Scolaris Content Display Scolaris Content Display

Mefloquina para la prevención del paludismo durante el viaje a zonas endémicas

Contraer todo Desplegar todo

Referencias

References to studies included in this review

Albright 2002 {published data only}

Albright TA, Binns HJ, Katz BZ. Side effects of and compliance with malaria prophylaxis in children. Journal of Travel Medicine 2002;9(6):289‐92. CENTRAL

Andersson 2008 {published data only}

Andersson H, Askling HH, Falck B, Rombo L. Well‐tolerated chemoprophylaxis uniformly prevented Swedish soldiers from Plasmodium falciparum malaria in Liberia, 2004‐2006. Military Medicine 2008;173(12):1194‐8. CENTRAL

Arthur 1990 {published data only}

Arthur JD, Echeverria P, Shanks GD, Karwacki J, Bodhidatta L, Brown JE. A comparative study of gastrointestinal infections in United States soldiers receiving doxycycline or mefloquine for malaria prophylaxis. American Journal of Tropical Medicine and Hygiene 1990;43(6):608‐13. CENTRAL

Belderok 2013 {published data only}

Belderok SM, van den Hoek A, Roeffen W, Sauerwein R, Sonder GJ. Adherence to chemoprophylaxis and Plasmodium falciparum anti‐circumsporozoite seroconversion in a prospective cohort study of Dutch short‐term travelers. PLoS ONE 2013;8(2):e56863. CENTRAL

Boudreau 1991 {published data only}

Boudreau EF, Pang LW, Chaikummao S, Witayarut C. Comparison of mefloquine, chloroquine plus pyrimethamine‐sulfadoxine (Fansidar), and chloroquine as malarial prophylaxis in eastern Thailand. Southeast Asian Journal of Tropical Medicine and Public Health1991; Vol. 22, issue 2:183‐9. CENTRAL

Boudreau 1993 {published data only}

Boudreau E, Schuster B, Sanchez J, Novakowski W, Johnson R, Redmond D, et al. Tolerability of prophylactic Lariam regimens. Tropical Medicine and Parasitology 1993;44(3):257‐65. CENTRAL

Bunnag 1992 {published data only}

Bunnag D, Malikul S, Chittamas S, Chindanond D, Harinasuta T, Fernex M, et al. Fansimef for prophylaxis of malaria: a double‐blind randomized placebo controlled trial. Southeast Asian Journal of Tropical Medicine and Public Health1992; Vol. 23, issue 4:777‐82. CENTRAL

Corominas 1997 {published data only}

Corominas N, Gascon J, Mejias T, Caparros F, Quinto L, Codina C, et al. Adverse drug reactions associated to the antimalarial chemoprophylaxis. Medicina Clinica 1997;108(20):772‐5. CENTRAL

Cunningham 2014 {published and unpublished data}

Cunningham J, Horsley J, Patel D, Tunbridge A, Lalloo DG. Compliance with long‐term malaria prophylaxis in British expatriates. Travel Medicine and Infectious Disease 2014;12(4):341‐8. CENTRAL

Davis 1996 {published data only}

Davis TM, Dembo LG, Kaye‐Eddie SA, Hewitt BJ, Hislop RG, Batty KT. Neurological, cardiovascular and metabolic effects of mefloquine in healthy volunteers: a double‐blind, placebo‐controlled trial. British Journal of Clinical Pharmacology 1996;42(4):415‐21. CENTRAL

Eick‐Cost 2017 {published data only}

Eick‐Cost A, Hu Z, Rohrbeck P, Clark L. Neuropsychiatric outcomes after mefloquine exposure among U.S. military service members. American Journal of Tropical Medicine and Hygiene 2017;96(1):159‐66. CENTRAL

Goodyer 2011 {published data only}

Goodyer L, Rice L, Martin A. Choice of and adherence to prophylactic antimalarials. Journal of Travel Medicine 2011;18(4):245‐9. CENTRAL

Hale 2003 {published data only}

Hale BR, Owusu‐Agyei S, Fryauff DJ, Koram KA, Adjuik M, Oduro AR, et al. A randomized, double‐blind, placebo‐controlled, dose‐ranging trial of tafenoquine for weekly prophylaxis against Plasmodium falciparum. Clinical Infectious Diseases 2003;36(5):541‐9. CENTRAL

Hill 2000 {published data only}

Hill DR. Health problems in a large cohort of Americans traveling to developing countries. Journal of Travel Medicine 2000;7(5):259‐66. CENTRAL

Hoebe 1997 {published data only}

Hoebe C, de Munter J, Thijs C. Adverse effects and compliance with mefloquine or proguanil antimalarial chemoprophylaxis. European Journal of Clinical Pharmacology 1997;52(4):269‐75. CENTRAL

Jute 2007 {published data only}

Jute S, Toovey S. Knowledge, attitudes and practices of expatriates towards malaria chemoprophylaxis and personal protection measures on a mine in Mali. American Journal of Tropical Medicine and Hygiene 2007;5(1):40‐3. CENTRAL

Kato 2013 {published and unpublished data}

Kato T, Okuda J, Ide D, Amano K, Takei Y, Yamaguchi Y. Questionnaire‐based analysis of atovaquone‐proguanil compared with mefloquine in the chemoprophylaxis of malaria in non‐immune Japanese travelers. Journal of Infection and Chemotherapy 2013;19(1):20‐3. CENTRAL

Korhonen 2007 {published and unpublished data}

Korhonen C, Peterson K, Bruder C, Jung P. Self‐reported adverse events associated with antimalarial chemoprophylaxis in peace corps volunteers. American Journal of Preventine Medicine 2007;33(3):194‐9. CENTRAL

Kuhner 2005 {published data only}

Kuhner S, Drager‐Hoppe HS, Dreesman J. Malaria chemoprophylaxis ‐ A survey on drug‐related adverse events. Medizinische Welt 2005;56(1‐2):51‐5. CENTRAL

Landman 2015 {published data only}

Landman KZ, Tan KR, Arguin PM. Adherence to malaria prophylaxis among Peace Corps volunteers in the Africa region, 2013. Travel Medicine and Infectious Disease 2015;13(1):61‐8. CENTRAL
Landman KZ, Tan KR, Arguin PM. Knowledge, attitudes, and practices regarding antimalarial chemoprophylaxis in U.S. Peace Corps volunteers ‐ Africa, 2013. Morbidity and Mortality Weekly Report 2014;63(23):516‐7. CENTRAL

Laver 2001 {published data only}

Laver SM, Wetzels J, Behrens RH. Knowledge of malaria, risk perception, and compliance with prophylaxis and personal and environmental preventive measures in travelers exiting Zimbabwe from Harare and Victoria Falls International airport. Journal of Travel Medicine 2001;8(6):298‐303. CENTRAL

Laverone 2006 {published data only}

Laverone E, Boccalini S, Bechini A, Belli S, Santini MG, Baretti S, et al. Travelers' compliance to prophylactic measures and behavior during stay abroad: Results of a retrospective study of subjects returning to a travel medicine center in Italy. Journal of Travel Medicine 2006;13(6):338‐44. CENTRAL

Lobel 2001 {published data only}

Lobel HO, Baker MA, Gras FA, Stennies GM, Meerburg P, Hiemstra E, et al. Use of malaria prevention measures by North American and European travelers to East Africa. Journal of Travel Medicine 2001;8(4):167‐72. CENTRAL

Mavrogordato 2012 {published data only}

Mavrogordato A, Lever AM. A cluster of Plasmodium vivax malaria in an expedition group to Ethiopia: Prophylactic efficacy of atovaquone/proguanil on liver stages of P. vivax. Journal of Infection 2012;65(3):269‐74. CENTRAL

Meier 2004 {published data only}

Meier CR, Wilcock K, Jick SS. The risk of severe depression, psychosis or panic attacks with prophylactic antimalarials. Drug Safety 2004;27(3):203‐13. CENTRAL

Napoletano 2007 {published data only}

Napoletano G, Bissoli P, Bisoffi Z, Todescato A, Gottardello L, Costa S, et al. Malaria chemoprophylaxis ‐ Follow up of returned travellers of Veneto Region, Italy. Giornale Italiano di Medicina Tropicale 2007;12(1‐4):13‐9. CENTRAL

Nosten 1994 {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. CENTRAL

Ohrt 1997 {published data only}

Ohrt C, Richie TL, Widjaja H, Shanks GD, Fitriadi J, Fryauff DJ, et al. Mefloquine compared with doxycycline for the prophylaxis of malaria in Indonesian soldiers. A randomized, double‐blind, placebo‐controlled trial. Annals of Internal Medicine1997; Vol. 126, issue 12:963‐72. CENTRAL

Overbosch 2001 {published data only}

Overbosch D, Schilthuis H, Bienzle U, Behrens RH, Kain KC, Clarke PD, et al. Atovaquone‐proguanil versus mefloquine for malaria prophylaxis in nonimmune travelers: results from a randomized, double‐blind study. Clinical Infectious Diseases 2001;33(7):1015‐21. CENTRAL

Pearlman 1980 {published data only}

Pearlman EJ, Doberstyn EB, Sudsok S, Thiemanun W, Kennedy RS, Canfield CJ. Chemosuppressive field trials in Thailand. IV. The suppression of Plasmodium falciparum and Plasmodium vivax parasitemias by mefloquine (WR 142,490, A 4‐quinolinemethanol). Americal Journal of Tropical Medicine and Hygiene 1980;29(6):1131‐7. CENTRAL

Petersen 2000 {published data only}

Petersen E, Ronne T, Ronn A, Bygbjerg I, Larsen SO. Reported side effects to chloroquine, chloroquine plus proguanil, and mefloquine as chemoprophylaxis against malaria in Danish travelers. Journal of Travel Medicine 2000;7(2):79‐84. CENTRAL

Philips 1996 {published data only}

Phillips MA, Kass RB. User acceptability patterns for mefloquine and doxycycline malaria chemoprophylaxis. Journal of Travel Medicine 1996;3(1):40‐5. CENTRAL

Potasman 2002 {published data only}

Potasman I, Weller B. Does mefloquine prophylaxis affect electroencephalographic patterns?. American Journal of Medicine 2002;112(2):147‐9. CENTRAL

Rack 2005 {published data only}

Rack J, Wichmann O, Kamara B, Gunther M, Cramer J, Schonfeld C, et al. Risk and spectrum of diseases in travelers to popular tourist destinations. Journal of Travel Medicine 2005;12(5):248‐53. CENTRAL

Rieckmann 1993 {published data only}

Rieckmann KH, Yeo AE, Davis DR, Hutton DC, Wheatley PF, Simpson R. Recent military experience with malaria chemoprophylaxis. Medical Journal of Australia 1993;158(7):446‐9. CENTRAL

Rietz 2002 {published data only}

Rietz G, Petersson H, Odenholt I. Many travellers suffer of side‐effects of malaria prophylaxis. Lakartidningen 2002;99(26‐7):2939‐44. CENTRAL

Salako 1992 {published data only}

Salako LA, Adio RA, Walker O, Sowunmi A, Sturchler D, Mittelholzer ML, et al. Mefloquine‐sulphadoxine‐pyrimethamine (Fansimef, Roche) in the prophylaxis of Plasmodium falciparum malaria: A double‐blind, comparative, placebo‐controlled study. Annals of Tropical Medicine and Parasitology 1992;86(6):575‐81. CENTRAL

Santos 1993 {published data only}

Santos JB, Prata A, Wanssa E. Mefloquine chemoprophylaxis of malaria in the Brazilian Amazonia. Revista da Sociedade Brasileira de Medicina Tropical 1993;26(3):157‐62. CENTRAL

Saunders 2015 {published data only}

Saunders DL, Garges E, Manning JE, Bennett K, Schaffer S, Kosmowski AJ, et al. Safety, tolerability, and compliance with long‐term antimalarial chemoprophylaxis in American soldiers in Afghanistan. American Journal of Tropical Medicine and Hygiene 2015;93(3):584‐90. CENTRAL

Schlagenhauf 1997 {published data only}

Schlagenhauf P, Lobel H, Steffen R, Johnson R, Popp K, Tschopp A, et al. Tolerance of mefloquine by SwissAir trainee pilots. American Journal of Tropical Medicine and Hygiene 1997;56(2):235‐40. CENTRAL

Schlagenhauf 2003 {published data only}

Schlagenhauf P, Johnson R, Schwartz E, Nothdurft HD, Steffen R. Evaluation of mood profiles during malaria chemoprophylaxis: a randomized, double‐blind, four‐arm study. Journal of Travel Medicine 2009;16(1):42‐45. CENTRAL
Schlagenhauf P, Tschopp A, Johnson R, Nothdurft HD, Beck B, Schwartz E, et al. Tolerability of malaria chemoprophylaxis in non‐immune travellers to sub‐Saharan Africa: multicentre, randomised, double blind, four arm study. BMJ 2003;327(7423):1078. CENTRAL

Schneider 2013 {published data only}

Schneider C, Adamcova M, Jick SS, Schlagenhauf P, Miller MK, Rhein HG, et al. Antimalarial chemoprophylaxis and the risk of neuropsychiatric disorders. Travel Medicine and Infectious Disease 2013;11(2):71‐80. CENTRAL
Schneider C, Adamcova M, Jick SS, Schlagenhauf P, Miller MK, Rhein HG, et al. Use of anti‐malarial drugs and the risk of developing eye disorders. Travel Medicine and Infectious Disease 2014;12(1):40‐7. CENTRAL

Schwartz 1999 {published data only}

Schwartz E, Regev‐Yochay G. Primaquine as prophylaxis for malaria for nonimmune travelers: A comparison with mefloquine and doxycycline. Clinical Infectious Diseases 1999;29(6):1502‐6. CENTRAL

Shamiss 1996 {published data only}

Shamiss A, Atar E, Zohar L, Cain Y. Mefloquine versus doxycycline for malaria prophylaxis in intermittent exposure of Israeli Air Force aircrew in Rwanda. Aviation, Space, and Environmental Medicine 1996;67(9):872‐3. CENTRAL

Sharafeldin 2010 {published data only}

Sharafeldin E, Soonawala D, Vandenbroucke JP, Hack E, Visser LG. Health risks encountered by Dutch medical students during an elective in the tropics and the quality and comprehensiveness of pre‐and post‐travel care. BMC Medical Education 2010;10:89. CENTRAL

Sonmez 2005 {published data only}

Sonmez A, Harlak A, Kilic S, Polat Z, Hayat L, Keskin O, et al. The efficacy and tolerability of doxycycline and mefloquine in malaria prophylaxis of the ISAF troops in Afghanistan. Journal of Infectious Diseases 2005;51(3):253‐8. CENTRAL

