine (first trimester) or artemether‐lumefantrine (second and third trimesters); severe malaria episodes were treated with parenteral quinine.

Outcomes

  • Maternal peripheral parasitaemia at delivery

  • Placental parasitaemia at delivery

  • Mean maternal haemoglobin at delivery

  • Maternal anaemia (< 10 g/dL) at delivery

  • Clinical malaria episodes during pregnancy

  • Cord blood parasitaemia at delivery

  • Cord blood anaemia

  • Mean birth weight

  • Low birth weight (< 2500 g)

  • Low birth weight by gravidity

  • Prematurity

  • Malaria in first year of life

  • Hospital admissions in first year of life

  • Malaria in first year of life (infant morbidity)

  • Hospital admissions in first year of life (infant morbidity)

  • Serious adverse events (SAEs) during pregnancy

  • Spontaneous abortions (< 20 complete weeks of gestation)

  • Stillbirths (> 20 complete weeks of gestation)

  • Congenital malformations

  • Maternal mortality

  • Neonatal mortality

  • Infant mortality

  • Vomiting

  • Headache

  • Fatigue/weakness

  • Dizziness

Notes

Country: Tanzania, Mozambique, Benin, and Gabon

Setting: antenatal care clinics

Transmission: mesoendemic in Tanzania and Mozambique, hyperendemic in Benin and Gabon

Resistance: resistance to sulfadoxine‐pyrimethamine due to long‐term sulfadoxine‐pyrimethamine for IPTp

Dates: 2009 to 2013

Funding: this study was funded by the European Developing Countries Clinical Trials Partnership (EDCTP; IP.2007.31080.002), the Malaria in Pregnancy Consortium, and the Instituto de Salud Carlos III (PI08/0564), in Spain. RG and MR were partially supported by grants from the Spanish Ministry of Health (ref. CM07/0015 and CM11/00278, respectively). The CISM receives core funding from the Spanish Agency for International Cooperation (AECID). LLITNs (Permanet) were donated by Vestergaard Frandsen.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The allocation of the participants to the study arms was done centrally by randomization stratified by country according to a 1:1:1 scheme. The sponsor’s institution biostatistician produced the computer‐generated randomization list for each recruiting site".

Allocation concealment (selection bias)

Low risk

Quote: "Treatment allocation for each participant was concealed in opaque sealed envelopes that were opened only after recruitment. Study participants were assigned a unique study number linked to the allocated treatment group".

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Quote: "The study was designed as an open‐label, randomized, three‐arm trial to compare two‐dose mefloquine with two‐dose SP for IPTp".

Blinding of outcome assessment (detection bias)
Efficacy

Low risk

No blinding of outcome assessment was reported, but the review authors judge that the efficacy outcome measurement is not likely to be influenced by lack of blinding.

Blinding of outcome assessment (detection bias)
Safety

High risk

No blinding of outcome assessment was reported; thus the review authors judge that the safety outcome measurement is likely to be influenced by lack of blinding.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All excluded participants, at any stage of the trial, are counted in the flow chart (both ITT and ATP cohorts). Missing outcome data were balanced in numbers across groups.

Selective reporting (reporting bias)

Low risk

Not observed. Protocol available

Other bias

Low risk

The study appears to be free of other sources of bias.

Gonzalez 2014b KEN MOZ TAN

Methods

Trial design: individually randomized, double‐blind, placebo‐controlled trial of 3 doses of IPTp

Follow‐up: at each scheduled and unscheduled visit, a standardized symptom questionnaire was completed, as were blood smears for malaria parasites, and haemoglobin if symptoms and/or signs were suggestive of malaria. On a monthly basis, adherence to cotrimoxazole and LLITN was assessed. At delivery, blood samples were collected for haematological and parasitological evaluation with CD4 cell count and HIV viral load. Weighting of newborns and gestational age at birth were recorded. Malaria parasite was determined 6 weeks after the end of pregnancy.

Adverse event monitoring: home visits by field workers were done 2 days after IPTp administration to assess drug tolerability.

Solicited and unsolicited adverse events (AEs) were assessed. The former were assessed by directed questioning of malaria‐related signs and symptoms during unscheduled visits, whereas the latter were assessed through open questioning during scheduled visits.

Participants

Numbers of participants randomized: 537 (placebo+cotrimoxazole), 534 (mefloquine+cotrimoxazole)

Inclusion criteria: HIV‐infected women of all gravidities attending the antenatal care clinic for the first time, did not receive IPTp during current pregnancy, permanent residence in the study area, gestational age of ≤ 28 weeks, HIV positive

Exclusion criteria: history of allergy to sulfa drugs or mefloquine; history of severe renal, hepatic, psychiatric, or neurological disease; mefloquine or halofantrine treatment in the preceding 4 weeks; participating in other intervention studies

Interventions

IPTp with mefloquine

  • 15 mg/kg single dose (maximum dosage would not exceed 1500 mg of mefloquine)

  • Three doses 1 month apart

IPTp with placebo

  • Identical to mefloquine tablets in shape and colour

  • Three doses 1 month apart

All study participants had monthly cotrimoxazole prophylaxis (fixed combination 800 mg of trimethroprim and 160 mg of sulfamethoxazole/tablet).

All study participants were given LLITNs.

The first dose was given at > 13 weeks of gestation.

All women were observed for 60 minutes following IPT administration. Women vomiting within the first 30 minutes were given a second full IPTp dose, and those vomiting 30 to 60 minutes after drug intake were given a half replacement dose.

Uncomplicated malaria episodes were treated with oral quinine (first trimester) or artemether‐lumefantrine (second and third trimesters); severe malaria episodes were treated with parenteral quinine.

Outcomes

  • Maternal peripheral parasitaemia at delivery (PCR)

  • Placental parasitaemia at delivery (blood smear and PCR)

  • Mean maternal haemoglobin at delivery

  • Maternal anaemia (< 9.5 g/dL) at delivery

  • Clinical malaria episodes during pregnancy

  • Cord blood parasitaemia at delivery

  • Mean birth weight

  • Low birth weight (< 2500 g)

  • Prematurity

  • Serious adverse events (SAEs) during pregnancy

  • Spontaneous abortions (< 20 complete weeks of gestation)

  • Stillbirths (> 20 weeks of gestation)

  • Congenital malformations

  • Maternal mortality

  • Perinatal mortality

  • Early neonatal mortality (< 7 days)

  • Neonatal mortality

  • Vomiting

  • Headache

  • Fatigue/weakness

  • Dizziness

Notes

Countries: Tanzania, Mozambique, and Kenya

Setting: antenatal care clinics

Transmission: mesoendemic in Tanzania and Mozambique, holoendemic in Kenya

Resistance: resistance to sulfadoxine‐pyrimethamine due to long‐term sulfadoxine‐pyrimethamine for IPTp

Dates: 2010 to 2013

Funding: this study was funded by the European Developing Countries Clinical Trials Partnership (EDCTP; IP.2007.31080.002), the Malaria in Pregnancy Consortium, and the Instituto de Salud Carlos III (PI08/0564), in Spain. RG and MR were partially supported by grants from the Spanish Ministry of Health (ref. CM07/0015 and CM11/00278, respectively). The CISM receives core funding from the Spanish Agency for international Cooperation (AECID). LLITNs (Permanet) were donated by Vestergaard Frandsen, and cotrimoxazole tablets (Septrin) by UCB Pharma, in Spain.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Quote: "The allocation of the participants to the study arms was done centrally by block randomization (block size of 6) stratified by country".