Sossouhounto 1995 {published data only}

Sossouhounto RT, Soro BN, Coulibaly A, Mittelholzer ML, Stuerchler D, Haller L. Mefloquine in the prophylaxis of P. Falciparum malaria. Journal of Travel Medicine 1995;2(4):221‐4. CENTRAL

Steffen 1993 {published data only}

Handschin JC, Wall M, Steffen R, Sturchler D. Tolerability and effectiveness of malaria chemoprophylaxis with mefloquine or chloroquine with or without co‐medication. Journal of Travel Medicine 1997;4(3):121‐7. CENTRAL
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. CENTRAL
Steffen R, Heusser R, Machler R, Bruppacher R, Naef U, Chen D, et al. Malaria chemoprophylaxis among European tourists in tropical Africa: use, adverse reactions, and efficacy. Bulletin of the World Health Organization 1990;68(3):313‐22. CENTRAL

Steketee 1996 {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, 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

Stoney 2016 {published data only}

Stoney RJ, Chen LH, Jentes ES, Wilson ME, Han PV, Benoit CM, et al. Malaria prevention strategies: adherence among Boston area travelers visiting malaria‐endemic countries. American Journal of Tropical Medicine and Hygiene 2016;94(1):136‐42. CENTRAL

Tan 2017 {published data only}

Tan KR, Henderson S, Williamson J, Ferguson RW, Wilkinson TM, Jung P, et al. Long term health outcomes among returned Peace Corps volunteers after malaria prophylaxis, 1995‐2014. Travel Medicine and Infectious Disease 2017;17:50‐55. CENTRAL

Terrell 2015 {published data only}

Terrell AG, Forde ME, Firth R, Ross DA. Malaria chemoprophylaxis and self‐reported impact on ability to work: mefloquine versus doxycycline. Journal of Travel Medicine 2015;22(6):383‐8. CENTRAL

Tuck 2016 {published data only}

Tuck J, Williams J. Malaria protection in Sierra Leone during the Ebola outbreak 2014/15; The UK military experience with malaria chemoprophylaxis Sep 14‐Feb 15. Travel Medicine and Infectious Diseases 2016;14(5):471‐4. CENTRAL

van Riemsdijk 1997 {published data only}

van Riemsdijk MM, van der Klauw MM, van Heest JA, Reedeker FR, Ligthelm RJ, Herings RM, et al. Neuro‐psychiatric effects of antimalarials. European Journal of Clinical Pharmacology 1997;52(1):1‐6. CENTRAL

van Riemsdijk 2002 {published data only}

van Riemsdijk MM, Sturkenboom MC, Ditters JM, Ligthelm RJ, Overbosch D, Stricker BH. Atovaquone plus chloroguanide versus mefloquine for malaria prophylaxis: a focus on neuropsychiatric adverse events. Clinical Pharmacology and Therapeutics 2002;72(3):294‐301. CENTRAL

Vuurman 1996 {published data only}

Vuurman EF, Muntjewerff ND, Uiterwijk MM, Van Veggel LM, Crevoisier C, Haglund L, et al. Effects of mefloquine alone and with alcohol on psychomotor and driving performance. European Journal of Clinical Pharmacology 1996;50(6):475‐82. CENTRAL

Waner 1999 {published data only}

Waner S, Durrhiem D, Braack LE, Gammon S. Malaria protection measures used by in‐flight travelers to South African game parks. Journal of Travel Medicine 1999;6(4):254‐7. CENTRAL

Weiss 1995 {published data only}

Weiss WR, Oloo AJ, Johnson A, Koech D, Hoffman SL. Daily primaquine is effective for prophylaxis against falciparum malaria in Kenya: Comparison with mefloquine, doxycycline, and chloroquine plus proguanil. Journal of Infectious Disease 1995;171(6):1569‐75. CENTRAL

Wells 2006 {published data only}

Wells TS, Smith TC, Smith B, Wang LZ, Hansen CJ, Reed RJ, et al. Mefloquine use and hospitalizations among US service members, 2002‐2004. American Journal of Tropical Medicine and Hygiene 2006;74(5):744‐9. CENTRAL

References to studies excluded from this review

Abraham 1999 {published data only}

Abraham C, Clift S, Grabowski P. Cognitive predicators of adherence to malaria prophylaxis regimens on return from a malarious region: protective study. Social Science and Medicine 1999;48(11):1641‐54. CENTRAL

Adera 1995 {published data only}

Adera T, Wolfe MS, McGuire‐Rugh K, Calhoun N, Marum L. Risk factors for malaria among expatriates living in Kampala, Uganda: The need for adherence to chemoprophylactic regimens. American Journal of Tropical Medicine and Hygiene 1995;52(3):207‐12. CENTRAL

Adshead 2014 {published data only}

Adshead S. The adverse effects of mefloquine in deployed military personnel. Journal of the Royal Naval Medical Service 2014;100(3):232‐7. CENTRAL

Angelin 2014 {published data only}

Angelin M, Evengard B, Palmgren H. Travel health advice: Benefits, compliance, and outcome. Scandinavian Journal of Infectious Diseases 2014;46(6):447‐53. CENTRAL

Anonymous 1991 {published data only}

Anonymous. Mefloquine ‐ A new antimalarial. Drug and Therapeutics Bulletin 1991;29(13):51‐2. CENTRAL

Anonymous 1998 {published data only}

Anonymous. Medication‐associated depression. WHO Drug Information 1998;12(2):81. CENTRAL

Anonymous 1998a {published data only}

Anonymous. Mefloquine effectiveness impaired by high withdrawal rates. WHO Drug Information 1998;12(1):7‐8. CENTRAL

Anonymous 2005 {published data only}

Anonymous. Mefloquine: Revised patient information. WHO Drug Information 2005;19(2):119. CENTRAL

Anonymous 2009 {published data only}

Anonymous. Mefloquine: interstitial pneumonia: rare events. Prescrire International 2009;18(102):167. CENTRAL

Artaso 2004 {published data only}

Artaso Irigoyen B, Langarica Eseverri M, Campos Mangas MC. Mefloquine‐induced acute psychosis. Psiquiatria Biologica 2004;11(4):164‐6. CENTRAL

Arthur 1990a {published data only}

Arthur JD, Shanks GD, Echeverria P. Mefloquine prophylaxis. Lancet 1990;335(8695):972. CENTRAL

Banerjee 2001 {published data only}

Banerjee D, Stanley PJ. Malaria chemoprophylaxis in UK general practitioners traveling to South Asia. Journal of Travel Medicine 2001;8(4):173‐5. CENTRAL

Barbero Gonzalez 2003 {published data only}

Barbero Gonzalez A, Alvarez de Toledo Saavedra F, Esteban Fernandez J, Pastor‐Sanchez R, Gil de Miguel A, Rodriguez Barrios JM, et al. Management of vaccinations and prophylaxis of international travellers from community pharmacy (VINTAF Study). Atencion Primaria 2003;32(5):276‐81. CENTRAL

Barrett 1996 {published data only}

Barrett PJ, Emmins PD, Clarke PD, Bradley DJ. Comparison of adverse events associated with use of mefloquine and combination of chloroquine and proguanil as antimalarial prophylaxis: postal and telephone survey of travelers. British Medical Journal 1996;313(7056):525‐8. CENTRAL

Berger 1998 {published data only}

Berger A. Science commentary: protection against malaria. British Medical Journal 1998;317(7171):1508. CENTRAL

Berman 2004 {published data only}

Berman J. Toxicity of commonly‐used antimalarial drugs. Travel Medicine and Infectious Disease 2004;2(3‐4):171‐84. CENTRAL

Bernado 1994 {published data only}

Bernardo M, Parellada E. Mefloquine and severe psychiatric disorder. Medicina Clinica 1994;102(15):596. CENTRAL

Bijker 2014 {published data only}

Bijker EM, Schats R, Obiero JM, Behet MC, Gemert GJ, Vegte‐Bolmer M, et al. Sporozoite immunization of human volunteers under mefloquine prophylaxis is safe, immunogenic and protective: a double‐blind randomized controlled clinical trial. PLoS ONE 2014;9(11):e112910. CENTRAL

Bjorkman 1991 {published data only}

Bjorkman A, Steffen R, Armengaud M, Picot N, Piccoli S. Malaria chemoprophylaxis with mefloquine. Lancet 1991;337(8755):1479‐80. CENTRAL

Black 2007 {published data only}

Black J. Lariam and Halfan. Journal of the Royal Society of Medicine 2007;100(8):355‐6. CENTRAL

Blanke 2003 {published data only}

Blanke CH. Increased malaria‐morbidity of long‐term travellers due to inappropriate chemoprophylaxis recommendations. Tropical Doctor 2003;33(2):117‐9. CENTRAL

Botella de Maglia 1999 {published data only}

Botella de Maglia J, Espacio Casanovas A. Prevention of malaria. Revista Clínica Española 1999;199(8):549‐50. CENTRAL

Bourgeade 1990 {published data only}

Bourgeade A, Tonin V, Keudjian F, Levy PY, Faugere B. Accidental mefloquine poisoning. Presse Medicale 1990;19(41):1903. CENTRAL

Brenier‐Pinchart 2000 {published data only}

Brenier‐Pinchart MP, Brion JP, Issartel B, Barro C, Pinel C, Ambroise‐Thomas P. Glucose‐6‐phosphate dehydrogenase deficiency and hemoglobinuric biliary fever after taking mefloquine. Presse Medicale 2000;29(3):142. CENTRAL

Brisson 2012 {published data only}

Brisson M, Brisson P. Compliance with antimalaria chemoprophylaxis in a combat zone. American Journal of Tropical Medicine and Hygiene 2012;86(4):587‐90. CENTRAL

Bruguera 2007 {published data only}

Bruguera M, Herrera S. Acute hepatitis associated with mefloquine therapy. Gastroenterologie Hepatologie 2007;30(2):102‐3. CENTRAL

Burke 1993 {published data only}

Burke BM. Mefloquine. Lancet 1993;341(8860):1605‐6. CENTRAL

Caillon 1992 {published data only}

Caillon E, Schmitt L, Moron P. Acute depressive symptoms after mefloquine treatment. American Jounal of Psychiatry 1992;149(5):712. CENTRAL

Carme 1997 {published data only}

Carme B, Peguet C, Nevez G. Compliance with and tolerance of mefloquine and chloroquine + proguanil malaria chemoprophylaxis in French short‐term travellers to sub‐Saharan Africa. Tropical Medicine and International Health 1997;2(10):953‐6. CENTRAL
Carme B, Peguet C, Nevez G. Malaria chemoprophylaxis: tolerance and compliance with mefloquine and proguanil/chloroquine combination in French tourists. Bulletin de la Societe de Pathologie Exotique 1997;90(4):273‐6. CENTRAL

Castot 1988 {published data only}

Castot A, Garnier R. The secondary effects of mefloquine. Concours Medical 1988;110(43):4003. CENTRAL

Cave 2003 {published data only}

Cave W, Pandey P, Osrin D, Shlim DR. Chemoprophylaxis use and the risk of malaria in travelers to Nepal. Journal of Travel Medicine 2003;10(2):100‐5. CENTRAL

Charles 2007 {published data only}

Charles BG, Blomgren A, Nasveld PE, Kitchener SJ, Jensen A, Gregory RM, et al. Population pharmacokinetics of mefloquine in military personnel for prophylaxis against malaria infection during field deployment. European Journal of Clinical Pharmacology 2007;63(3):271‐8. CENTRAL

Chin 2016 {published data only}

Chin BS, Kim JY, Gianella S, Lee M. Travel pattern and prescription analysis at a single travel clinic specialized for yellow fever vaccination in South Korea. Infection and Chemotherapy 2016;48(1):20‐30. CENTRAL

Clifford 2009 {published data only}

Clifford D, Brew B, Cinque P, Gorelik L, Bennett D, Panzara MA, et al. Design of a clinical trial of mefloquine in patients with progressive multifocal leukoencephalopathy. 25th Congress of the European Committee for Treatment and Research in Multiple Sclerosis; 2009 Sep 9‐12; Dusseldorf Germany. Dusseldorf, 2009:S87. CENTRAL

Clift 1996 {published data only}

Clift S, Grabowski P. Malaria prophylaxis and the media. Lancet 1996;348(9023):344. CENTRAL

Clyde 1976 {published data only}

Clyde DF, McCarthy VC, Miller RM, Hornick RB. Suppressive activity of mefloquine in sporozoite‐induced human malaria. Antimicrobial Agents and Chemotherapy 1976;9(3):384‐6. CENTRAL

Cobelens 1997 {published data only}

Cobelens FG, van Thiel PP. There is no evidence of more symptoms with mefloquine than with other drugs in malaria prophylaxis. Nederlands Tijdschrift voor Geneeskunde 1997;141(16):794‐5; author reply 796. CENTRAL

Cohen 1997 {published data only}

Cohen J. Mefloquine prophylaxis ‐ revisited. Australian Family Physician 1997;26(8):909. CENTRAL

Conget 1993 {published data only}

Conget JJ, Navarro M, Navarro P, Corachan M. Prophylaxis with mefloquine and changes in thyroidal hormones. Medicina Clinica 1993;100(13):516. CENTRAL

Conrad 1997 {published data only}

Conrad KA, Kiser WR. Response to doxycycline vs. mefloquine. Military Medicine 1997;162(6):iii. CENTRAL

Corbett 1996 {published data only}

Corbett EL, Doherty JF, Behrens RH. Adverse events associated with mefloquine. Study in returned travelers confirms authors' findings. British Medical Journal 1996;313(7071):1552. CENTRAL

Coulaud 1986 {published data only}

Coulaud JP. Chemoprophylaxis of malaria. Medecine et Maladies Infectieuses 1986;16(12):746. CENTRAL

Croft 1996 {published data only}

Croft AM, World MJ. Neuropsychiatric reactions with mefloquine chemoprophylaxis. Lancet 1996;347(8997):326. CENTRAL

Croft 1997 {published data only}

Croft AM, Clayton TC, World MJ. Side effects of mefloquine prophylaxis for malaria: an independent randomized controlled trial. Transactions of the Royal Society of Tropical Medicine and Hygiene 1997;91(2):199‐203. CENTRAL

Del Cacho 2001 {published data only}

Del Cacho Ma E, Martinez M, Tuset M, Biarnes C, Mejias T, Gascon J, et al. Advice program for travelers about antimalarial prohilaxis. Atencion Farmaceutica 2001;3(3):171‐6. CENTRAL

Dia 2010 {published data only}

Dia A, Gautret P, Adheossi E, Bienaime A, Gaillard C, Simon F, et al. Illness in French travelers to Senegal: Prospective cohort follow‐up and sentinel surveillance data. Journal of Travel Medicine 2010;17(5):296‐302. CENTRAL

Durrheim 1999 {published data only}

Durrheim DN, Gammon S, Waner S, Braack LE. Antimalarial prophylaxis‐‐use and adverse events in visitors to the Kruger National Park. Suid‐Afrikaanse Tydskrif vir Geneeskunde 1999;89(2):170‐5. CENTRAL

Eamsila 1993 {published data only}

Eamsila C, Singharaj P, Yooyen P, Chatnugrob P, Nopavong Na Ayuthya A, Webster HK, et al. Prevention of Plasmodium falciparum malaria by Fansimef and Lariam in the northeastern part of Thailand. Southeast Asian Journal of Tropical Medicine and Public Health 1993;24(4):672‐6. CENTRAL

El Jaoudi 2010 {published data only}

El Jaoudi R, Benziane H, Khabbal Y, Elomri N, Lamsaouri J, Cherrah Y. Long‐term malaria prophylaxis with mefloquine: a study of adverse drug reactions. Therapie 2010;65(5):439‐45. CENTRAL

Fernando 2016 {published data only}

Fernando SD, Dharmawardana P, Semege S, Epasinghe G, Senanayake N, Rodrigo C, et al. The risk of imported malaria in security forces personnel returning from overseas missions in the context of prevention of re‐introduction of malaria to Sri Lanka. Malaria Journal 2016;15(1):144. CENTRAL

Fujii 2007 {published data only}

Fujii T, Kaku K, Jelinek T, Kimura M. Malaria and mefloquine prophylaxis use among Japan ground self‐defense force personnel deployed in East Timor. Journal of Travel Medicine 2007;14(4):226‐32. CENTRAL

Hamer 2008 {published data only}

Hamer DH, Ruffing R, Callahan MV, Lyons SH, Abdullah AS. Knowledge and use of measures to reduce health risks by corporate expatriate employees in western Ghana. Journal of Travel Medicine 2008;15(4):237‐42. CENTRAL

Hellgren 1990 {published data only}

Hellgren U, Angel VH, Bergqvist Y, Arvidsson A, Forero‐Gomez J, Rombo L. Plasma concentrations of sulfadoxine‐pyrimethamine and of mefloquine during regular long term malaria prophylaxis. Transactions of the Royal Society of Tropical Medicine and Hygiene 1990;84(1):46‐9. CENTRAL

Hopperus 1996 {published data only}

Hopperus Buma AP, Thiel PP, Lobel HO, Ohrt C, Ameijden EJ, Veltink RL, et al. Long‐term malaria chemoprophylaxis with mefloquine in Dutch marines in Cambodia. Journal of Infectious Diseases 1996;173(6):1506‐9. CENTRAL

Jaspers 1996 {published data only}

Jaspers CA, Hopperus Buma AP, van Thiel PP, van Hulst RA, Kager PA. Tolerance of mefloquine chemoprophylaxis in Dutch military personnel. American Journal of Tropical Medicine and Hygiene 1996;55(2):230‐4. CENTRAL

Jensen 1998 {published data only}

Jensen JJ. Mefloquine: neuropsychiatric adverse effects are often severe and persistent long after withdrawal of the drug. Ugeskrift for Laeger 1998;160(16):2413. CENTRAL

Karbwang 1991 {published data only}

Karbwang J, Molunto P, Bangchang KN, Banmairuroi V, Bunnag D, Harinasuta T. Pharmacokinetics of prophylactic mefloquine. Southeast Asian Journal of Tropical Medicine and Public Health 1991;22(4):519‐22. CENTRAL

Karbwang 1991a {published data only}

Karbwang J, Bangchang KN, Supapojana A, Bunnag D, Harinasuta T. Pharmacokinetics of prophylactic mefloquine in Thai healthy volunteers. Southeast Asian Journal of Tropical Medicine and Public Health 1991;22(1):68‐71. CENTRAL

Khaliq 2001 {published data only}

Khaliq Y, Gallicano K, Carignan CT, Cooper C, McCarthy A. Pharmacokinetic interaction between mefloquine and ritonavir in healthy volunteers. British Journal of Clinical Pharmacology 2001;51(6):591‐600. CENTRAL

Kimura 2006 {published data only}

Kimura M, Kawakami K, Hashimoto M, Hamada M. Malaria prevention and stand‐by emergency treatment among Japanese travelers. Travel Medicine and Infectious Disease 2006;4(2):81‐5. CENTRAL

Kitchener 2003 {published data only}

Kitchener S, Nasveld P, Russell B, Elmes N. An outbreak of malaria in a forward battalion on active service in East Timor. Military Medicine 2003;168(6):457‐9. CENTRAL

Kitchener 2005 {published data only}

Kitchener SJ, Nasveld PE, Gregory RM, Edstein MD. Mefloquine and doxycycline malaria prophylaxis in Australian soldiers in East Timor. Medical Journal of Australia 2005;182(4):168‐71. CENTRAL

Kok 1997 {published data only}

Kok PW, Puls FT, Zonderland HD. Not more symptoms with mefloquine use than with other drugs in malaria prophylaxis. Nederlands Tijdschrift Voor Geneeskunde 1997;141(18):898. CENTRAL

Kollaritsch 2000 {published data only}

Kollaritsch H, Karbwang J, Wiedermann G, Mikolasek A, Na‐Bangchang K, Wernsdorfer WH. Mefloquine concentration profiles during prophylactic dose regimens. Wiener Klinische Wochenschrift 2000;112(10):441‐7. CENTRAL

Kozarsky 1993 {published data only}

Kozarsky P, Eaton M. Use of mefloquine for malarial chemoprophylaxis in its first year of availability in the United States. Clinical Infectious Diseases 1993;16(1):185‐6. CENTRAL

Landry 2006 {published data only}

Landry P, Iorillo D, Darioli R, Burnier M, Genton B. Do travelers really take their mefloquine malaria chemoprophylaxis? Estimation of adherence by an electronic pillbox. Journal of Travel Medicine 2006;13(1):8‐14. CENTRAL

Lapierre 1983 {published data only}

Lapierre J, Devant J, Coquelin B, Faurant C, Galal AA. Results of an experiment on chemoprophylaxis of malaria using mefloquine in Cambodia (Cambodia‐Thai border region). Bulletin de la Societe de Pathologie Exotique et de ses Filiales 1983;76(4):357‐63. CENTRAL

Lim 2005 {published data only}

Lim DS. Dermatology in the military: An East Timor study. International Journal of Dermatology 2005;44(4):304‐11. CENTRAL

Lobel 1993 {published data only}

Lobel HO, Bernard KW, Williams SL, Hightower AW, Patchen LC, Campbell C. Effectiveness and tolerance of long‐term malaria prophylaxis with mefloquine. Need for a better dosing regimen. JAMA 1991;265(3):361‐4. CENTRAL
Lobel HO, Miani M, Eng T, Bernard KW, Hightower AW, Campbell CC. Long‐term malaria prophylaxis with weekly mefloquine. Lancet 1993;341(8849):848‐51. CENTRAL

Looareesuwan 1987 {published data only}

Looareesuwan S, White NJ, Warrell DA, Forgo I, Schwartz DE, et al. Studies of mefloquine bioavailability and kinetics using a stable isotope technique: comparison of Thai patients with falciparum malaria and healthy Caucasian volunteers. British Journal of Clinical Pharmacology 1987;24(1):37‐42. CENTRAL

MacArthur 2002 {published data only}

MacArthur JR, Parise ME, Steketee RW. Relationships between mefloquine blood levels, gender, and adverse reactions. American Journal of Tropical Medicine and Hygiene 2002;66(5):445; author reply 446‐7. CENTRAL

Malvy 2006 {published data only}

Malvy D, Pistone T, Rezvani A, Lancon F, Vatan R, Receveur MC, et al. Risk of malaria among French adult travellers. Travel Medicine and Infectious Disease 2006;4(5):259‐69. CENTRAL

Marcy 1996 {published data only}

Marcy SM, Wilson ME. Malaria prophylaxis in young children and pregnant women. Pediatric Infectious Disease Journal 1996;15(1):101‐2. CENTRAL

Massey 2007 {published data only}

Massey P, Durrheim DN, Speare R. Inadequate chemoprophylaxis and the risk of malaria. Australian Family Physician 2007;36(12):1058‐60. CENTRAL

Matsumura 2005 {published data only}

Matsumura T, Fujii T, Miura T, Koibuchi T, Endo T, Nakamura H, et al. Questionnaire‐based analysis of mefloquine chemoprophylaxis for malaria in a Japanese population. Journal of Infection and Chemotherapy 2005;11(4):196‐8. CENTRAL

Meszaros 1996 {published data only}

Meszaros K. Acute psychosis caused by mefloquine prophylaxis?. Canadian Journal of Psychiatry 1996;41(3):196. CENTRAL

Michel 2007 {published data only}

Michel R, Ollivier L, Meynard JB, Guette C, Migliani R, Boutin JP. Outbreak of malaria among policemen in French Guiana. Military Medicine 2007;172(9):977‐81. CENTRAL

Mimica 1983 {published data only}

Mimica I, Fry W, Eckert G, Schwartz DE. Multiple‐dose kinetic study of mefloquine in healthy male volunteers. Chemotherapy 1983;29(3):184‐7. CENTRAL

Mizuno 2006 {published data only}

Mizuno Y, Kudo K, Kano S. Chemoprophylaxis according to the guidelines on malaria prevention for Japanese overseas travellers. Southeast Asian Journal of Tropical Medicine and Public Health 2006;37(Suppl 3):11‐4. CENTRAL

Mizuno 2010 {published data only}

Mizuno Y, Kudo K, Kano S. Mefloquine chemoprophylaxis against malaria in Japanese travelers: Results of a study on adverse effects. Tropical Medicine and Health 2010;38(3):103‐6. CENTRAL

Moon 2011 {published data only}

Moon J, Deye G, Miller L, Fracisco S, Miller RS, Tosh D, et al. Malaria infection in individuals taking mefloquine does not induce antibody response to MSP142. 58th Annual Meeting of the American Society of Tropical Medicine and Hygiene; 2009 November 18‐22; Washington, USA. Washington, USA: American Journal of Tropical Medicine and Hygiene, 2009; Vol. 81:156‐7. CENTRAL
Moon JE, Deye GA, Miller L, Fracisco S, Miller RS, Tosh D, et al. Plasmodium falciparum infection during suppressive prophylaxis with mefloquine does not induce an antibody response to merozoite surface protein‐1(42). American Journal of Tropical Medicine and Hygiene 2011;84(5):825‐29. CENTRAL

Morales de Naime 1989 {published data only}

Morales de Naime L, Kosidub H, Martínez Iturriza L. Prophylactic malaria with pyrimethamine, sulfadoxine and mefloquine [Paludismo tratamiento profiláctico con pirimetamina, sulfadoxina y mefloquina]. Medicina Interna 1989;5(3‐4):125‐37. CENTRAL

Munawar 2012 {published data only}

Munawar CM, Khan TP, Hyder MF. Effect of mefloquine on eye. Pakistan Journal of Medical and Health Sciences 2012;6(2):340‐2. CENTRAL

Mølle 2000 {published data only}

Mølle I, Christensen KL, Hansen PS, Dragsted UB, Aarup M, Mads Buhl R. Use of medical chemoprophylaxis and antimosquito precautions in Danish malaria patients and their traveling companions. Journal of Travel Medicine 2000;7(5):253‐8. CENTRAL

Namikawa 2008 {published data only}

Namikawa K, Kikuchi H, Kato S, Takizawa Y, Konta A, Iida T, et al. Knowledge, attitudes, and practices of Japanese travelers towards malaria prevention during overseas travel. Travel Medicine and Infectious Disease 2008;6(3):137‐41. CENTRAL

Nasveld 2010 {published data only}

Dow GS, McCarthy WF, Reid M, Smith B, Tang D, Shanks GD. A retrospective analysis of the protective efficacy of tafenoquine and mefloquine as prophylactic anti‐malarials in non‐immune individuals during deployment to a malaria‐endemic area. Malaria Journal 2014;13(1):49. CENTRAL
Nasveld PE, Edstein MD, Reid M, Brennan L, Harris IE, Kitchener SJ, et al. Randomized, double‐blind study of the safety, tolerability, and efficacy of tafenoquine versus mefloquine for malaria prophylaxis in nonimmune subjects. Antimicrobial Agents and Chemotherapy 2010;54(2):792‐8. CENTRAL

Nevin 2010 {published data only}

Nevin RL. Mefloquine prescriptions in the presence of contraindications: Prevalence among US military personnel deployed to Afghanistan, 2007. Pharmacoepidemiology and Drug Safety 2010;19(2):206‐10. CENTRAL

Nevin 2012 {published data only}

Nevin RL. Biased measurement of neuropsychiatric adverse effects of pediatric mefloquine treatment. Pediatric Infectious Diseases Journal 2012;31(1):102; author reply 102‐3. CENTRAL

Nosten 1990 {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

Nosten 1999 {published data only}

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. CENTRAL

Nwokolo 2001 {published data only}

Nwokolo C, Wambebe C, Akinyanju O, Raji AA, Audu BS, Emodi IJ, et al. Mefloquine versus proguanil in short‐term malaria chemoprophylaxis in sickle cell anaemia. Clinical Drug Investigation 2001;21(8):537‐44. CENTRAL

Olanrewaju 2000 {published data only}

Olanrewaju I W, Lin L. Mefloquine chemoprophylaxis in Chinese railway workers on contract in Nigeria. Journal of Travel Medicine 2000;7(3):116‐9. CENTRAL

Ollivier 2004 {published data only}

Ollivier L, Tifratene K, Josse R, Keundjian A, Boutin JP. The relationship between body weight and tolerance to mefloquine prophylaxis in non‐immune adults: results of a questionnaire‐based study. Annals of Tropical Medicine and Parasitology 2004;98(6):639‐41. CENTRAL

Peetermans 2001 {published data only}

Peetermans WE, Van Wijngaerden E. Implementation of pretravel advice: Good for malaria, bad for diarrhoea. Acta Clinica Belgica 2001;56(5):284‐8. CENTRAL

Peragallo 1999 {published data only}

Peragallo MS, Sabatinelli G, Sarnicola G. Compliance and tolerability of mefloquine and chloroquine plus proguanil for long‐term malaria chemoprophylaxis in groups at particular risk (the military). Transactions of the Royal Society of Tropical Medicine and Hygiene 1999;93(1):73‐7. CENTRAL

Peragallo 2002 {published data only}

Peragallo MS, Croft AM, Kitchener SJ. Malaria during a multinational military deployment: the comparative experience of the Italian, British and Australian Armed Forces in East Timor. Transactions of the Royal Society of Tropical Medicine and Hygiene 2002;96(5):481‐2. CENTRAL