Allocation concealment (selection bias)

Low risk

Quote: "The Pharmacy Department of the Hospital Clinic in Barcelona produced and safeguarded the computer‐generated randomization list for each recruiting site until unblinding, and carried out the masking, labelling, and packaging of all study interventional drugs. Study number allocation for each participant was concealed in opaque sealed envelopes that were sequentially numbered and opened only after recruitment by study health personnel".

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Quote: "Study participants were assigned a unique study
number linked to the allocated treatment group. Investigators, laboratory staff, care providers, and study participants were blinded to intervention throughout the study".

Placebo tablets were identical to mefloquine tables in shape and colour.

Blinding of outcome assessment (detection bias)
Efficacy

Low risk

Quote: "Investigators, laboratory staff, care providers, and study participants were blinded to intervention throughout the study".

Blinding of outcome assessment (detection bias)
Safety

Low risk

Quote: "Investigators, laboratory staff, care providers, and study participants were blinded to intervention throughout the study".

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All excluded participants, at any stage of the trial, are counted in the flow chart (both ITT and ATP cohorts). Missing outcome data were balanced in numbers across groups.

Selective reporting (reporting bias)

Low risk

Not observed. Protocol available

Other bias

Low risk

The study appears to be free of other sources of bias.

Nosten 1994 THA

Methods

Trial design: double‐blind, placebo‐controlled trial. Phase 1 and phase 2

Follow‐up: in both phases, weekly visits included assessment of weight, temperature, pulse, blood pressure, fundal height, presence of oedema and anaemia, a symptom questionnaire on gastrointestinal and central nervous system side effects, malaria blood smear, electrocardiogram, and haematology and biochemistry every 2 weeks. Treatment of malaria and anaemia and food supply were provided when needed. At phase 2, expanded questionnaires and Romberg test were used. At delivery, measurement of newborn weight, details of labour, cord and maternal blood samples (malaria and anaemia), and placental biopsy were included. At phase 2, autopsy of death was performed in newborns. Follow‐up consisted of different measurements in children until 2 years of age (weight, height, head and arm circumferences) and determination of age when baby could first crawl, sit, walk, and talk. At phase 2, age at first symptomatic malaria, malaria blood smear, haematocrit, and full clinical examination were performed.

Adverse event monitoring: weekly symptom questionnaire focusing on gastrointestinal, neurological, dermatological, and systemic symptoms

Participants

Numbers of participants randomized: 170 (mefloquine ‐ 60 phase 1, 110 phase 2), 169 (placebo ‐ 59 phase 1, 110 phase 2)

Inclusion criteria: women of all gravidities and unknown HIV status (not tested) who attended the ANC clinic and were at > 20 weeks of estimated gestation.

Exclusion criteria: women not meeting inclusion criteria.

Interventions

IPTp with mefloquine

  • Phase 1: 500 mg of base loading dose followed by 250 mg weekly for 4 weeks and thereafter 125 mg weekly until delivery

  • Phase 2: 250 mg of base weekly given for 4 weeks followed by 125 mg weekly until delivery

IPTp with placebo

  • Identical to mefloquine tablets (weekly dosage)

The first dose was given at > 20 weeks of gestation.

Anaemia was treated with ferrous sulphate and folic acid. Uncomplicated Plasmodium falciparum malaria was treated with quinine sulphate, P vivax with chloroquine sulphate, and severe malaria with intravenous quinine dihydrochloride.

Outcomes

  • Maternal peripheral parasitaemia during pregnancy

  • Placental malaria

  • Mean birth weight

  • Low birth weight

  • Prematurity

  • Stillbirths

  • Spontaneous abortions

  • Congenital malformations

  • Maternal mortality

  • Infant mortality

Notes

Country: Thailand

Setting: 3 camps for displaced people: phase 1 antenatal clinics, phase 2 hospital

Dates: 1987 to 1990

Transmission: seasonal malaria transmission (mesoendemic)

Resistance: resistances to mefloquine, quinine, chloroquine, and antifolates

Funding: United Nations Development Programme/World Bank/World Health Organization Special Programme for Research and Training in Tropical Diseases; Wellcome Trust of Great Britain; Prevention Fundation

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Women were randomized to receive mefloquine or placebo. Not well explained how women were randomized

Allocation concealment (selection bias)

Unclear risk

Not explained

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Double‐blind trial

Quote: "The investigators were unaware of the randomization".

Placebo tablets were identical to mefloquine tablets.

Blinding of outcome assessment (detection bias)
Efficacy

Unclear risk

Not explained

Blinding of outcome assessment (detection bias)
Safety

Unclear risk

Not explained

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All excluded participants and those who decided to drop out are correctly reported along with reasons. Missing outcome data were balanced in numbers across groups.

Selective reporting (reporting bias)

Unclear risk

Results of cord and maternal blood smears are not shown (published elsewhere?). No protocol is available. Nothing else was observed.

Other bias

Low risk

The study appears to be free of other sources of bias.

Abbreviations: AE: adverse event; AECID: Spanish Agency for International Cooperation; ANC: antenatal care; ATP: adenosine triphosphate; CISM: Centro de Investigação em Saúde da Manhiça; IPTp: intermittent preventive treatment for malaria in pregnancy; IPTp‐mefloquine: intermittent preventive mefloquine treatment in pregnancy; IPTp‐sulfadoxine‐pyrimethamine: intermittent preventive sulfadoxine‐pyrimethamine treatment in pregnancy; ITT: intention‐to‐treat; LLITN: long‐lasting insecticide‐treated net; PCR: polymerase chain reaction; PMTCT: prevention of mother‐to‐child transmission; SAE: serious adverse event.

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Balocco 1992

Letter to editor reporting on the results of pregnancy of 24 women exposed to mefloquine in early pregnancy. The report was excluded because it did not meet the inclusion criteria.

Briand 2015

This publication reports the findings of a re‐analysis of previous published data comparing mefloquine with sulphadoxine‐pyrimethamine for IPTp in Benin using a multiple outcome approach, which allowed the joint assessment of efficacy and tolerability. This analysis was not included in the review because the original study (Briand 2009 BEN) was already included and it did not add additional data.

Denoeud‐Ndam 2012

Study comparing mefloquine tolerability as IPTp between HIV‐infected and uninfected women participating in three included trials from Benin (Briand 2009 BEN and Denoeud‐Ndam 2014a and b). This analysis was excluded from the review because it did not provide additional data from already included trials.

Nosten 1990 THA

The study was designed as a dose‐finding pharmacokinetic study in 20 pregnant women in the third trimester of pregnancy who received mefloquine as prophylaxis. The trial did not compare the safety and efficacy of mefloquine with another antimalarial drug and thus, it did not meet inclusion criteria.

Phillips‐Howard 1998

Publication reporting on a data analysis of reported use of mefloquine during the 1st trimester of pregnancy in European travellers. This analysis was excluded from the review because it did not meet inclusion criteria.

Schlagenhauf 2012

This publication presents the analysis of the reports of exposure to mefloquine in pregnancy received by the Roche post‐marketing surveillance system. This analysis was excluded from the review because it did not meet inclusion criteria.

Smoak 1997

This publication reports a case series of 72 US soldiers who inadvertently took mefloquine during pregnancy for prophylaxis. This publication was excluded from the review because it did not meet inclusion criteria.

Steketee 1996 MAL

We were not convinced that allocation was unbiased.

Quote: "The assignment of regimens was based on the clinic day of enrolment. All women making their first antenatal clinic visit on a given day were assigned to the same regimen; the following clinic day, enrolled women were assigned a different regimen".

We noted bias in allocation supported by statistically and clinically significant differences between intervention groups (3 groups under different chloroquine regimens versus 1 group under mefloquine regimen).