Peragallo 2014 {published data only}

Peragallo MS, Sarnicola G, Boccolini D, Romi R, Mammana G. Risk assessment and prevention of malaria among Italian troops in Afghanistan, 2002 to 2011. Journal of Travel Medicine 2014;21(1):24‐32. CENTRAL

Philips 1994 {published data only}

Phillips M. Antimalarial mefloquine. Medical Journal of Australia 1994;161(3):227‐8. CENTRAL

Phillips 1996 {published data only}

Phillips M. Adverse events associated with mefloquine. Women may be more susceptible to adverse events. British Medical Journal 1996;313(7071):1552‐3. 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 Travel Medicine 1998;5(3):121‐6. CENTRAL

Pistone 2007 {published data only}

Pistone T, Guibert P, Gay F, Malvy D, Ezzedine K, Receveur MC, et al. Malaria risk perception, knowledge and prophylaxis practices among travellers of African ethnicity living in Paris and visiting their country of origin in sub‐Saharan Africa. Transactions of the Royal Society of Tropical Medicine and Hygiene 2007;101(10):990‐5. CENTRAL

Port 2011 {published data only}

Port A, Cottrell G, Dechavanne C, Briand V, Bouraima A, Guerra J, et al. Prevention of malaria during pregnancy: assessing the effect of the distribution of IPTp through the national policy in Benin. American Journal of Tropical Medicine and Hygiene 2011;84(2):270‐5. CENTRAL

Potasman 2000 {published data only}

Potasman I, Beny A, Seligmann H. Neuropsychiatric problems in 2,500 long‐term young travelers to the tropics. Journal of Travel Medicine 2000;7(1):5‐9. CENTRAL

Quinn 2016 {published data only}

Quinn JC. Better approach needed to detect and treat military personnel with adverse effects from mefloquine. BMJ 2016;352:i838. CENTRAL

Reisinger 1989 {published data only}

Reisinger EC, Horstmann RD, Dietrich M. Tolerance of mefloquine alone and in combination with sulfadoxine‐pyrimethamine in the prophylaxis of malaria. Transactions of the Royal Society of Tropical Medicine and Hygiene 1989;83(4):474‐7. CENTRAL

Rieckmann 1974 {published data only}

Rieckmann KH, Trenholme GM, Williams RL, Carson PE, Frischer H, Desjardins RE. Prophylactic activity of mefloquine hydrochloride (WR 142490) in drug‐resistant malaria. Bulletin of the World Health Organization 1974;51(4):375‐7. CENTRAL

Rieke 1993 {published data only}

Rieke B, Fleischer K. Permanent health impairment after staying in malaria areas. Versicherungsmedizin 1993;45(6):197‐202. CENTRAL

Ries 1993 {published data only}

Ries S. Cerebral spasm during malaria prophylaxis with mefloquine. Deutsche Medizinische Wochenschrift 1993;118(51‐2):1911‐2. CENTRAL

Ringqvist 2015 {published data only}

Ringqvist A, Bech P, Glenthøj B, Petersen E. Acute and long‐term psychiatric side effects of mefloquine: a follow‐up on Danish adverse event reports. Travel Medicine and Infectious Disease 2015;13(1):80‐8. CENTRAL

Rombo 1993 {published data only}

Rombo L, Angel VH, Friman G, Hellgren U, Mittelholzer ML, Sturchler D. Comparative tolerability and kinetics during long‐term intake of Lariam and Fansidar for malaria prophylaxis in nonimmune volunteers. Tropical Medicine and Parasitology 1993;44(3):254‐6. CENTRAL

Rønn 1998 {published data only}

Rønn AM, Rønne‐Rasmussen J, Gøtzsche PC, Bygbjerg IC. Neuropsychiatric manifestations after mefloquine therapy for Plasmodium falciparum malaria: comparing a retrospective and a prospective study. Tropical Medicine and International Health 1998;3(2):83‐8. CENTRAL

Sallent 1997 {published data only}

Sallent LV. Anti‐malarial prophylaxis: The role of the general practitioner. Atencion Primaria 1997;20(10):558‐62. CENTRAL

Schlagenhauf 1996 {published data only}

Schlagenhauf P, Steffen R, Lobel H, Johnson R, Letz R, Tschopp A, et al. Mefloquine tolerability during chemoprophylaxis: focus on adverse event assessments, stereochemistry and compliance. Tropical Medicine and International Health 1996;1(4):485‐94. CENTRAL

Scott 1993 {published data only}

Scott R. Malaria chemoprophylaxis. South African Medical Journal 1993;83(11):861. CENTRAL

Smail 1991 {published data only}

Smail A, Ducroix JP, Cohen G, Baillet J. Current malaria prophylactic therapy for travellers servicemen. La Semaine des Hopitaux de Paris 1991;67(40‐41):1824‐7. 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 Disease 1997;176(3):831‐3. CENTRAL

Suriyamongkol 1991 {published data only}

Suriyamongkol V, Timsaad S, Shanks GD. Mefloquine chemoprophylaxis of soldiers on the Thai‐Cambodian border. Southeast Asian Journal of Tropical Medicine and Public Health 1991;22(4):515‐8. CENTRAL

Tansley 2010 {published data only}

Tansley R, Lotharius J, Priestley A, Bull F, Duparc S, Mohrle J. A randomized, double‐blind, placebo‐controlled study to investigate the safety, tolerability, and pharmacokinetics of single enantiomer (+)‐mefloquine compared with racemic mefloquine in healthy persons. American Journal of Tropical Medicine and Hygiene 2010;83(6):1195‐201. CENTRAL

ter Kuile 1993 {published data only}

ter Kuile FO, Nosten F, Luxemburger C, White NJ. Mefloquine prophylaxis. Lancet 1993;342(8870):551. CENTRAL

Todd 1997 {published data only}

Todd GD, Hopperus Buma AP, Green MD, Jaspers CA, Lobel HO. Comparison of whole blood and serum levels of mefloquine and its carboxylic acid metabolite. American Journal of Tropical Medicine and Hygiene 1997;57(4):399‐402. CENTRAL

Turner 2014 {published data only}

Turner C, Sabin C, Chiodini P, Bhagani S, Johnson M, Zuckerman J. Cross‐sectional study investigating the prescription, adherence and tolerability of malaria prophylaxis in HIV‐positive travellers. 3rd Joint Conference of the British HIV Association, BHIVA with the British Association for Sexual Health and HIV, BASHH Liverpool United Kingdom; 2014 April 1‐4; Liverpool, UK. Liverpool, UK: HIV Medicine, 2014; Vol. 15:127. CENTRAL

Valerio 2005 {published data only}

Valerio L, Martinez O, Sabria M, Esteve M, Urbiztondo L, Roca C. High‐risk travel abroad overtook low‐risk travel from 1999 to 2004: Characterization and trends in 2,622 Spanish travelers. Journal of Travel Medicine 2005;12(6):327‐31. CENTRAL

Van Genderen 2007 {published data only}

Van Genderen PJ, Koene HR, Spong K, Overbosch D. Atovaquone‐proguanil versus mefloquine for malaria prophylaxis in nonimmune travelers: Results from a randomized, double‐blind study. Journal of Travel Medicine 2007;14(2):92‐5. CENTRAL

Van Grootheest 1999 {published data only}

Van Grootheest AC, Van Puijenbroek EP, Heeringa M. Adverse effects of mefloquine: Agitation undermines objective assessment. Pharmaceutisch Weekblad 1999;134(4):114‐6. CENTRAL

van Riemsdijk 2004 {published data only}

Aarnoudse AL, van Schaik RH, Dieleman J, Molokhia M, van Riemsdijk MM, Ligthelm RJ, et al. MDR1 gene polymorphisms are associated with neuropsychiatric adverse effects of mefloquine. Clinical Pharmacology and Therapeutics 2006;80(4):367‐74. CENTRAL
van Riemsdijk MM, Ditters JM, Sturkenboom MC, Tulen JH, Ligthelm RJ, Overbosch D, et al. Neuropsychiatric events during prophylactic use of mefloquine before travelling. British Journal of Clinical Pharmacology 2002;58(6):441‐5. CENTRAL
van Riemsdijk MM, Sturkenboom MC, Ditters JM, Tulen JH, Ligthelm RJ, Overbosch D, et al. Low body mass index is associated with an increased risk of neuropsychiatric adverse events and concentration impairment in women on mefloquine. British Journal of Clinical Pharmacology 2004;57(4):506‐12. CENTRAL

Venturini 2011 {published data only}

Venturini E, Chiappini E, Mannelli F, Bonsignori F, Galli L, De Martino M. Malaria prophylaxis in African and Asiatic children traveling to their parents' home country: A Florentine study. Journal of Travel Medicine 2011;18(3):161‐4. CENTRAL

Wagner 1986 {published data only}

Wagner WH. Fight against malaria ‐ chemotherapy and immunoprophylaxis. Part 1. Chemotherapy. Arzneimittel Forschung 1986;36(1):1‐9. CENTRAL

Wallace 1996 {published data only}

Sánchez JL, DeFraites RF, Sharp TW, Hanson RK. Mefloquine or doxycycline prophylaxis in US troops in Somalia. Lancet 1993;341(8851):1021‐2. CENTRAL
Wallace MR. Malaria among United States troops in Somalia. American Journal of Medicine 1996;100(1):49‐55. CENTRAL

Weinke 1991 {published data only}

Weinke T, Trautmann M, Held T, Weber G, Eichenlaub D, Fleischer K, et al. Neuropsychiatric side effects after the use of mefloquine. American Journal of Tropical Medicine and Hygiene 1991;45(1):86‐91. CENTRAL

White 2016 {published data only}

White C. UK troops to continue to receive mefloquine despite concern over adverse events. BMJ 2016;354:i5030. CENTRAL

Win 1985 {published data only}

Win K, Thwe Y, Lwin TT, Win K. Combination of mefloquine with sulfadoxine‐pyrimethamine compared with two sulfadoxine‐pyrimethamine combinations in malaria chemoprophylaxis. Lancet 1985;2(8457):694‐5. CENTRAL

Winstanley 1999 {published data only}

Winstanley P, Behrens R. Malaria prophylaxis with mefloquine: Neurological and psychiatric adverse drug reactions. Prescribers' Journal 1999;39(3):161‐5. CENTRAL

Wolters 1997 {published data only}

Wolters BA, Bosje T, Luinstra‐Passchier MJ. Not more problems with mefloquine compared to other antimalarial prophylactics. Nederlands Tijdschrift voor Geneeskunde 1997;141(7):331‐4. CENTRAL

ACROBAT‐NSRI tool

Sterne JAC, Higgins JPT, Reeves BC on behalf of the development group for ACROBAT‐NRSI. A Cochrane Risk Of Bias Assessment Tool: for Non‐Randomized Studies of Interventions (ACROBAT‐NRSI). Version 1.0.0 [September 2014]. Available from: http://www.riskofbias.info[Accessed September 2017].

Behrens 2015

Behrens R, Neave P, Jones P. Imported malaria among people who travel to visit friends and relatives: is current UK policy effective or does it need a strategic change?. Malaria Journal 2015;14:149.

Briand 2009

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.

Bukiwra 2014

Bukirwa H, Unnikrishnan B, Kramer CV, Sinclair D, Nair S, Tharyan P. Artesunate plus pyronaridine for treating uncomplicated Plasmodium falciparum malaria. Cochrane Database of Systematic Reviews 2014, Issue 3. [DOI: 10.1002/14651858.CD006404.pub2]

CDC 2015a

Centers for Disease Control and Prevention. Malaria biology. 2015. www.cdc.gov/malaria/about/biology(accessed September 2017).

CDC 2015b

Centers for Disease Control and Prevention. Drugs for malaria prevention table. Travelers' health. 2015. Yellow book. wwwnc.cdc.gov/travel/diseases/malaria(accessed September 2017).

Covidence 2017 [Computer program]

Veritas Health Innovation. Covidence. Version accessed prior to 4 October 2017. Melbourne, Australia: Veritas Health Innovation, 2017.

Croft 2005

Croft AM. Malaria: prevention in travellers. BMJ Clinical Evidence 2010;2010:0903.

EMA 2014

European Medicines Agency. PRAC recommendations on signals adopted at the PRAC meeting of 3‐6 March 2014. EMA/PRAC/65788/2014. ema.europa.eu/docs/en_GB/document_library/PRAC_recommendation_on_signal/2014/03/WC500163626.pdf (accessed prior to 19 September 2017).

eMC 2015a

electronic Medicines Compendium. Summary of product characteristics: Larium. medicines.org.uk/emc/medicine/1701#UNDESIRABLE_EFFECTS 2015 (accessed September 2017).

eMC 2015b

electronic Medicines Compendium. Glossary. medicines.org.uk/emc/glossary/#SPC 2015 (accessed September 2017).

FDA 2013

USA Food, Drug Administration. FDA drug safety communication: mefloquine. fda.gov/drugs/drugsafety/ucm362227.htm 2013 (accessed September 2017).

González 2014

González R, Hellgren U, Greenwood B, Menéndez C. Mefloquine safety and tolerability in pregnancy: a systematic literature review. Malaria Journal 2014;13:75.

González 2014a

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):e1001733.

Handschin 1997

Handschin JC, Wall M, Steffen R, Stürchler D. Tolerability and effectiveness of malaria chemoprophylaxis with mefloquine or chloroquine with or without co‐medication. Journal of Travel Medicine 1997;4(3):121‐7.

Higgins 2011

Higgins JPT, Altman DG, Sterne JAC (editors). Chapter 8: Assessing risk of bias in included studies. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0 [updated March 2011]. http://training.cochrane.org/handbook. The Cochrane Collaboration, 2011. Available at www.cochrane‐handbook.org, (accessed prior to 19 September 2017).

Høgh 2000

Høgh B, Clarke PD, Camus D, et al. Atovaquone‐proguanil versus chloroquine‐proguanil for malaria prophylaxis in non‐immune travellers: a randomised, double‐blind study. Lancet 2000;356:1888‐94.

ICH 1994

International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH). Clinical safety data management: definitions and standards for expedited reporting E2A. 1994. ICH harmonised tripartite guideline. https://www.imim.cat/media/upload/arxius/MEDIA436.pdf (accessed prior to 19 September 2017).

Lefebvre 2011

Lefebvre C, Manheimer E, Glanville J. Chapter 6: Searching for studies. Cochrane Handbook for Systematic Reviews of Interventions, Version 5.1.0 [updated March 2011]. training.cochrane.org/handbook (accessed prior to 19 September 2017).

Loke 2007

Loke YK, Price D, Herxheimer A. Systematic reviews of adverse effects: framework for a structured approach. BMC Medical Research Methodology 2007;7:32.

Loke 2011

Loke YK, Price D, Herxheimer A. Chapter 14: Adverse effects. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0 [updated March 2011]. training.cochrane.org/handbook. The Cochrane Collaboration, 2011. Available at www.cochrane‐handbook.org, (accessed prior to 19 September 2017).

McCarthy 2015

McCarthy S. Malaria prevention, mefloquine neurotoxicity, neuropsychiatric illness, and risk‐benefit analysis in the Australian Defence Force. Journal of Parasitology Research 2015;2015:287651. [DOI: 10.1155/2015/287651]

MedDRA 2016

Medical Dictionary for Regulatory Activities. Introductory guide MedDRA version 19.0. 2016. International Federation of Pharmaceutical Manufacturers and Associations. meddra.org/sites/default/files/guidance/file/intguide_19_0_english.pdf(accessed September 2017).

Meremikwu 2008

Meremikwu MM, Donegan S, Esu E. Chemoprophylaxis and intermittent treatment for preventing malaria in children. Cochrane Database of Systematic Reviews 2008, Issue 2. [DOI: 10.1002/14651858.CD003756.pub3]

Muanda 2015

Muanda FT, Chaabane S, Boukhris T, Santos F, Sheehy O, Perreault S, et al. Antimalarial drugs for preventing malaria during pregnancy and the risk of low birthweight: a systematic review and meta‐analysis of randomized and quasi‐randomized trials. BMC Medicine 2015;13:193. [DOI: 10.1186/s12916‐015‐0429‐x]

Murray 2014

Murray CJ, Ortblad KF, Guinovart C, Lim SS, Wolock TM, Roberts DA, et al. Global, regional, and national incidence and mortality for HIV, tuberculosis, and malaria during 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014;384(9947):1005‐70.

Nevin 2015

Nevin R. Rational risk‐benefit decision‐making in the setting of military mefloquine policy. Journal of Parasitology Research 2015;2015:260106. [DOI: 10.1155/2015/260106]

Oniyangi 2006

Oniyangi O, Omari AA. Malaria chemoprophylaxis in sickle cell disease. Cochrane Database of Systematic Reviews 2006, Issue 4. [DOI: 10.1002/14651858.CD003489.pub2]

PHAC 2014

Public Health Agency of Canada. Canadian recommendations for the prevention and treatment of malaria. Canada Communicable Disease Report. 2014. publications.gc.ca/collections/collection_2014/aspc‐phac/HP40‐102‐2014‐eng.pdf(accessed September 2017).

PHE 2015

Public Health England Advisory Committee for Malaria Prevention for UK Travellers. Guidelines for malaria prevention in travellers from the UK. 2015. gov.uk/government/publications/malaria‐prevention‐guidelines‐for‐travellers‐from‐the‐uk(accessed September 2017).

Radeva‐Petrova 2014

Radeva‐Petrova D, Kayentao K, ter Kuile F, 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]

Reeves 2011

Reeves BC, Deeks JJ, Higgins JPT, Wells GA. Chapter 13: Including non‐randomized studies. Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0 [updated March 2011]. training.cochrane.org/handbook (accessed prior to 19 September 2017).

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.

Rodrigo 2016

Rodrigo C, Rajapakse S, Fernando S. Primaquine or tafenoquine for preventing malaria in people travelling to or living in endemic areas. Cochrane Database of Systematic Reviews 2016, Issue 6. [DOI: 10.1002/14651858.CD012242]

Schlagenhauf 2003

Schlagenhauf P, Tschopp A, Johnson R, Nothdurft HD, Beck B, Schwartz E, et al. Tolerability of malaria chemoprophylaxis in non‐immune travellers to sub‐Saharan Africa: multicentre, randomised, double blind, four arm study. BMJ 2003;327(7423):1078.

Schlagenhauf 1999

Schlagenhauf P. Mefloquine for malaria chemoprophylaxis 1992‐1998: a review. Journal of Travel Medicine 1999;6(2):122‐33.

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.

Shanks 2005

Shanks GD, Edstein MD. Modern malaria chemoprophylaxis. Drugs 2005;65(12):2091‐110.

Tickell‐Painter 2017

Tickell‐Painter M, Saunders R, Mayaan N, Lutje V, Mateo‐Urdiales A, Garner P. Deaths and parasuicides associated with mefloquine chemoprophylaxis: a systematic review. Travel Medicine and Infectious Disease (in press).

Treiber 2010

Treiber M, Wernsdorfer G, Wiedermann U, Congpuong K, Wernsdorfer WH. Sensitivity to artemisinin, mefloquine and quinine of Plasmodium falciparum in northwestern Thailand. Wiener Klinische Wochenschrift 2010;122(Suppl 3):52‐6.

Treiber 2011

Treiber M, Wernsdorfer G, Wiedermann U, Congpuong K, Sirichaisinthop J, Wernsdorfer WH. Sensitivity of Plasmodium vivax to chloroquine, mefloquine, artemisinin and atovaquone in north‐western Thailand. Wiener Klinische Wochenschrift 2011;123(Suppl 1):20‐5.

UK Parliament 2016

UK Parliament 2016. Commons select committee, UK Parliament. 'Larium should be 'drug of last resort' for troops'. parliament.uk/business/committees/committees‐a‐z/commons‐select/defence‐committee/news‐parliament‐2015/lariam‐report‐published‐16‐17/(accessed September 2017).

Warrell 2002

Warrell D, Gilles H. Essential Malariology. 4th Edition. New York: CRC Press, 2002.

WHO 2015

World Health Organization. Guidelines for the treatment of malaria. 2015. who.int/malaria/publications/atoz/9789241549127/en/(accessed September 2017).

WHO 2016

World Health Organization. World malaria report 2016. http://www.who.int/malaria/publications/world‐malaria‐report‐2016/en/(accessed September 2017).

WHO 2017

World Health Organization. Malaria, International travel and health. Chapter 7. who.int/ith/2017‐ith‐chapter7.pdf?ua=1 2017 (accessed 19 September 2017).

WHO‐ART 2008

Uppsala Monitoring Centre. WHO adverse reaction terminology (WHO‐ART). https://www.who‐umc.org/vigibase/services/learn‐more‐about‐who‐art/ (accessed prior to 19 September 2017).

References to other published versions of this review

Jacquerioz 2007

Jacquerioz FA, Croft AM. Drugs for preventing malaria in travellers. Cochrane Database of Systematic Reviews 2007, Issue 2. [DOI: 10.1002/14651858.CD006491]

Jacquerioz 2009

Jacquerioz FA, Croft AM. Drugs for preventing malaria in travellers. Cochrane Database of Systematic Reviews 2009, Issue 10. [DOI: 10.1002/14651858.CD006491.pub2]

Jacquerioz 2015

Jacquerioz FA, Croft AM. Drugs for preventing malaria in travellers. Cochrane Database of Systematic Reviews 2015, Issue 10. [DOI: 10.1002/14651858.CD006491.pub3]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Albright 2002

Methods

Design: retrospective cohort study.

Study dates: November 1997 to January 2000

Malaria transmission pattern and local antimalarial drug resistance: various destinations, not specified.

Adverse event monitoring: one off telephone interview with parents whose children had previously been prescribed antimalarial prophylaxis.

Participants

Number enrolled: 177 fit inclusion criteria and interviewed, 190 contacted

Inclusion criteria: children aged ≤ 13 years who visited the travel clinic at the Children’s Memorial Hospital in Chicago within the study dates. Subjects who were not on other medications.

Exclusion criteria: "...data were only included if the child was living with the interviewed parent while taking the antimalarial". "Unwillingness to participate in the study and language barriers".

Factors influencing drug allocation: "children... instructed to take mefloquine or chloroquine for malaria prophylaxis".

Country of recruitment: USA.

Country of malaria exposure: various; Africa 58%, Central or South America 21%, India 12% or Eastern Asia 9%.

Duration of exposure to malaria: various, not specified.

Type of participants: travellers

Interventions

1. Mefloquine*

2. Chloroquine*

*dosing regimen not specified

Outcomes

1. Adverse effects; any, nausea, vomiting, diarrhoea, headache, insomnia, abnormal dreams

2. Serious adverse effects

3. Discontinuations of study drug due to adverse effects

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex and destination of travel were recorded, but were not reported across prophylactic regimens

2. Selection of participants into the study: low

Non‐response rate 1.6%

3. Measurement of interventions: moderate

The prescription was provided by a travel clinic, but participants were asked to recall if they discontinued their medication 2.8 to 28 months after visiting

4. Departures from intended interventions: serious

Information was collected up to 2 years after taking the drug. No information was captured on switches.

5. Missing data: low

All information was collected at one time point, there were no losses to follow‐up.

6. Measurement of outcomes: serious

The outcome measure was subjective, participants and personnel were not blinded.

7. Selection of the reported results: low

All outcomes included in the introduction were reported in the results

8. Other: low

"The authors had no financial or other conflicts of interest to disclose"

Andersson 2008

Methods

Design: prospective cohort study

Study dates: March 2004 to November 2006

Malaria transmission pattern and local antimalarial drug resistance: malaria attack rate of 44% with P falciparum in another similar study at the time

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 690 soldiers sent questionnaire, 609 respondents

Inclusion criteria: all Swedish soldiers deployed to Liberia within the study dates

Exclusion criteria: none stated.

Factors influencing drug allocation: "...mefloquine was prescribed to almost all soldiers in the first two contingents and to about two‐thirds in the last three contingents. The remaining soldiers were recommended atovaquone/ proguanil. The latter group consisted mainly of those with body weight < 70 kg and those who had already experienced adverse events with mefloquine. No other drug regimes were used".

Country of recruitment: Sweden

Country of malaria exposure: Liberia

Duration of exposure to malaria: 6 months

Type of participants: military

Interventions

1. Mefloquine*

2. Atovaquone‐proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; any, nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams nightmares, insomnia sleep disturbance, depression

2. Serious adverse events; serious

3. Adverse events; other (concentration difficulties, mouth ulcers, fever, muscle pain)

4. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

5. Clinical cases of malaria

6. Overall satisfaction with the drug

7. Whether they would take the drug again

8. Measures of adherence to the drug regimen (data provided on aggregate)

Notes

Funding sources: Not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Information on potential confounders is not provided across prophylactic groups

2. Selection of participants into the study: moderate

609/690 (88%) response rate

3. Measurement of interventions: low

All participants were issued with the study drug.

4. Departures from intended interventions: low

Switches were recorded and reported

5. Missing data: serious

Outcomes were reported from 3 of 5 cohorts. No information was provided for 2 remaining cohorts.

6. Measurement of outcomes: serious

The outcome measure was subjective, participants and personnel were not blinded.

7. Selection of the reported results: low

All outcomes prespecified in the introduction were reported.

8. Other: moderate

Study sponsor not mentioned, but 2 study authors worked for GlaxoSmithKline

Arthur 1990

Methods

Design: RCT

Study dates: June to August 1988

Malaria transmission pattern and local drug resistance: local chloroquine resistance

Adverse event monitoring: blood taken at induction and at days 57 and 70 of treatment. Interviews regarding side effects when sera taken. Stool sample at induction, at end of exercise and at any time participants sought medical care.

Participants

Number enrolled: 270

Inclusion criteria: soldiers (aged 18 to 40 years), awaiting deployment to Thailand

Exclusion criteria: previous history of gastrointestinal illness

Country of recruitment: USA

Country of malaria exposure: Thailand

Duration of exposure to malaria: 5 weeks

Type of participants: soldiers, non‐immune

Interventions

1. Mefloquine (1 x 250 mg tablet) once weekly, starting 1 week before travel and continuing throughout the period of deployment.*

2. Doxycycline (1 capsule containing doxycycline hyclate 100 mg) once daily, starting 1 week before travel and continuing throughout the period of deployment*

Co‐interventions: Both groups given doxycycline 100mg daily for suppression of P falciparum and primaquine 45 mg weekly for elimination of liver hypnozoites for 6 weeks on return to the USA.

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Serious adverse event

3. Adverse events; diarrhoea

4. Discontinuation of study drug due to adverse effects

5. Measures of adherence to the drug regimen

Outcomes assessed not included in the review:

6. Laboratory tests; enteric pathogens

7. Adverse events; nausea, vomiting, headache, dizziness (data provided on aggregate)

Notes

Funding sources: Pfizer Inc supplied active and placebo doxycycline; Hoffman‐La Roche Inc supplied active and placebo mefloquine

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Volunteers were assigned from a computer generated random number list to receive daily doxycycline or weekly mefloquine"

Allocation concealment (selection bias)

Unclear risk

Comment: Unclear how the tablets were labelled and whether allocation concealment occurred

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"Soldiers receiving mefloquine also received identical appearing doxycycline placebo capsules daily, and those receiving daily doxycycline received weekly mefloquine placebo tablets"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no explanation of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

High risk

"Of the 270 volunteers who were deployed, 253 were correctly taking the assigned study malaria prophylaxis on arrival in Korat"

Comment: Reasons for not taking medication were not reported. Method of detection for malaria, frequency and duration of follow‐up were not reported

Incomplete outcome data (attrition bias); safety

Low risk

Comment: 17 participants (6%) were not "correctly taking the prophylaxis on arrival to Korat" and were excluded from the analysis. Data were not stratified by time point

Selective reporting (reporting bias); efficacy

Low risk

"None of the soldiers developed malaria"

Selective reporting (reporting bias); safety

Unclear risk

Comment: data for general side effects (e.g. headaches) were presented for the study population but not for each group

Other bias

Unclear risk

Comment: study sponsor not mentioned

Belderok 2013

Methods

Design: prospective cohort study

Study dates: October 2006 to October 2007

Malaria transmission pattern and local antimalarial drug resistance: various destinations, not specified

Adverse event monitoring: not performed

Participants

Number enrolled: 945

Inclusion criteria: People aged ≥ 18 years were eligible if they were planning to travel for 1 to 13 weeks to one or more malaria‐endemic countries.