Vanhauwere 1998

Study evaluating 1627 reports of mefloquine exposure pregnancy, mainly for chemoprophylaxis received by the Roche Post‐marketing surveillance system between 1986 and 1996.This analysis was excluded from the review because it did not meet inclusion criteria.

Characteristics of ongoing studies [ordered by study ID]

Akinyotu 2015 NIG

Trial name or title

A comparative study of mefloquine and SP as prophylaxis against malaria in pregnant HIV‐infected patients

Methods

Allocation: randomized

Intervention model: parallel assignment

Masking: single‐blind (outcomes assessor)

Primary purpose: prevention

Participants

Inclusion criteria:

  • Pregnant HIV‐infected patients

  • Gestational age ≥ 16 weeks

  • No history of use of mefloquine or sulphadoxine

  • Pyrimethamine 4 weeks before recruitment

Exclusion criteria:

  • Anaemia packed cell volume < 30%

  • Pre‐existing medical conditions ‐ diabetes mellitus, hypertension

  • Allergy to sulphadoxine‐pyrimethamine or mefloquine

  • Non‐consenting patients

  • Multiple gestation

  • Known psychiatric illness

  • Known seizure disorder

  • History of severe renal or hepatic disease

Interventions

  • Mefloquine: 250 mg 3 doses 4 weeks apart

  • Sulfadoxine‐pyrimethamine: 500 mg sulphadoxine and 25 mg pyrimethamine, 3 tablets 4 weeks apart for 3 doses

Outcomes

No information available

Starting date

September 2015

Contact information

Oriyomi O Akinyotu, MBBS; Ibadan: +2348035044590; [email protected]

Notes

We contacted the study authors, but they could not provide the data to us because the study was part of a thesis not yet defended.

Data and analyses

Open in table viewer
Comparison 1. Mefloquine versus sulfadoxine‐pyrimethamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical malaria episodes during pregnancy Show forest plot

2

Rate Ratio (Fixed, 95% CI)

0.83 [0.65, 1.05]

Analysis 1.1

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 1 Clinical malaria episodes during pregnancy.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 1 Clinical malaria episodes during pregnancy.

2 Maternal peripheral parasitaemia at delivery Show forest plot

2

5455

Risk Ratio (M‐H, Fixed, 95% CI)

0.65 [0.48, 0.86]

Analysis 1.2

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 2 Maternal peripheral parasitaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 2 Maternal peripheral parasitaemia at delivery.

3 Placental malaria Show forest plot

2

4668

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.58, 1.86]

Analysis 1.3

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 3 Placental malaria.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 3 Placental malaria.

4 Mean haemoglobin at delivery Show forest plot

2

5588

Mean Difference (IV, Fixed, 95% CI)

0.10 [0.01, 0.19]

Analysis 1.4

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 4 Mean haemoglobin at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 4 Mean haemoglobin at delivery.

5 Maternal anaemia at delivery Show forest plot

2

5469

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.76, 0.94]

Analysis 1.5

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 5 Maternal anaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 5 Maternal anaemia at delivery.

6 Severe maternal anaemia at delivery Show forest plot

2

5469

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.58, 1.48]

Analysis 1.6

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 6 Severe maternal anaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 6 Severe maternal anaemia at delivery.

7 Cord blood parasitaemia Show forest plot

2

5309

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.13, 1.46]

Analysis 1.7

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 7 Cord blood parasitaemia.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 7 Cord blood parasitaemia.

8 Cord blood anaemia Show forest plot

1

4006

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.87, 1.23]

Analysis 1.8

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 8 Cord blood anaemia.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 8 Cord blood anaemia.

9 Mean birth weight Show forest plot

2

5241

Mean Difference (IV, Fixed, 95% CI)

2.52 [‐25.66, 30.69]

Analysis 1.9

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 9 Mean birth weight.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 9 Mean birth weight.

10 Low birth weight Show forest plot

2

5641

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.78, 1.17]

Analysis 1.10

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 10 Low birth weight.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 10 Low birth weight.

11 Low birth weight by gravidity Show forest plot

2

5641

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.84, 1.13]

Analysis 1.11

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 11 Low birth weight by gravidity.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 11 Low birth weight by gravidity.

11.1 Primigravidae

2

1576

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.80, 1.30]

11.2 Multigravidae

2

4065

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.78, 1.14]

12 Prematurity Show forest plot

2

4640

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.76, 1.40]

Analysis 1.12

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 12 Prematurity.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 12 Prematurity.

13 Malaria in first year of life Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.97 [0.82, 1.15]

Analysis 1.13

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 13 Malaria in first year of life.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 13 Malaria in first year of life.

14 Hospital admissions in first year of life Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.93 [0.75, 1.17]

Analysis 1.14

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 14 Hospital admissions in first year of life.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 14 Hospital admissions in first year of life.

15 SAEs during pregnancy Show forest plot

1

4674

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.81, 1.20]

Analysis 1.15

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 15 SAEs during pregnancy.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 15 SAEs during pregnancy.

16 Stillbirths and abortions Show forest plot

2

6219

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.91, 1.58]

Analysis 1.16

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 16 Stillbirths and abortions.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 16 Stillbirths and abortions.

17 Congenital malformations Show forest plot

2

5931

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.51, 2.37]

Analysis 1.17

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 17 Congenital malformations.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 17 Congenital malformations.

18 Maternal mortality Show forest plot

2

6219

Risk Ratio (M‐H, Random, 95% CI)

2.41 [0.27, 21.23]

Analysis 1.18

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 18 Maternal mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 18 Maternal mortality.

19 Neonatal mortality Show forest plot

2

6134

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.67, 1.43]

Analysis 1.19

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 19 Neonatal mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 19 Neonatal mortality.

20 Infant mortality Show forest plot

1

Rate Ratio (Fixed, 95% CI)

1.00 [0.66, 1.52]

Analysis 1.20

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 20 Infant mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 20 Infant mortality.

21 AEs: vomiting Show forest plot

2

6272

Risk Ratio (M‐H, Fixed, 95% CI)

4.76 [4.13, 5.49]

Analysis 1.21

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 21 AEs: vomiting.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 21 AEs: vomiting.

22 AEs: fatigue/weakness Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

4.62 [1.80, 11.85]

Analysis 1.22

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 22 AEs: fatigue/weakness.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 22 AEs: fatigue/weakness.

23 AEs: dizziness Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

4.21 [3.36, 5.27]

Analysis 1.23

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 23 AEs: dizziness.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 23 AEs: dizziness.

24 AEs: headache Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.25, 1.94]

Analysis 1.24

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 24 AEs: headache.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 24 AEs: headache.

Open in table viewer
Comparison 2. Mefloquine plus cotrimoxazole versus cotrimoxazole

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical malaria episodes during pregnancy Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.76 [0.33, 1.76]

Analysis 2.1

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 1 Clinical malaria episodes during pregnancy.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 1 Clinical malaria episodes during pregnancy.

2 Maternal peripheral parasitaemia at delivery (PCR) Show forest plot

2

989

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.30, 0.93]

Analysis 2.2

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 2 Maternal peripheral parasitaemia at delivery (PCR).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 2 Maternal peripheral parasitaemia at delivery (PCR).

3 Placental malaria (blood smear) Show forest plot

2

1144

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.89]

Analysis 2.3

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

4 Placental malaria (PCR) Show forest plot

2

977

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.14, 0.57]

Analysis 2.4

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 4 Placental malaria (PCR).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 4 Placental malaria (PCR).