Exclusion criteria: None stated

Factors influencing drug allocation: "Dutch national guidelines for travelers’ health advice"

Country of recruitment: Netherlands

Regions of malaria exposure: various; Asia 48%, Africa 30% and Latin America 22%

Duration of exposure to malaria: various; 49% ≤ 13 days, 35% 14 to 28 days and 9% ≥ 29 days

Type of participants: travellers

Interventions

1. Mefloquine: taken 3 weeks prior to arrival, during trip and for 4 weeks after return, dose and frequency of dose not specified

2. Atovaquone‐proguanil: 1 day prior to arrival, during trip and for 7 days after return, dose and frequency of dose not specified

3. Proguanil: On day of arrival, during trip and for 4 weeks after return, dose and frequency of dose not specified

Outcomes

Included in the review:

1. Measures of adherence to the drug regime

Outcomes assessed not included in the review:

2. Clinical cases of malaria

3. Predictors of adherence to malaria prophylaxis

4. Use of antimosquito preventive measures

Notes

Funding sources: The Amsterdam Academic Collaborative Center on Public Health is financially supported by the Netherlands Organization for Health Research and Development (ZonMw; grant number 7115 0001, http://www.zonmw.nl/nl/)

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Length of stay, travel destination, age and sex were not reported across groups

2. Selection of participants into the study: moderate

Non‐response rates were not reported

3. Measurement of interventions: low

Participants made daily diary entries during travel

4. Departures from intended interventions: low

Participants made daily diary entries during travel

5. Missing data: low

Information was collected at one time point

6. Measurement of outcomes: moderate

Outcome assessors were not blinded, methods were comparable across groups

7. Selection of the reported results: low

Outcomes were reported for 610/620 participants

8. Other: low

Government funding

Boudreau 1991

Methods

Design: RCT

Study dates: July 1983 to March 1984

Malaria transmission pattern and local antimalarial drug resistance: "in this area we believe the efficacy of chloroquine prophylaxis at the time of the study was negligible"

Adverse event monitoring: "at each 2 week visit… history of symptoms over the previous fortnight was obtained. Patients were asked about fever, chills, headache, nausea, vomiting, diarrhoea, anorexia, rash, myalgia and dysuria or abnormally coloured urine". Laboratory studies were performed at baseline and at 6 weeks in participants who had not developed malaria

Participants

Number enrolled: 501

Inclusion criteria: "Only males 21 years of age or over were accepted"

Exclusion criteria: "All participants were required to have a negative malaria smear (after examination of 200 fields on thick smear) on entry into the study". "...the use of other antimalarials or antibiotics"

Country of recruitment: Cambodia

Country of malaria exposure: Cambodia

Duration of exposure to malaria: ongoing in semi immune population, 14 week study period

Type of participants: Thai gem miners with a degree of immunity

Interventions

Included in review comparisons:

1. Mefloquine (2 x 250 mg tablet) fortnightly for 14 weeks*

2. Chloroquine (1 x 300 mg tablet) weekly*

Not included in review comparisons:

3. Fansidar (2 x 500 mg sufadoxine and 25 mg pyrimethamine) fortnightly and chloroquine (1 x 300 mg tablet) weekly*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Adverse events; other (myalgias, rash)

Outcomes assessed not included in the review:

3. Laboratory tests; haematocrit, complete blood count, transaminase levels, total and direct bilirubin, alkaline phosphatase, blood urea nitrogen

4. Adverse events; headache, anorexia, fever, chills, nausea, diarrhoea or vomiting (data provided on aggregate)

Notes

Funding sources: Support for this study was from the USA Army Medical Research and Development Command

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Assignment… is a 4:3:2 ratio"

Comment: method of sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

Comment: no details of allocation concealment were reported

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

"Every two weeks in a double blind fashion one of the investigators administered five tablets to each subject"

Comment: not mentioned whether placebo tablets had an identical appearance

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no mention of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Unclear risk

"Only 194 patients completed the study until positivity or end of the 14 weeks observation period". "Therefore of the original 501 enrollees, 63 were discarded due to positivity at week 0 and 104 were discarded since they never returned beyond week 0".

Comment: Losses to follow‐up during the study was not reported across groups

Incomplete outcome data (attrition bias); safety

Unclear risk

"Only 194 patients completed the study until positivity or end of the 14 weeks observation period...Any subject missing one appointment was excluded from the study though each subject's records up to the time of exclusion were entered into the survival analysis...After 3 weeks post treatment and a negative malaria smear some patients wishing to continue were reentered under a new study number and were assigned a double blind randomized treatment"

Selective reporting (reporting bias); efficacy

Unclear risk

Comment: number of people contracting malaria in each group and person‐weeks in the study were reported

Selective reporting (reporting bias); safety

Unclear risk

"There were no significant differences in frequency of complaints among the study groups for headache, anorexia, fever, chills, nausea, diarrhoea, or vomiting".

Comment: Data for specific adverse events not reported. Methods section states participants were asked about dysuria and abnormally coloured urine, but this was not reported in the results

Other bias

Low risk

Support for this study was from the USA Army Medical Research and Development Command

Boudreau 1993

Methods

Design: RCT

Study dates: not mentioned

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: "At each visit, the subject answered two computerised questionnaires (the Environmental Symptoms Questionnaire and the Profile of Mood States) [and] a physician interview was performed"

Participants

Number enrolled: 359

Inclusion criteria: "males at least 18 years old, met military weight standards, were available for weekly administration of medications and monitoring during the 13 week study period, and were willing to give informed consent"

Exclusion criteria: "treatment with beta‐blocking agents or other cardiotropic drugs, underlying chronic disease, history of cardiac arrhythmia, medical history of psychiatric or neurological problems within the last 5 years, anaemia or impaired hepatic or renal function. Women were excluded from participation in the study due to the risk of teratogenicity involved when the drug is used in early pregnancy"

Country of recruitment: USA

Country of malaria exposure: not applicable

Duration of exposure to malaria: not applicable

Type of participants: military, non‐travellers

Interventions

1. Mefloquine (1 x 250 mg tablet), larium 228 mg base (F Hoffman La Roche) weekly for 11 weeks

2. Mefloquine (1 x 250 mg tablet), larium 228 mg base (F Hoffman La Roche) weekly for 11 weeks, with loading dose of 1 x 250 mg tablet daily for 3 days during the first week

3. Chloroquine (1 x 300 mg tablet), 300 mg base (F Hoffman La Roche) weekly for 11 weeks

Outcomes

Included in the review:

1. Adverse events; nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia

2. Adverse events; other (irritability, poor concentration, anger, moodiness, abdominal distension, anorexia, environmental symptoms questionnaire (ESQ), sleep assessment, Profile of Mood States questionnaire)

Outcomes assessed not included in the review:

3. Laboratory tests: haemoglobin, haematocrit, platelets, white blood cell count, alanine aminotransferase, blood urea nitrogen and creatinine

4. Analysis of the dizziness index on the ESQ

5. Spontaneous comments on the ESQ (data provided on aggregate)

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"...military personnel were assigned to drug groups in a ratio of approximately 3:3:1…stratification was performed by major subordinate command so that equal proportions of each study group would be represented in each MSC"

Comment: not mentioned how the randomisation code was generated

Allocation concealment (selection bias)

Unclear risk

Comment: method allocation concealment not mentioned

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"...the ‘double dummy’ method of blinding was employed with either chloroquine or mefloquine placebos administered with active drug… In addition, during the first week of the study, on days two and three, a single mefloquine tablet or placebo was administered. Both drugs and placebos had an extremely bitter taste... identical placebo tablets"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no description provided of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Unclear risk

Comment: 15 medical withdrawals are reported within the study. It is unclear whether these are the only losses to follow up which occurred, or whether they occurred in the mefloquine loading dose group or weekly administration group.

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

High risk

‘table 5 outlines the percent of the group with symptoms only when significance was demonstrated’ ‘selected haematology and biochemistry tests were performed… no significant differences were noted among the three drugs when comparing the mean values’

Comment: data is not fully reported for ‘other symptoms’; only significant results are reported for the ESQ, and data for spontaneous comments on the ESQ are not reported; data is not fully reported for the POMS.

Other bias

Unclear risk

Comment: study sponsor not mentioned, but the lead author is attributed to ‘Pharmaceutical Systems Incorporated’

Bunnag 1992

Methods

Design: RCT

Study dates: July 1987 to January 1988

Malaria transmission pattern and local antimalarial drug resistance: "a malaria endemic area". Reports chloroquine, sulfadoxine‐pyrimethamine and quinine resistance within Thailand at the time of the study.

Adverse event monitoring: "volunteers asked about adverse events at each visit (weeks 4, 9, 14, 19, 24, 28)...starting week 14, volunteers reporting adverse events were interviewed by members of the hospital team; most of them were also seen by principal investigators"

Participants

Number enrolled: 605 randomized, 3 excluded because of baseline parasitaemia

Inclusion criteria: "...healthy male volunteers, aged between 16 and 60, living in this area, were recruited"

Exclusion criteria: "persons with a known history of allergy against sulphonamides, with an evidence illness of fever, or which a positive blood film (with or without symptomatic malaria) were excluded"

Country of recruitment: Thailand

Country of malaria exposure: Thailand

Duration of exposure to malaria: trial duration 24 weeks

Type of participants: Thai residents in a malaria‐endemic area (presumed semi‐immune)

Interventions

Included in the review:

1. Mefloquine (1 tablet containing 125 mg mefloquine) once weekly, double dose during first 4 weeks*

2. Chloroquine (1 tablet containing 300 mg chloroquine) once weekly*

3. Placebo

Not included in the review:

4. Fansifem (1 tablet containing 125 mg mefloquine, 250 mg sulfadoxine, 12.5 mg pyrimethamine) once weekly, double dose during first 2 weeks*

5. Fansidar (1 tablet containing 500 mg sulfadoxine, 25 mg pyrimethamine) once weekly*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Adverse events; any

3. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

4. Laboratory tests; haematocrit, white blood cell count and neutrophil count

Notes

Funding sources: "The project was jointly organized and conducted by the Malaria Division, Department of Communicable Disease, Ministry of Public Health; the Hoffman‐La Roche company, Basel, Switzerland; and The Faculty of Tropical Medicines, Mahidol University, Bangkok"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Eligible volunteers were randomly assigned to treatment groups"

Comment: method of random sequence generation not reported

Allocation concealment (selection bias)

Unclear risk

"The tablets were identical in appearance; they were packed in numbered blister packs and were in addition labelled weeks 1‐24... the coded test drugs for weeks 1‐4 were given to every subject"

Comment: no mention of concealed opaque envelopes or central allocation

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"A randomised double blind trial…the tablets were identical in appearance; they were packed in numbered blister packs"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no explanation provided of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Unclear risk

"Of the 605 subjects originally randomised, 3 were excluded because of baseline parasitaemia... Although some of the volunteers left the study for personal reasons (moving away from the area)"

Comment: numbers lost to follow up have not been reported

Incomplete outcome data (attrition bias); safety

Unclear risk

"94% (116/123) in the mefloquine group and 98% (119/121) in the placebo group were included for adverse event reporting"

"Although some of the volunteers left the study for personal reasons (moving away from the area)"

Comment: numbers lost to follow‐up were not reported

Selective reporting (reporting bias); efficacy

Low risk

Comment: Malaria cases were fully reported

Selective reporting (reporting bias); safety

High risk

Comment: Data were collected but not reported for adherence to drug regimen. Data were provided on aggregate across all time points. The number of adverse events were reported but not types or severity

Other bias

High risk

"The project was jointly organized and conducted by the Malaria Division, Department of Communicable Disease, Ministry of Public Health; the Hoffman‐La Roche company, Basel, Switzerland; and The Faculty of Tropical Medicines, Mahidol University, Bangkok"

Corominas 1997

Methods

Design: retrospective cohort study

Study dates: June 1992 to July 1994

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 1511 questionnaires distributed, 1054 respondents

Inclusion criteria: travellers who visited areas with a risk of malaria infection who were travelling on short trips < 6 weeks duration

Exclusion criteria: none mentioned

Factors influencing drug allocation: The fact of participating in this study did not change at all the typical prophylaxis when performing, which followed the usual criteria (Google Translate = "El hecho de participar en este estudio no cambio en absoluto el tipico de profilaxis al realizar, que siguio los criterios habituales"

Country of recruitment: Spain

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine (1 x 250 mg tablet) weekly, starting 1 week prior to travel, during the trip and 4 weeks following return from the malaria‐endemic area

2. Chloroquine (5 mg/kg) weekly, starting 1 week prior to travel, during the trip and 4 weeks following return from the malaria‐endemic area

Outcomes assessed not included in the review:

3. Chloroquine and proguanil (chloroquine base 5 mg/kg, once weekly plus proguanil 100 mg daily, if weight < 55 kg and 200 mg daily if weight > 55 kg) starting 1 week prior to travel, during the trip and 4 weeks following return from the malaria‐endemic area

Outcomes

Included in the review:

1. Adverse effects; any, vertigo, visual impairment, nausea, vomiting, abdominal pain, diarrhoea, insomnia, anxiety, depression, pruritis

2. Adverse effects; other (irritability)

3. Discontinuations of study drugs due to adverse effects

Outcomes assessed not included in the review:

4. Mean number of symptoms reported per traveller

5. Adverse effects; other, incidence < 1% (amnesia, tremor, paraesthesia, seizures, hyper‐reflexia, drowsiness, asthenia, nervousness, difficulty concentrating, mouth ulcers, acne, cardiac rhythm disturbance)

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex was reported across groups. No other confounders were reported

2. Selection of participants into the study: serious

1054/1511 (70%) response rate

3. Measurement of interventions: low

The antimalarial prescription was provided by a travel clinic which also performed the study

4. Departures from intended interventions: moderate

Discontinuations were reported across groups. It is unclear if information regarding switches was obtained

5. Missing data: low

All participants were included in the analysis. All information was included at one time point

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective, participants and personnel were not blinded

7. Selection of the reported results: moderate

The analysis of the relationship of symptoms by weight was reported only for mefloquine

8. Other: no information

No information was provided regarding the study sponsor

Cunningham 2014

Methods

Design: cross‐sectional cohort study

Study dates: questionnaire emailed July 2012, reminder emails were circulated at 8 and 12 weeks

Malaria transmission pattern and local antimalarial drug resistance: various destinations, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 579 questionnaires emailed, 327 responses

Inclusion criteria: all Foreign and Commonwealth Office staff posted to a malaria‐endemic area

Exclusion criteria: none stated

Factors influencing drug allocation: "prophylaxis based on the Advisory Committee on Malaria Prevention in UK Travellers (ACMP) guidelines"

Country of recruitment: various, not specified

Country of malaria exposure: various, not specified

Duration of exposure to malaria: 0 to 3 months N = 16 (4.9%), 4 to 6 months N = 26 (8.0%), 7 to 12 months N = 46 (14.1%), 13 to 36 months N = 75 (22.9%), > 36 months N = 167 (51.1%)

Type of participants: UK Foreign and Commonwealth Office staff

Interventions

1. Mefloquine*

2. Atovaquone‐proguanil*

3. Doxycycline*

4. Chloroquine*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; psychiatric disorders (abnormal dreams)

2. Adverse effects; other (skin sensitivity, indigestion, other psychological)

Outcomes assessed not included in the review:

3. Clinical cases of malaria

4. Background knowledge of malaria

5. Attitudes regarding malaria prophylaxis

6. Use of personal protective measures

7. Impact of pregnancy on malaria prevention

8. Measures of adherence to drug regimen (data provided on aggregate)

Notes

Funding sources: not mentioned

Communications with study authors: the study authors provided us with access to the full original data set. Thedata set differed from findings in the published version of the paper, and we were unable to determine the cause for differences. The included figures were from the full data set

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

No information on confounders was provided across prophylaxis groups

2. Selection of participants into the study: serious

Response rate for the survey was 56.5%

3. Measurement of interventions: moderate

Participants were asked to self‐report which medications they were prescribed. Compliance rate was 25%

4. Departures from intended interventions: serious

No questions were included in the questionnaire regarding switches between chemoprophylactic regimens

5. Missing data: low

All participants were included in the analysis

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

The entire questionnaire was provided in full, all outcomes included were reported

8. Other: no information

Study sponsor not mentioned

Davis 1996

Methods

Design: RCT

Study dates: not mentioned

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: daily self‐reported diary. Three medical check ups for laboratory and other tests

Participants

Number enrolled: 106 randomized, 95 completed all study procedures

Inclusion criteria: "healthy adult staff and students at teaching hospitals in Perth, Western Australia"

Exclusion criteria: "Those with a past history of psychiatric conditions, or neurological, cardiac, hepatic or renal disease were excluded, as were pregnant or breastfeeding females and those with a known allergy to, or taking medication known to interact with quinolone drugs. None of the subjects had taken mefloquine in the 3 months before the study"

Country of recruitment: Australia

Country of malaria exposure: not applicable

Duration of follow up: 7 weeks

Type of participants: non‐immune non‐travellers

Interventions

1. Mefloquine (1 x 250 mg tablet), with placebo dose followed 1 week later by 250 mg mefloquine weekly, active treatment duration 4 weeks

2. Placebo, 1 tablet weekly, duration 5 weeks

Outcomes

Included in the review:

1. Measure of adherence to the drug regimen

2. Adverse events: other outcome measures (symbol digit modalities test, digit span forwards and backwards test, ECG, hearing loss at 6kHz)

Outcomes assessed not included in the review:

3. Laboratory tests: serum glucose, insulin, ionized calcium, phosphate, magnesium and albumin concentrations

4. Adverse events: headache, lethargy, abdominal pain, diarrhoea, cough, nausea; study reports events occurring in the first week (after both groups had received placebo) and the relative risk of symptoms worsening over time

Notes

Funding sources: "We thank… F. Hoffman La Roche & Co. for financial support"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"...allocation… was by a random number code generated by independent Fremantle Hospital Pharmacy staff"

Allocation concealment (selection bias)

Low risk

"...who kept the code strictly confidential until the last volunteer had completed the protocol"

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"Tablets were prepared in individually numbered but otherwise unlabelled containers... identical placebo tablets…"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Allocation of active or placebo formulation was by a random number code generated by independent Freemantle hospital staff who kept the code strictly confidential"

Comment: not mentioned whether outcomes assessors were blinded

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Low risk

"Of 106 randomised volunteers, 95 (90%) completed all study procedures... eight subjects withdrew after initial assessment and three after the second. Follow‐up of these individuals revealed no toxicity in those allocated mefloquine"

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

High risk

Comment: not all symptoms were reported, only those occurring in > 10% of participants in both groups. Absolute numbers of participants experiencing each symptom after mefloquine/placebo commenced not provided, only relative risk of symptoms worsening over time

Other bias

High risk

"We thank… F. Hoffman La Roche & Co. for financial support"

Eick‐Cost 2017

Methods

Design: Retrospective cohort study

Study dates: 1 January 2008 to 30 June 2013

Malaria transmission pattern and local antimalarial drug resistance: Various, not specified

Adverse event monitoring: Data collected retrospectively from the Defense Medical Surveillance System, the Pharmacy Data Transaction Service and the Theater Medical Data Store

Participants

Number enrolled: 367,840

Inclusion criteria: Active component service members who filled a prescription for mefloquine, doxycycline or atovaquone‐proguanil

Exclusion criteria: Doxycycline and atovaquone‐proguanil prescriptions were excluded if the service member previously or concurrently received mefloquine. Doxycycline prescriptions were restricted to 100 mg, once daily, tabular form, minimum 30 day prescription

Factors influencing drug allocation: Not specified

Country of recruitment: USA

Country of malaria exposure: Various, not specified

Duration of exposure to malaria: Various, not specified

Type of participants: Military

Interventions

1. Mefloquine (250 mg weekly)

2. Atovaquone‐ proguanil*

3. Doxycycline (100 mg, tabular form, daily dose, 30 day minimum prescription)

*dosing regimen not specified

Outcomes

1. Adverse events (anxiety disorders, depressive disorders, psychoses, insomnia, vertigo)

2. Adverse events; other (adjustment disorders, post‐traumatic stress disorder, tinnitus, suicidal ideation, convulsions, hallucinations, paranoia, confusion)

Notes

Funding source: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Identified confounders were measured and not balanced across groups

2. Selection of participants into the study: low

Start of intervention and start of follow‐up coincided for most participants. Retrospective medical records were used, therefore there were no non‐responders

3. Measurement of interventions: moderate

Information regarding drug prescriptions were obtained from a medical database, without any verification that users took the prescription

4. Departures from intended interventions: serious

Discontinuations and switches between prophylactic regimes were not recorded in the database

5. Missing data: low

All records in the research database were included in the analysis

6. Measurement of outcomes: moderate

Participants and outcome assessors (physicians) were not blinded. However, information was collected anonymously and on aggregate. Participants were unaware of their participation at the time of seeking healthcare

7. Selection of the reported results: low

Outcome data were reported for all outcomes prespecified for analysis

8. Other: no information

No information was available regarding the study sponsor.

Goodyer 2011

Methods

Design: prospective cohort study

Study dates: December 2004 to April 2006

Malaria transmission pattern and local antimalarial drug resistance: various destinations, not specified

Adverse event monitoring: "a post travel questionnaire… approximately 1 week after they were due to complete their course of medication"

Participants

Number enrolled: 252 recruited, 185 completed pre‐ and post‐travel questionnaires

Inclusion criteria: "...to be eligible, travelers had to be at least 18 years of age and to have been prescribed or supplied... an antimalarial medication as a result of planned travel for a duration of 28 days or less."

Exclusion criteria: "travelers participating in other prospective clinical research or observational studies, pregnant travelers or travelers planning to get pregnant during the study were excluded"

Factors influencing drug allocation: "Treatment choice was solely at the discretion of the traveler and practitioner"

Country of recruitment: UK

Country of malaria exposure: various, not reported

Duration of exposure to malaria: various, median 14 days (interquartile range 9 to 20)

Type of participants: travellers

Interventions

1. Mefloquine*

2. Atovaquone‐proguanil*

3. Doxycycline*

*dosing regimen not specified

Outcomes

Included in the review:

1. Any adverse effects

2. Measures of adherence to the drug regimen

Outcomes assessed not included in the review:

3. Relative importance of factors in choice of antimalarial drugs, for both healthcare professionals and travellers

Notes

Funding sources: "The study was commissioned and paid for by GlaxoSmithKline"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

"There were statistically significant differences in mean age"

Several other confounders were not reported across groups

2. Selection of participants into the study: moderate

No information is provided regarding people who did not wish to participate

3. Measurement of interventions: low

The antimalarial prescription was provided by a travel clinic which also performed the study

4. Departures from intended interventions: moderate

No information was captured regarding switches between interventions of interest

5. Missing data: serious

185/252 participants completed the pre‐ and post‐travel questionnaire. Interim loss to follow up 27%

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

The number of reported side effects was reported, but not the types or severity

8. Other: serious

Funded by GlaxoSmithKline; the role of the study sponsor was not made clear

Hale 2003

Methods

Design: RCT

Study dates: not mentioned

Malaria transmission pattern and local antimalarial drug resistance: "the 20‐week cumulative incidence of reinfection by P. falciparum to be nearly 100%". No mention of local drug resistance patterns

Adverse event monitoring: "...during the prophylaxis and follow‐up phases, health workers visited the subjects 3 times weekly. Subjects with physical complaints were examined by a study physician the next day or on an emergent basis, as needed. Hematologic analysis was done on days 4 and 10 after starting the loading dose phase and during weeks 4, 8, 12, and 15. Biochemical analysis was done during weeks 4, 8, 12, and 15"

Participants

Number enrolled: 530 enrolled and completed radical cure regimen. 509 participants took at least 1 dose of the weekly study drug or placebo and comprised the full intention‐to‐treat data set

Inclusion criteria: "Inclusion criteria included the following: age of 18–60 years (men) or 50–60 years (women); lack of significant systemic illness as determined by history, physical examination, and clinical laboratory test results (including negative results of a urine pregnancy test for women); and absence of seizures or other neuropsychiatric illness (past or present)"

Exclusion criteria: "The high rate of pregnancy and breast‐feeding in women aged 18–49 years precluded their enrollment... G6PD deficiency accounted for 179 of 338 exclusions"

Country of recruitment: Ghana

Country of malaria exposure: Ghana

Duration of exposure to malaria: trial duration 12 weeks

Type of participants: Ghanain residents, semi‐immune

Interventions

Included in the review:

1. Mefloquine (1 x 250 mg tablet, salt), weekly, with supervised 3 day loading dose*

2. Placebo, with supervised 3 day loading dose*

Not included in the review:

3. Tafenoquine (1 x 25 mg tablet, base), weekly, with supervised 3 day loading dose*

4. Tafenoquine (1 x 50 mg tablet, base), weekly, with supervised 3 day loading dose*

5. Tafenoquine (1 x 100 mg tablet, base), weekly, with supervised 3 day loading dose*

6. Tafenoquine (1 x 200 mg tablet, base), weekly, with supervised 3 day loading dose*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1.Clinical cases of malaria

2. Adverse events; any, abdominal pain, diarrhoea, headache

3. Adverse events; other (gastritis, back pain, myalgia, polyarthralgia/arthralgia, respiratory tract infection, sore throat, rash)

4. Discontinuation of study drug due to adverse effects

Outcomes assessed not included in the review:

5. Laboratory tests; haematological and biochemical analyses

Notes

Funding sources: USA Army Medical Materiel Development

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"The randomization code was generated in blocks of 11 numbers"

Comment: not mentioned how randomization code was produced

Allocation concealment (selection bias)

Unclear risk

"Code numbers were assigned according to the chronological order of appearance of the subjects at screening. Study drugs were prepackaged and prelabeled with a unique study number according to the randomization code"

Comment: no mention of opaque sealed envelopes

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

"A ‘double‐dummy’ design allowed double‐blind administration of tafenoquine and mefloquine active drugs and their corresponding placebos"

"A placebo (tafenoquine placebo, GlaxoSmith‐Kline; mefloquine placebo, Hoffmann‐La Roche) served as the negative comparator"

Comment: does not report that the tablets were identical

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"All slides positive for the presence of malaria causing parasites, and an equal number of randomly selected slides with negative results were reevaluated by a second (blinded) microscopist."

Comment: no other mention of outcome assessors being blinded and does not report that the researchers were blinded

Incomplete outcome data (attrition bias); efficacy

Low risk

"Data analysis for efficacy used 2 data sets: the 'full, intent‐to‐treat' data set (n=509), comprising all subjects who took at least 1 dose of the weekly study drug or placebo, and the 'per‐protocol' data set (n=428), comprising those subjects who strictly fulfilled the protocol criteria"

Incomplete outcome data (attrition bias); safety

Low risk

Comment: The safety and tolerability analyses included data for all participants who received at least 1 dose of the study drug or placebo (N = 513)

Selective reporting (reporting bias); efficacy

Low risk

Comment: total number of participants with positive blood smear result at any time during prophylaxis was reported. Clinical cases of malaria were reported

Selective reporting (reporting bias); safety

High risk

"There were 9 serious adverse events in the study... No serious adverse events were considered by study physicians to be related to the study drug, and no deaths occurred"

Comment: Data for serious adverse events were not attributed to the drug regimen. No information was provided on how causality was assessed

Other bias

High risk

Acknowledgement of "Philip Pickford and Rachel Moate (GlaxoSmithKline), for statistical and editorial advisement"

Hill 2000

Methods

Design: retrospective cohort study

Study dates: June 1989 to May 1991

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire. "Any reported illness was followed up by telephone interview about the nature of the illness, during which time more complete information was obtained using standardized questions"

Participants

Number enrolled: 869 participants enrolled, 822 completed follow‐up

Inclusion criteria: all individuals attending the International Traveler’s Medical Service at the University of Connecticut Health Center and traveling for ≤ 90 days

Exclusion criteria: none mentioned

Factors influencing drug allocation: "prior to travel each person was given extensive counseling and written material on the prevention of malaria and traveler’s diarrhea. They were given prescriptions for prophylactic antimalarials"

Country of recruitment: USA

Country of malaria exposure: Various: Indian subcontinent 21%, central and east Africa 20%, South America 16%, Southeast Asia 14%, West Africa 10%, Central America and Mexico 10%, North Africa 65, East Asia 6%, Carribean 5%, Southern Africa 5%, Middle East 3%

Duration of exposure to malaria: median 19 days (up to 90 days)

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Chloroquine*

Not included in the review:

2. Chloroquine‐proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Any adverse effects

2. Discontinuations of study drug due to adverse effects

3. Measures of adherence to the drug regime

Outcomes assessed not included in the review:

4. Clinical cases of malaria

5. Adverse events (provided for entire cohort, not by type of malaria prophylaxis)

6. Adverse effects; other (all gastrointestinal disorders, all nervous system disorders ‐ no comparative data provided)

7. Illness during and following travel

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex, destination and duration of travel were measured but not reported across groups

2. Selection of participants into the study: moderate

Non‐response rate was not reported.