5 Mean haemoglobin at delivery Show forest plot

2

1167

Mean Difference (IV, Random, 95% CI)

0.07 [‐0.32, 0.46]

Analysis 2.5

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 5 Mean haemoglobin at delivery.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 5 Mean haemoglobin at delivery.

6 Maternal anaemia at delivery (< 9.5 g/dL) Show forest plot

2

1197

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.73, 1.20]

Analysis 2.6

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 6 Maternal anaemia at delivery (< 9.5 g/dL).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 6 Maternal anaemia at delivery (< 9.5 g/dL).

7 Maternal severe anaemia at delivery Show forest plot

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.41, 2.08]

Analysis 2.7

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 7 Maternal severe anaemia at delivery.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 7 Maternal severe anaemia at delivery.

8 Cord blood parasitaemia Show forest plot

2

1166

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.13]

Analysis 2.8

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 8 Cord blood parasitaemia.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 8 Cord blood parasitaemia.

9 Mean birth weight Show forest plot

2

1220

Mean Difference (IV, Random, 95% CI)

‐25.75 [‐86.99, 35.49]

Analysis 2.9

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 9 Mean birth weight.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 9 Mean birth weight.

10 Low birth weight Show forest plot

2

1220

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.89, 1.60]

Analysis 2.10

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 10 Low birth weight.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 10 Low birth weight.

11 Prematurity Show forest plot

2

824

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.58, 1.96]

Analysis 2.11

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 11 Prematurity.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 11 Prematurity.

12 SAEs during pregnancy Show forest plot

2

1347

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.50, 0.95]

Analysis 2.12

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 12 SAEs during pregnancy.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 12 SAEs during pregnancy.

13 Spontaneous abortions and stillbirths Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.42, 2.98]

Analysis 2.13

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 13 Spontaneous abortions and stillbirths.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 13 Spontaneous abortions and stillbirths.

14 Congenital malformations Show forest plot

2

1312

Risk Ratio (M‐H, Fixed, 95% CI)

0.61 [0.22, 1.67]

Analysis 2.14

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 14 Congenital malformations.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 14 Congenital malformations.

15 Maternal mortality Show forest plot

2

1347

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.13, 2.01]

Analysis 2.15

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 15 Maternal mortality.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 15 Maternal mortality.

16 Neonatal mortality Show forest plot

2

1239

Risk Ratio (M‐H, Fixed, 95% CI)

1.32 [0.65, 2.69]

Analysis 2.16

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 16 Neonatal mortality.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 16 Neonatal mortality.

17 Mother‐to‐child transmission HIV Show forest plot

2

1019

Risk Ratio (M‐H, Fixed, 95% CI)

1.92 [1.13, 3.25]

Analysis 2.17

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 17 Mother‐to‐child transmission HIV.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 17 Mother‐to‐child transmission HIV.

18 AEs: vomiting Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

20.88 [1.40, 311.66]

Analysis 2.18

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 18 AEs: vomiting.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 18 AEs: vomiting.

19 AEs: fatigue/weakness Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

2.95 [0.26, 32.93]

Analysis 2.19

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 19 AEs: fatigue/weakness.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 19 AEs: fatigue/weakness.

20 AEs: dizziness Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

16.34 [0.39, 684.99]

Analysis 2.20

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 20 AEs: dizziness.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 20 AEs: dizziness.

21 AEs: headache Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

0.76 [0.28, 2.10]

Analysis 2.21

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 21 AEs: headache.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 21 AEs: headache.

Open in table viewer
Comparison 3. Mefloquine versus cotrimoxazole

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Maternal peripheral parasitaemia at delivery (PCR) Show forest plot

1

98

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.72]

Analysis 3.1

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 1 Maternal peripheral parasitaemia at delivery (PCR).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 1 Maternal peripheral parasitaemia at delivery (PCR).

2 Placental malaria (PCR) Show forest plot

1

94

Risk Ratio (M‐H, Fixed, 95% CI)

0.73 [0.13, 4.15]

Analysis 3.2

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 2 Placental malaria (PCR).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 2 Placental malaria (PCR).

3 Placental malaria (blood smear) Show forest plot

1

108

Risk Ratio (M‐H, Fixed, 95% CI)

0.35 [0.01, 8.30]

Analysis 3.3

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

4 Mean haemoglobin at delivery Show forest plot

1

100

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.67, 0.47]

Analysis 3.4

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 4 Mean haemoglobin at delivery.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 4 Mean haemoglobin at delivery.

5 Maternal anaemia at delivery (< 9.5 g/dL) Show forest plot

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.26, 3.16]

Analysis 3.5

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 5 Maternal anaemia at delivery (< 9.5 g/dL).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 5 Maternal anaemia at delivery (< 9.5 g/dL).

6 Mean birth weight Show forest plot

1

120

Mean Difference (IV, Fixed, 95% CI)

‐102.0 [‐255.52, 51.52]

Analysis 3.6

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 6 Mean birth weight.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 6 Mean birth weight.

7 Low birth weight Show forest plot

1

120

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.56, 4.13]

Analysis 3.7

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 7 Low birth weight.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 7 Low birth weight.

8 Prematurity Show forest plot

1

125

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.33, 3.56]

Analysis 3.8

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 8 Prematurity.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 8 Prematurity.

9 SAEs during pregnancy Show forest plot

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.28, 4.07]

Analysis 3.9

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 9 SAEs during pregnancy.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 9 SAEs during pregnancy.

10 Stillbirths Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

4.30 [0.49, 37.49]

Analysis 3.10

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 10 Stillbirths.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 10 Stillbirths.

11 Spontaneous abortions Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

1.07 [0.07, 16.84]

Analysis 3.11

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 11 Spontaneous abortions.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 11 Spontaneous abortions.

12 Congenital malformations Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

1.07 [0.16, 7.41]

Analysis 3.12

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 12 Congenital malformations.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 12 Congenital malformations.

13 Maternal mortality Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.13

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 13 Maternal mortality.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 13 Maternal mortality.

14 Neonatal mortality Show forest plot

1

129

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.07, 16.39]

Analysis 3.14

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 14 Neonatal mortality.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 14 Neonatal mortality.

15 Infant deaths after 7 days Show forest plot

1

129

Risk Ratio (M‐H, Fixed, 95% CI)

2.10 [0.19, 22.54]

Analysis 3.15

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 15 Infant deaths after 7 days.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 15 Infant deaths after 7 days.

16 AEs: vomiting Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

13.43 [3.31, 54.54]

Analysis 3.16

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 16 AEs: vomiting.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 16 AEs: vomiting.

17 AEs: fatigue/weakness Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

6.99 [1.64, 29.81]

Analysis 3.17

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 17 AEs: fatigue/weakness.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 17 AEs: fatigue/weakness.

18 AEs: dizziness Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

52.60 [3.26, 848.24]

Analysis 3.18

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 18 AEs: dizziness.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 18 AEs: dizziness.

19 AEs: headache Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.39]

Analysis 3.19

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 19 AEs: headache.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 19 AEs: headache.

Open in table viewer
Comparison 4. Mefloquine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Maternal peripheral parasitaemia during pregnancy Show forest plot

1

339

Risk Ratio (M‐H, Fixed, 95% CI)

0.13 [0.05, 0.33]

Analysis 4.1

Comparison 4 Mefloquine versus placebo, Outcome 1 Maternal peripheral parasitaemia during pregnancy.

Comparison 4 Mefloquine versus placebo, Outcome 1 Maternal peripheral parasitaemia during pregnancy.

2 Placental malaria Show forest plot

1

220

Risk Ratio (M‐H, Fixed, 95% CI)

0.14 [0.01, 2.68]

Analysis 4.2

Comparison 4 Mefloquine versus placebo, Outcome 2 Placental malaria.

Comparison 4 Mefloquine versus placebo, Outcome 2 Placental malaria.

3 Mean birth weight Show forest plot

1

290

Mean Difference (IV, Fixed, 95% CI)

‐80.0 [‐184.65, 24.65]

Analysis 4.3

Comparison 4 Mefloquine versus placebo, Outcome 3 Mean birth weight.

Comparison 4 Mefloquine versus placebo, Outcome 3 Mean birth weight.

4 Low birth weight Show forest plot

1

290

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.78, 2.48]

Analysis 4.4

Comparison 4 Mefloquine versus placebo, Outcome 4 Low birth weight.

Comparison 4 Mefloquine versus placebo, Outcome 4 Low birth weight.

5 Prematurity Show forest plot

1

199

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.15, 1.53]

Analysis 4.5

Comparison 4 Mefloquine versus placebo, Outcome 5 Prematurity.

Comparison 4 Mefloquine versus placebo, Outcome 5 Prematurity.

6 Stillbirths Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [0.86, 8.08]

Analysis 4.6

Comparison 4 Mefloquine versus placebo, Outcome 6 Stillbirths.

Comparison 4 Mefloquine versus placebo, Outcome 6 Stillbirths.

7 Spontaneous abortions Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.04, 5.22]

Analysis 4.7

Comparison 4 Mefloquine versus placebo, Outcome 7 Spontaneous abortions.

Comparison 4 Mefloquine versus placebo, Outcome 7 Spontaneous abortions.

8 Congenital malformations Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

3.82 [0.43, 33.83]

Analysis 4.8

Comparison 4 Mefloquine versus placebo, Outcome 8 Congenital malformations.

Comparison 4 Mefloquine versus placebo, Outcome 8 Congenital malformations.

9 Maternal mortality Show forest plot

1

339

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.12, 71.85]

Analysis 4.9

Comparison 4 Mefloquine versus placebo, Outcome 9 Maternal mortality.

Comparison 4 Mefloquine versus placebo, Outcome 9 Maternal mortality.

10 Infant mortality Show forest plot

1

288

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.63, 1.74]

Analysis 4.10

Comparison 4 Mefloquine versus placebo, Outcome 10 Infant mortality.

Comparison 4 Mefloquine versus placebo, Outcome 10 Infant mortality.

Indicators and impact of malaria infection in mothers and infants.
Figuras y tablas -
Figure 1

Indicators and impact of malaria infection in mothers and infants.

Conceptual framework of malaria chemoprevention. Reproduced under the terms of a Creative Commons Licence from Radeva‐Petrova 2014.
Figuras y tablas -
Figure 2

Conceptual framework of malaria chemoprevention. Reproduced under the terms of a Creative Commons Licence from Radeva‐Petrova 2014.

Study flow diagram.
Figuras y tablas -
Figure 3

Study flow diagram.

‘Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 4

‘Risk of bias' summary: review authors' judgements about each risk of bias item for each included study.

‘Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 5

‘Risk of bias' graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 1 Clinical malaria episodes during pregnancy.
Figuras y tablas -
Analysis 1.1

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 1 Clinical malaria episodes during pregnancy.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 2 Maternal peripheral parasitaemia at delivery.
Figuras y tablas -
Analysis 1.2

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 2 Maternal peripheral parasitaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 3 Placental malaria.
Figuras y tablas -
Analysis 1.3

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 3 Placental malaria.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 4 Mean haemoglobin at delivery.
Figuras y tablas -
Analysis 1.4

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 4 Mean haemoglobin at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 5 Maternal anaemia at delivery.
Figuras y tablas -
Analysis 1.5

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 5 Maternal anaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 6 Severe maternal anaemia at delivery.
Figuras y tablas -
Analysis 1.6

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 6 Severe maternal anaemia at delivery.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 7 Cord blood parasitaemia.
Figuras y tablas -
Analysis 1.7

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 7 Cord blood parasitaemia.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 8 Cord blood anaemia.
Figuras y tablas -
Analysis 1.8

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 8 Cord blood anaemia.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 9 Mean birth weight.
Figuras y tablas -
Analysis 1.9

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 9 Mean birth weight.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 10 Low birth weight.
Figuras y tablas -
Analysis 1.10

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 10 Low birth weight.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 11 Low birth weight by gravidity.
Figuras y tablas -
Analysis 1.11

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 11 Low birth weight by gravidity.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 12 Prematurity.
Figuras y tablas -
Analysis 1.12

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 12 Prematurity.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 13 Malaria in first year of life.
Figuras y tablas -
Analysis 1.13

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 13 Malaria in first year of life.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 14 Hospital admissions in first year of life.
Figuras y tablas -
Analysis 1.14

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 14 Hospital admissions in first year of life.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 15 SAEs during pregnancy.
Figuras y tablas -
Analysis 1.15

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 15 SAEs during pregnancy.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 16 Stillbirths and abortions.
Figuras y tablas -
Analysis 1.16

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 16 Stillbirths and abortions.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 17 Congenital malformations.
Figuras y tablas -
Analysis 1.17

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 17 Congenital malformations.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 18 Maternal mortality.
Figuras y tablas -
Analysis 1.18

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 18 Maternal mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 19 Neonatal mortality.
Figuras y tablas -
Analysis 1.19

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 19 Neonatal mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 20 Infant mortality.
Figuras y tablas -
Analysis 1.20

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 20 Infant mortality.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 21 AEs: vomiting.
Figuras y tablas -
Analysis 1.21

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 21 AEs: vomiting.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 22 AEs: fatigue/weakness.
Figuras y tablas -
Analysis 1.22

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 22 AEs: fatigue/weakness.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 23 AEs: dizziness.
Figuras y tablas -
Analysis 1.23

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 23 AEs: dizziness.

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 24 AEs: headache.
Figuras y tablas -
Analysis 1.24

Comparison 1 Mefloquine versus sulfadoxine‐pyrimethamine, Outcome 24 AEs: headache.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 1 Clinical malaria episodes during pregnancy.
Figuras y tablas -
Analysis 2.1

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 1 Clinical malaria episodes during pregnancy.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 2 Maternal peripheral parasitaemia at delivery (PCR).
Figuras y tablas -
Analysis 2.2

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 2 Maternal peripheral parasitaemia at delivery (PCR).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).
Figuras y tablas -
Analysis 2.3

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 4 Placental malaria (PCR).
Figuras y tablas -
Analysis 2.4

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 4 Placental malaria (PCR).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 5 Mean haemoglobin at delivery.
Figuras y tablas -
Analysis 2.5

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 5 Mean haemoglobin at delivery.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 6 Maternal anaemia at delivery (< 9.5 g/dL).
Figuras y tablas -
Analysis 2.6

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 6 Maternal anaemia at delivery (< 9.5 g/dL).

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 7 Maternal severe anaemia at delivery.
Figuras y tablas -
Analysis 2.7

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 7 Maternal severe anaemia at delivery.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 8 Cord blood parasitaemia.
Figuras y tablas -
Analysis 2.8

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 8 Cord blood parasitaemia.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 9 Mean birth weight.
Figuras y tablas -
Analysis 2.9

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 9 Mean birth weight.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 10 Low birth weight.
Figuras y tablas -
Analysis 2.10

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 10 Low birth weight.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 11 Prematurity.
Figuras y tablas -
Analysis 2.11

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 11 Prematurity.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 12 SAEs during pregnancy.
Figuras y tablas -
Analysis 2.12

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 12 SAEs during pregnancy.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 13 Spontaneous abortions and stillbirths.
Figuras y tablas -
Analysis 2.13

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 13 Spontaneous abortions and stillbirths.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 14 Congenital malformations.
Figuras y tablas -
Analysis 2.14

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 14 Congenital malformations.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 15 Maternal mortality.
Figuras y tablas -
Analysis 2.15

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 15 Maternal mortality.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 16 Neonatal mortality.
Figuras y tablas -
Analysis 2.16

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 16 Neonatal mortality.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 17 Mother‐to‐child transmission HIV.
Figuras y tablas -
Analysis 2.17

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 17 Mother‐to‐child transmission HIV.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 18 AEs: vomiting.
Figuras y tablas -
Analysis 2.18

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 18 AEs: vomiting.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 19 AEs: fatigue/weakness.
Figuras y tablas -
Analysis 2.19

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 19 AEs: fatigue/weakness.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 20 AEs: dizziness.
Figuras y tablas -
Analysis 2.20

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 20 AEs: dizziness.

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 21 AEs: headache.
Figuras y tablas -
Analysis 2.21

Comparison 2 Mefloquine plus cotrimoxazole versus cotrimoxazole, Outcome 21 AEs: headache.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 1 Maternal peripheral parasitaemia at delivery (PCR).
Figuras y tablas -
Analysis 3.1

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 1 Maternal peripheral parasitaemia at delivery (PCR).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 2 Placental malaria (PCR).
Figuras y tablas -
Analysis 3.2

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 2 Placental malaria (PCR).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).
Figuras y tablas -
Analysis 3.3

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 3 Placental malaria (blood smear).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 4 Mean haemoglobin at delivery.
Figuras y tablas -
Analysis 3.4

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 4 Mean haemoglobin at delivery.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 5 Maternal anaemia at delivery (< 9.5 g/dL).
Figuras y tablas -
Analysis 3.5

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 5 Maternal anaemia at delivery (< 9.5 g/dL).

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 6 Mean birth weight.
Figuras y tablas -
Analysis 3.6

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 6 Mean birth weight.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 7 Low birth weight.
Figuras y tablas -
Analysis 3.7

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 7 Low birth weight.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 8 Prematurity.
Figuras y tablas -
Analysis 3.8

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 8 Prematurity.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 9 SAEs during pregnancy.
Figuras y tablas -
Analysis 3.9

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 9 SAEs during pregnancy.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 10 Stillbirths.
Figuras y tablas -
Analysis 3.10

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 10 Stillbirths.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 11 Spontaneous abortions.
Figuras y tablas -
Analysis 3.11

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 11 Spontaneous abortions.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 12 Congenital malformations.
Figuras y tablas -
Analysis 3.12

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 12 Congenital malformations.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 13 Maternal mortality.
Figuras y tablas -
Analysis 3.13

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 13 Maternal mortality.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 14 Neonatal mortality.
Figuras y tablas -
Analysis 3.14

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 14 Neonatal mortality.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 15 Infant deaths after 7 days.
Figuras y tablas -
Analysis 3.15

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 15 Infant deaths after 7 days.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 16 AEs: vomiting.
Figuras y tablas -
Analysis 3.16

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 16 AEs: vomiting.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 17 AEs: fatigue/weakness.
Figuras y tablas -
Analysis 3.17

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 17 AEs: fatigue/weakness.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 18 AEs: dizziness.
Figuras y tablas -
Analysis 3.18

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 18 AEs: dizziness.

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 19 AEs: headache.
Figuras y tablas -
Analysis 3.19

Comparison 3 Mefloquine versus cotrimoxazole, Outcome 19 AEs: headache.

Comparison 4 Mefloquine versus placebo, Outcome 1 Maternal peripheral parasitaemia during pregnancy.
Figuras y tablas -
Analysis 4.1

Comparison 4 Mefloquine versus placebo, Outcome 1 Maternal peripheral parasitaemia during pregnancy.

Comparison 4 Mefloquine versus placebo, Outcome 2 Placental malaria.
Figuras y tablas -
Analysis 4.2

Comparison 4 Mefloquine versus placebo, Outcome 2 Placental malaria.

Comparison 4 Mefloquine versus placebo, Outcome 3 Mean birth weight.
Figuras y tablas -
Analysis 4.3

Comparison 4 Mefloquine versus placebo, Outcome 3 Mean birth weight.

Comparison 4 Mefloquine versus placebo, Outcome 4 Low birth weight.
Figuras y tablas -
Analysis 4.4

Comparison 4 Mefloquine versus placebo, Outcome 4 Low birth weight.

Comparison 4 Mefloquine versus placebo, Outcome 5 Prematurity.
Figuras y tablas -
Analysis 4.5

Comparison 4 Mefloquine versus placebo, Outcome 5 Prematurity.

Comparison 4 Mefloquine versus placebo, Outcome 6 Stillbirths.
Figuras y tablas -
Analysis 4.6

Comparison 4 Mefloquine versus placebo, Outcome 6 Stillbirths.

Comparison 4 Mefloquine versus placebo, Outcome 7 Spontaneous abortions.
Figuras y tablas -
Analysis 4.7

Comparison 4 Mefloquine versus placebo, Outcome 7 Spontaneous abortions.

Comparison 4 Mefloquine versus placebo, Outcome 8 Congenital malformations.
Figuras y tablas -
Analysis 4.8

Comparison 4 Mefloquine versus placebo, Outcome 8 Congenital malformations.

Comparison 4 Mefloquine versus placebo, Outcome 9 Maternal mortality.
Figuras y tablas -
Analysis 4.9

Comparison 4 Mefloquine versus placebo, Outcome 9 Maternal mortality.

Comparison 4 Mefloquine versus placebo, Outcome 10 Infant mortality.
Figuras y tablas -
Analysis 4.10

Comparison 4 Mefloquine versus placebo, Outcome 10 Infant mortality.

Summary of findings for the main comparison. Mefloquine compared with sulfadoxine‐pyrimethamine for preventing malaria in pregnant women

Mefloquine compared with sulfadoxine‐pyrimethamine for preventing malaria in pregnant women

Patient or population: HIV‐uninfected pregnant women
Setting: Benin, Gabon, Mozambique, and Tanzania
Intervention: mefloquine
Comparison: sulfadoxine‐pyrimethamine

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(trials)

Certainty of the evidence
(GRADE)

Comments (compared with sulfadoxine‐pyrimethamine)

Risk with sulfadoxine‐pyrimethamine

Risk with mefloquine

Clinical malaria episodes during pregnancy

IRR 0.83
(0.65 to 1.05)


(2 RCTs)

⊕⊕⊕⊝
HIGHa

Mefloquine results in little or no difference in the incidence of clinical malaria episodes during pregnancy

Maternal peripheral parasitaemia at delivery

43 per 1000

28 per 1000

(20 to 37)

RR 0.65

(0.48 to
0.86)

5455

(2 RCTs)

⊕⊕⊕⊝
HIGHa

Mefloquine results in lower maternal peripheral parasitaemia at delivery

Placental malaria

52 per 1000

54 per 1000
(30 to 97)

RR 1.04
(0.58 to 1.86)

4668
(2 RCTs)

⊕⊕⊝⊝
LOWa,b,c

Due to imprecision and heterogeneity

Mefloquine may result in little or no difference in placental parasitaemia

Maternal anaemia at delivery

219 per 1000

184 per 1000
(166 to 206)

RR 0.84
(0.76 to 0.94)

5469
(2 RCTs)

⊕⊕⊕⊝
MODERATEa,d

Due to imprecision

Mefloquine probably results in fewer women anaemic at delivery

Low birth weight

117 per 1000

111 per 1000
(91 to 137)

RR 0.95
(0.78 to 1.17)

5641
(2 RCTs)

⊕⊕⊕⊝
HIGHa

Mefloquine results in little or no difference in low birth weight

Stillbirths and abortions

31 per 1000

37 per 1000
(28 to 49)

RR 1.20
(0.91 to 1.58)

6219
(2 RCTs)

⊕⊕⊕⊝
HIGHa

Mefloquine results in little or no difference in stillbirths or abortions

AEs: vomiting

82 per 1000

390 per 1000
(338 to 449)

RR 4.76
(4.13 to 5.49)

6272
(2 RCTs)

⊕⊕⊕⊕
HIGHa

Mefloquine results in a four‐fold increase in vomiting

AEs: dizziness

94 per 1000

396 per 1000
(316 to 496)

RR 4.21
(3.36 to 5.27)

6272
(2 RCTs)

⊕⊕⊕⊝
HIGHa,b

Mefloquine results in a four‐fold increase in dizziness

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
Abbreviations: CI: confidence interval; IRR: incidence rate ratio; RCT: randomized controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

aAlthough one trial has serious risk of bias, the other is of high certainty and exclusion of the smaller trial has little effect on the estimate of effect.
bDowngraded by 1 for imprecision: Confidence intervals range from considerable benefit to considerable harm.
cDowngraded by 1 for heterogeneity: Substantive qualitative heterogeneity is evident in the meta‐analysis.
dConfidence intervals include little or no important difference to a 24% reduction in anaemic women. The estimate of 16% is judged to be clinically important.

Figuras y tablas -
Summary of findings for the main comparison. Mefloquine compared with sulfadoxine‐pyrimethamine for preventing malaria in pregnant women
Summary of findings 2. Mefloquine plus cotrimoxazole compared with cotrimoxazole for preventing malaria in pregnant women

Mefloquine plus cotrimoxazole compared with cotrimoxazole for preventing malaria in pregnant women

Patient or population: HIV‐infected pregnant women
Setting: Benin, Kenya, Mozambique, and Tanzania
Intervention: mefloquine plus cotrimoxazole
Comparison: cotrimoxazole

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Number of participants
(trials)

Certainty of the evidence
(GRADE)

Comments (compared with cotrimoxazole)

Risk with cotrimoxazole

Risk with mefloquine plus cotrimoxazole

Clinical malaria episodes during pregnancy

IRR 0.76 (0.33 to 1.76)

(1 RCT)

⊕⊕⊕⊕
HIGH

Mefloquine results in little or no difference in the incidence of clinical malaria episodes during pregnancy

Maternal peripheral parasitaemia at delivery (PCR)

66 per 1000

34 per 1000

(20 to 62)

RR 0.52

(0.30 to 0.93)

989

(2 RCTs)

⊕⊕⊕⊝
MODERATEa

Mefloquine probably results in lower maternal peripheral parasitaemia at delivery

Placental malaria (PCR)

68 per 1000

19 per 1000
(10 to 39)

RR 0.28
(0.14 to 0.57)

977
(2 RCTs)

⊕⊕⊕⊕
HIGHa

Mefloquine plus cotrimoxazole results in fewer women with placental malaria at delivery

Maternal anaemia at delivery

178 per 1000

168 per 1000
(130 to 214)

RR 0.94
(0.73 to 1.20)

1197
(2 RCTs)

⊕⊕⊕⊝
MODERATEa

Mefloquine plus cotrimoxazole probably results in little or no difference in maternal anaemia cases at delivery

Low birth weight

118 per 1000

141 per 1000
(105 to 188)

RR 1.20
(0.89 to 1.60)

1220
(2 RCTs)

⊕⊕⊕⊝
MODERATEa

Mefloquine plus cotrimoxazole probably results in little or no difference in low birth weight

Spontaneous abortions and stillbirths

50 per 1000

56 per 1000
(21 to 149)

RR 1.12
(0.42 to 2.98)

1347
(2 RCTs)

⊕⊝⊝⊝
VERY LOWa,b,c

Mefloquine plus cotrimoxazole may result in little or no difference in spontaneous abortions and stillbirths

AEs: vomiting

30 per 1000

239 per 1000

(144 to 396)

RR 7.95

(4.79 to 13.18)

1055

(1 RCT)d

⊕⊕⊕⊕
HIGH

Mefloquine plus cotrimoxazole results in an eight‐fold increase in vomiting

AEs: dizziness

75 per 1000

296 per 1000

(214 to 411)

RR 3.94

(2.85 to 5.46)

1055

(1 RCT)e

⊕⊕⊕⊕
HIGH

Mefloquine plus cotrimoxazole results in a four‐fold increase in dizziness

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
Abbreviations: CI: confidence interval; IRR: incidence rate ratio; RCT: randomized controlled trial; RR: risk ratio.

GRADE Working Group grades of evidence.
High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: The true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: The true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: The true effect is likely to be substantially different from the estimate of effect.

aAlthough one trial has serious risk of bias, the other is of high certainty and exclusion of the smaller trial has little effect on the estimate of effect.
bDowngraded by 1 for inconsistency: Trials showed substantial heterogeneity.
cDowngraded by 1 for imprecision: Confidence intervals range from considerable benefit to considerable harm.
dA second RCT, Denoeud‐Ndam 2014a BEN, reported 50 events in the mefloquine+cotrimoxazole group and 0 in the control group (cotrimoxazole), with RR 101 (95% CI 6.29 to 1621.68). This trial was open and participants knew to which group they were allocated. Meta‐analysis causes a paradoxically very wide CI. Because of this distortion, we have used the results from Gonzalez 2014b KEN MOZ TAN in the grade table.
eA second RCT, Denoeud‐Ndam 2014a BEN, reported 52 events in the mefloquine+cotrimoxazole group and 0 in the control group (cotrimoxazole), with RR 105 (95% CI 6.54 to 1685.03). This trial was open and participants knew to which group they were allocated. Meta‐analysis causes a paradoxically very wide CI with the lower 95% CI. Because of this distortion, we have used the results from Gonzalez 2014b KEN MOZ TAN in this ‘Summary of findings' table.

Figuras y tablas -
Summary of findings 2. Mefloquine plus cotrimoxazole compared with cotrimoxazole for preventing malaria in pregnant women
Comparison 1. Mefloquine versus sulfadoxine‐pyrimethamine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical malaria episodes during pregnancy Show forest plot

2

Rate Ratio (Fixed, 95% CI)

0.83 [0.65, 1.05]

2 Maternal peripheral parasitaemia at delivery Show forest plot

2

5455

Risk Ratio (M‐H, Fixed, 95% CI)

0.65 [0.48, 0.86]

3 Placental malaria Show forest plot

2

4668

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.58, 1.86]

4 Mean haemoglobin at delivery Show forest plot

2

5588

Mean Difference (IV, Fixed, 95% CI)

0.10 [0.01, 0.19]

5 Maternal anaemia at delivery Show forest plot

2

5469

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.76, 0.94]

6 Severe maternal anaemia at delivery Show forest plot

2

5469

Risk Ratio (M‐H, Random, 95% CI)

0.93 [0.58, 1.48]

7 Cord blood parasitaemia Show forest plot

2

5309

Risk Ratio (M‐H, Random, 95% CI)

0.44 [0.13, 1.46]

8 Cord blood anaemia Show forest plot

1

4006

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.87, 1.23]

9 Mean birth weight Show forest plot

2

5241

Mean Difference (IV, Fixed, 95% CI)

2.52 [‐25.66, 30.69]

10 Low birth weight Show forest plot

2

5641

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.78, 1.17]

11 Low birth weight by gravidity Show forest plot

2

5641

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.84, 1.13]

11.1 Primigravidae

2

1576

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.80, 1.30]

11.2 Multigravidae

2

4065

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.78, 1.14]

12 Prematurity Show forest plot

2

4640

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.76, 1.40]

13 Malaria in first year of life Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.97 [0.82, 1.15]

14 Hospital admissions in first year of life Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.93 [0.75, 1.17]

15 SAEs during pregnancy Show forest plot

1

4674

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.81, 1.20]

16 Stillbirths and abortions Show forest plot

2

6219

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.91, 1.58]

17 Congenital malformations Show forest plot

2

5931

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.51, 2.37]

18 Maternal mortality Show forest plot

2

6219

Risk Ratio (M‐H, Random, 95% CI)

2.41 [0.27, 21.23]

19 Neonatal mortality Show forest plot

2

6134

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.67, 1.43]

20 Infant mortality Show forest plot

1

Rate Ratio (Fixed, 95% CI)

1.00 [0.66, 1.52]

21 AEs: vomiting Show forest plot

2

6272

Risk Ratio (M‐H, Fixed, 95% CI)

4.76 [4.13, 5.49]

22 AEs: fatigue/weakness Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

4.62 [1.80, 11.85]

23 AEs: dizziness Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

4.21 [3.36, 5.27]

24 AEs: headache Show forest plot

2

6272

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.25, 1.94]

Figuras y tablas -
Comparison 1. Mefloquine versus sulfadoxine‐pyrimethamine
Comparison 2. Mefloquine plus cotrimoxazole versus cotrimoxazole

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical malaria episodes during pregnancy Show forest plot

1

Rate Ratio (Fixed, 95% CI)

0.76 [0.33, 1.76]

2 Maternal peripheral parasitaemia at delivery (PCR) Show forest plot

2

989

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.30, 0.93]

3 Placental malaria (blood smear) Show forest plot

2

1144

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.29, 0.89]

4 Placental malaria (PCR) Show forest plot

2

977

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.14, 0.57]

5 Mean haemoglobin at delivery Show forest plot

2

1167

Mean Difference (IV, Random, 95% CI)

0.07 [‐0.32, 0.46]

6 Maternal anaemia at delivery (< 9.5 g/dL) Show forest plot

2

1197

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.73, 1.20]

7 Maternal severe anaemia at delivery Show forest plot

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.41, 2.08]

8 Cord blood parasitaemia Show forest plot

2

1166

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.03, 3.13]

9 Mean birth weight Show forest plot

2

1220

Mean Difference (IV, Random, 95% CI)

‐25.75 [‐86.99, 35.49]

10 Low birth weight Show forest plot

2

1220

Risk Ratio (M‐H, Fixed, 95% CI)

1.20 [0.89, 1.60]

11 Prematurity Show forest plot

2

824

Risk Ratio (M‐H, Random, 95% CI)

1.07 [0.58, 1.96]

12 SAEs during pregnancy Show forest plot

2

1347

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.50, 0.95]

13 Spontaneous abortions and stillbirths Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.42, 2.98]

14 Congenital malformations Show forest plot

2

1312

Risk Ratio (M‐H, Fixed, 95% CI)

0.61 [0.22, 1.67]

15 Maternal mortality Show forest plot

2

1347

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.13, 2.01]

16 Neonatal mortality Show forest plot

2

1239

Risk Ratio (M‐H, Fixed, 95% CI)

1.32 [0.65, 2.69]

17 Mother‐to‐child transmission HIV Show forest plot

2

1019

Risk Ratio (M‐H, Fixed, 95% CI)

1.92 [1.13, 3.25]

18 AEs: vomiting Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

20.88 [1.40, 311.66]

19 AEs: fatigue/weakness Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

2.95 [0.26, 32.93]

20 AEs: dizziness Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

16.34 [0.39, 684.99]

21 AEs: headache Show forest plot

2

1347

Risk Ratio (M‐H, Random, 95% CI)

0.76 [0.28, 2.10]

Figuras y tablas -
Comparison 2. Mefloquine plus cotrimoxazole versus cotrimoxazole
Comparison 3. Mefloquine versus cotrimoxazole

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Maternal peripheral parasitaemia at delivery (PCR) Show forest plot

1

98

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.03, 1.72]

2 Placental malaria (PCR) Show forest plot

1

94

Risk Ratio (M‐H, Fixed, 95% CI)

0.73 [0.13, 4.15]

3 Placental malaria (blood smear) Show forest plot

1

108

Risk Ratio (M‐H, Fixed, 95% CI)

0.35 [0.01, 8.30]

4 Mean haemoglobin at delivery Show forest plot

1

100

Mean Difference (IV, Fixed, 95% CI)

‐0.10 [‐0.67, 0.47]

5 Maternal anaemia at delivery (< 9.5 g/dL) Show forest plot

1

100

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.26, 3.16]

6 Mean birth weight Show forest plot

1

120

Mean Difference (IV, Fixed, 95% CI)

‐102.0 [‐255.52, 51.52]

7 Low birth weight Show forest plot

1

120

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.56, 4.13]

8 Prematurity Show forest plot

1

125

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.33, 3.56]

9 SAEs during pregnancy Show forest plot

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.28, 4.07]

10 Stillbirths Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

4.30 [0.49, 37.49]

11 Spontaneous abortions Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

1.07 [0.07, 16.84]

12 Congenital malformations Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

1.07 [0.16, 7.41]

13 Maternal mortality Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

14 Neonatal mortality Show forest plot

1

129

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.07, 16.39]

15 Infant deaths after 7 days Show forest plot

1

129

Risk Ratio (M‐H, Fixed, 95% CI)

2.10 [0.19, 22.54]

16 AEs: vomiting Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

13.43 [3.31, 54.54]

17 AEs: fatigue/weakness Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

6.99 [1.64, 29.81]

18 AEs: dizziness Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

52.60 [3.26, 848.24]

19 AEs: headache Show forest plot

1

139

Risk Ratio (M‐H, Fixed, 95% CI)

0.21 [0.01, 4.39]

Figuras y tablas -
Comparison 3. Mefloquine versus cotrimoxazole
Comparison 4. Mefloquine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Maternal peripheral parasitaemia during pregnancy Show forest plot

1

339

Risk Ratio (M‐H, Fixed, 95% CI)

0.13 [0.05, 0.33]

2 Placental malaria Show forest plot

1

220

Risk Ratio (M‐H, Fixed, 95% CI)

0.14 [0.01, 2.68]

3 Mean birth weight Show forest plot

1

290

Mean Difference (IV, Fixed, 95% CI)

‐80.0 [‐184.65, 24.65]

4 Low birth weight Show forest plot

1

290

Risk Ratio (M‐H, Fixed, 95% CI)

1.39 [0.78, 2.48]

5 Prematurity Show forest plot

1

199

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.15, 1.53]

6 Stillbirths Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [0.86, 8.08]

7 Spontaneous abortions Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

0.48 [0.04, 5.22]

8 Congenital malformations Show forest plot

1

311

Risk Ratio (M‐H, Fixed, 95% CI)

3.82 [0.43, 33.83]

9 Maternal mortality Show forest plot

1

339

Risk Ratio (M‐H, Fixed, 95% CI)

2.95 [0.12, 71.85]

10 Infant mortality Show forest plot

1

288

Risk Ratio (M‐H, Fixed, 95% CI)

1.04 [0.63, 1.74]

Figuras y tablas -
Comparison 4. Mefloquine versus placebo