3. Measurement of interventions: low

The antimalarial prescription was provided by a travel clinic which also performed the study

4. Departures from intended interventions: moderate

Information was provided on discontinuations, but no information was captured on switches between interventions

5. Missing data: low

Information on adverse effects was available for all participants who ever filled the prescription for the study drug (571/612, 93%)

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

It is unclear which questions were included in the questionnaire. Information was provided on aggregate

8. Other: no information

No information provided on study sponsor

Hoebe 1997

Methods

Design: retrospective cohort study

Study dates: January to June 1995

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: one‐off telephone interview between 4 and 20 weeks post‐travell

Participants

Number enrolled: 454 eligible travellers, 300 successfully contacted and agreed to participate

Inclusion criteria: subjects who visited the travel vaccination service of the regional public health institute in Maastricht if they had returned from their journey to tropical countries between 4 and 20 weeks previously. The group of non‐users was formed by people who travelled either to tropical countries without malaria risk or to cities in malarious areas, and by travellers who were prescribed an antimalarial drug but did not commence use

Exclusion criteria: participants who had a serious adverse reaction to mefloquine in the first week

Country of recruitment: Netherlands

Region of malaria exposure: various; Asia, Africa, South America

Duration of exposure to malaria: mean ˜3 weeks (range 1 to 9 weeks)

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine (1 x 250 mg tablet) weekly, taken 1 week prior to leaving, during travel and 4 weeks after departure

2. Non‐users of antimalarials

Not included in the review:

3. Proguanil (1 x 100 mg tablet) twice daily, taken during travel and 4 weeks after departure

Outcomes

1. Adverse events; any, nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia, anxiety, depression, pruritis

2. Adverse events; other (palpitations, severity of symptoms, time point of symptoms in relation to drug taking)

3. Discontinuations of study drug due to adverse effects

4. Measure of adherence to the drug regimen

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Travel destination varies significantly between users of mefloquine and non‐users of prophylaxis (6.7% America mefloquine versus 29.0% non‐users)

2. Selection of participants into the study: low

13/454 (2.8%) of travellers successfully contacted refused to participate

3. Measurement of interventions: low

Prescription was provided by a travel clinic which also performed the study, and discontinuations were reported

4. Departures from intended interventions: moderate

No information regarding switches been interventions of interest was reported

5. Missing data: moderate

"If somebody discontinued drug use within a certain period, symptoms that occurred in the following period were not counted"

Comment: Mefloquine has a half life of 17 to 21 days

6. Measurement of outcomes: moderate

"The participants were specifically asked about symptoms instead of adverse effects...To hide our focus on symptoms as adverse effects of the drugs, participants were informed that the aim of the study was to investigate symptoms during travelling. We structured the questionnaire so that the interviewers asked about symptoms first and drug use last, in order to blind them to the drug used when addressing symptoms"

7. Selection of the reported results: low

All prespecified outcomes were reported.

8. Other: no information

Funding source was not mentioned

Jute 2007

Methods

Design: cross‐sectional cohort study

Study dates: 2003

Malaria transmission pattern and local antimalarial drug resistance: during the dry season (considered a low risk malaria season). Local chloroquine/proguanil resistance

Adverse event monitoring: Patient self‐reported questionnaire

Participants

Number enrolled: 90 questionnaires distributed, 68 responses

Inclusion criteria: "all expatriate employees at the mine"

Exclusion criteria: non mentioned

Country of recruitment: Mali

Country of malaria exposure: Mali

Duration of exposure to malaria: various, not specified

Type of participants: long‐term expatriates

Interventions

Included in the review:

1. Mefloquine

2. Doxycycline

3. Atovaquone‐proguanil

Not included in the review:

4. Chloroquine‐proguanil

Outcomes

1. Adverse effects; any

Notes

Study sponsor not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex was recorded but not reported across chemoprophylaxis groups. Duration of travel was not reported. Destination of travel was set by the study design.

2. Selection of participants into the study: serious

68/90 response rate (76%)

3. Measurement of interventions: no information

It was unclear whether information on participants chemoprophylaxis was taken from medical records or patient self‐reporting

4. Departures from intended interventions: moderate

No information regarding switches between interventions of interest were reported. Discontinuations were reported

5. Missing data: low

All information was collected at one time point

6. Measurement of outcomes: serious

The outcome measure was subjective. There was no mention of participants or outcome assessors being blinded.

7. Selection of the reported results: no information

No information was provided regarding which topics were included within the questionnaire

8. Other: no information

Funding source was not mentioned

Kato 2013

Methods

Design: cross‐sectional cohort study

Study dates: June 2009 to June 2011

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 1119 eligible travellers, 316 enrolled

Inclusion criteria: "travelers who visited Hibiya Clinic, and requested antimalarial drugs for malaria chemoprophylaxis from June 2009 to June 2011"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "The choice of anti‐malarial drug was supported by sufficient explanation about the advantages and disadvantages (efficacy, method, duration, side effect, cost and approval) of each drug"

Country of recruitment: Japan

Region of malaria exposure: various (n): East Africa 76, West Africa 63, South Africa 50, Southeast Asia 36, Central Africa 36, South Pacific 21, South America 16, India 8, North Africa 5, Central America 1

Duration of exposure to malaria: mean 20.0 ± 9.6 days in the atovaquone‐proguanil group and 59.0 ± 15.9 days in the mefloquine group

Type of participants: travellers

Interventions

1. Mefloquine (1 x 250 mg tablet, Mephaquin; Mepha) weekly, starting 1 week prior to arrival, during the stay, and continuing for 4 weeks after leaving the endemic area

2. Atovaquone‐proguanil (1 tablet containing 250 mg atovaquone and 100 mg proguanil, Malarone; GlaxoSmithKline) daily, starting 2 days prior to arrival, during the stay, and for 1 week after leaving the endemic area

Outcomes

1. Adverse effects (any vertigo/dizziness, nausea, abdominal pain, diarrhoea, headache, insomnia, depression, any cardiovascular, any gastrointestinal, any psychoneurotic, allergic reaction)

2. Discontinuations of study drug due to adverse effects

Notes

Funding sources: not mentioned

Communications with the study authors: the study authors provided us with disaggregated study data for the following outcomes: vertigo/dizziness, nausea, abdominal pain, diarrhoea, headache, insomnia, depression. Because we did not get receive the full disaggregated data set, we also retained this study in the analysis of groups of symptoms

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

PTravellers in the mefloquine group were significantly younger than travellers in the A/P group (p=0.01)"

2. Selection of participants into the study: serious

"316 of 1119 travelers (28.2 %) were enrolled"

3. Measurement of interventions: low

The prescription has been provided by travel clinic which also performed the study and discontinuations have been reported

4. Departures from intended interventions: moderate

No information was available regarding switches between interventions of interest

5. Missing data: low

One participant in the mefloquine group appears to be missing from the adverse events analysis. No reason was given

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

Study authors provided us with disaggregated study data for individual outcomes

8. Other: serious

"The authors wish to acknowledge that Makoto Ono and Tomoko Kawamura of GlaxoSmithKline are highly appreciated for conducting Data Management and Statistics Analysis of this study"

Korhonen 2007

Methods

Design: prospective cohort study

Study dates: 1 August 2005 to 31 July 2006.

Malaria transmission pattern and local antimalarial drug resistance: various, chloroquine resistance specified by country of destination

Adverse event monitoring: "Peace Corps medical staff in these countries were provided surveys for distribution during mandatory in‐country volunteer training sessions"

Participants

Number enrolled: 2701 (6216 Peace Corps volunteers during the time period)

Inclusion criteria: "all Peace Corps countries with malaria risk"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "Volunteers are provided chemoprophylaxis (either chloroquine, mefloquine, doxycycline, or atovaquone/proguanil)... medical officers can provide alternative chemoprophylaxis regimens for volunteers when adverse events or other factors require the cessation of any medication"

Country of recruitment: various

Country of malaria exposure: various

Duration of exposure to malaria: "6 months or longer"

Type of participants: Peace Corps volunteers

Interventions

Included in the review:

1. Mefloquine*

2. Chloroquine*

3. Doxycycline*

4. Atovaquone‐proguanil*

*dosing regimen not specified

Outcomes

1. Adverse effects; any (mild, moderate, severe, sought medical advice), nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia, depression, anxiety, visual disturbance

2. Adverse effects; other (unsteadiness, hair loss, weakness, itchy skin, photosensitivity, yeast infection)

3. Serious adverse effects

4. Discontinuations of study drug due to adverse effects

Notes

Funding sources: "CK and PJ are employed by the Peace Corps, which has a significant number of volunteers taking anti‐malarial medications. There were no other financial disclosures"

Communications with study authors:

The study authors provided us with access to the disaggregated study data for the specific symptoms mentioned above. The questionnaire in the paper allowed participants to describe side effects from the antimalarial they were currently taking, and any regimen they had previously used. For non‐serious side effects, in line with the original paper, we only included side effects for the subject's original regimen. Where subjects had previously taken more than one regimen, we only include side effects for whichever regimen to which the participant attributed the greater number of side effects; this affected 70/2701 participants. This analysis resulted in a decrease in the effect size for side effects attributed to mefloquine. For serious side effects (hospitalizations) and discontinuations we included all participants entries for all regimens. In addition, our denominator differed from the original paper because we did not exclude participants who had been in post for fewer than six months

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

"The questionnaire did not collect demographic information because of privacy concerns"

Comment: destination has been reported, but not by type of antimalarial chemoprophylaxis. Duration was set by the study design

2. Selection of participants into the study: serious

"A total of 2701 surveys were received yielding a response rate of 43%"

3. Measurement of interventions: moderate

Participants were asked to self‐report which prophylaxis they were currently taking and had previously taken

4. Departures from intended interventions: moderate

Switches between interventions of interest were reported. Approximately 1/3 of study participants had switched prophylactic regimens

5. Missing data: low

We were able to include all participants in the study analysis because we had access to the original data set

6. Measurement of outcomes: serious

"If respondents identified any adverse event, the survey instructed them to self‐report which drug they believed caused the adverse event"

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

We were able to include all results in the analysis because we had access to the original data set

8. Other: low

No evidence of pharmaceutical company funding

Kuhner 2005

Methods

Design: prospective cohort study

Study dates: 2000 to 2003

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: retrospective patient self‐reporting questionnaire

Participants

Number enrolled: 495 enrolled, 284 response rate

Inclusion criteria: unclear. Users of the travel medicine department of the lower Saxony regional health office in Hanover, Germany

Exclusion criteria: None mentioned

Factors influencing drug allocation: "the prescriptions of medications followed individual consultation"

Country of recruitment: Germany

Country of malaria exposure: various, not specified

Duration of exposure to drug: atovaquone‐proguanil mean 2.6 weeks, mefloquine mean 7 weeks

Type of participants: short‐term travellers

Interventions

Included in the review:

1. Mefloquine*

2. Atovaquone‐proguanil*

Not included in the review:

3. Chloroquine‐proguanil*

4. Chloroquine (not included in the study analysis)

*dosing regimen not specified

Outcomes

1. Adverse effects; any, nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia, pruritis

2. Adverse effects; other (concentration difficulties, palpitations, circulation disorders, rash)

3. Discontinuations of study drug due to adverse effects

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex, age and duration of travel were reported but not balanced across groups

2. Selection of participants into the study: serious

284/495 (59.8%) response rate

3. Measurement of interventions: low

The prescription was provided by a travel clinic which also performed the study; switches and discontinuations were recorded and reported.

4. Departures from intended interventions: moderate

No information was provided regarding switches between prophylactic regimens

5. Missing data: low

All information was collected at one time point

6. Measurement of outcomes: serious

The outcome measure was subjective. There was no mention of outcome assessors being blinded

7. Selection of the reported results: moderate

Insufficient information was provided regarding the questionnaire to know whether all outcomes were reported

8. Other: no information

Study sponsor not mentioned

Landman 2015

Methods

Design: prospective cohort study

Study dates: 19 August to 30 September 2013

Malaria transmission pattern and local antimalarial drug resistance: various

Adverse event monitoring: participant self‐reported questionnaire

Participants

Number enrolled: 3207 emails sent, 1184 unique, valid responses received

Inclusion criteria: "(volunteers in) Peace Corps offices of all 23 countries with active posts in the Africa region to all active Volunteers in‐country"

Exclusion criteria: Volunteers serving in Ethiopia, Kenya, Tanzania, Namibia, Botswana, South Africa

Region of recruitment: African region except Ethiopia, Kenya, Tanzania, Namibia, Botswana, South Africa

Factors influencing drug allocation: "all prophylaxis options (mefloquine, doxycycline, atovaquone‐proguanil) [are] equally available... They are instructed to individualize their choice of agent based on area‐specific recommendations, drug contraindications and precautions, drug tolerance, and dosing schedule"

Country of malaria exposure: various: Togo (3.7%), Sierra Leone (6.3%), Uganda (7.8%), Liberia (5.6%), Malawi (2.0%), Cameroon (11.4%), Benin (10.2%), Burkina Faso (1.9%), Zambia (6.0%), Mozambique (4.5%), Ghana (10.8%), Rwanda (5.4%), Gambia (4.4%), Madagascar (11.1%), Swaziland (2.3%)

Duration of exposure to malaria: various, not specified

Type of participants: Peace Corps volunteers

Interventions

1. Mefloquine*

2. Atovaquone‐proguanil*

3. Doxycycline*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any, vertigo, headache, abnormal dreams, insomnia, anxiety, depression, psychosis

2. Adverse effects; other (any neuropsychiatric disorder, any gastrointestinal disorder, any skin or subcutaneous disorder, limb numbness, tinnitus, 'constitutional', genitourinary)

3. Measures of adherence to the drug regimen

Outcomes assessed not included in the review:

4. Reasons for non‐adherence (not ascribed to prophylactic regimen, provided on aggregate),

5. Malaria knowledge

6. Health behaviours

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

The age, sex and BMI of included participants was not recorded. The destination and duration of travel was not reported by prophylactic regimen

2. Selection of participants into the study: serious

1184/3248 (36%) response rate

3. Measurement of interventions: moderate

Travellers were asked to self‐report which prophylaxis they were taking at various time points during treatment

4. Departures from intended interventions: serious

"Two hundred seventy‐six (35%) respondents reported having changed prophylaxis at some point during their service"

Comment: this was not provided by prophylactic regimen

5. Missing data: low

703/781 (90%) participants reported data for adherence; 733/781 (94%) participants reported data for adverse events. Data were only included from the 2015 version of the publication

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

All outcomes prespecified in the methods section were reported

8. Other: no information

Study sponsor not mentioned

Laver 2001

Methods

Design: cross‐sectional cohort study

Study dates: February 2000

Malaria transmission pattern and local antimalarial drug resistance: "during February 2000, which was a peak period of malaria transmission in Zimbabwe"

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 660

Inclusion criteria: Passengers in Harare and Victoria Falls international airport during February 2000

Exclusion criteria: "Children under the age of 18 were excluded on the assumption that parents probably influence their health seeking behavior... Excluded, were travelers from the African continent and VIP travelers who exited through special departure lounges"

Factors influencing drug allocation: no infromation provided

Country of recruitment: Zimbabwe

Country of malaria exposure: Zimbabwe

Duration of exposure to malaria: various: 1 week or less, N = 317; 8 days to 2 weeks, N = 144; 15 days to 4 weeks, N = 90; > 4 weeks, N = 41

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Doxycycline*

3. Chloroquine*

Not included in the review:

4. Proguanil*

5. Dapsone and pyrimethamine*

6. Chloroquine and proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

2. Sources of pre‐travel health advice

3. Knowledge about malaria transmission

4. Knowledge about malaria prevention

5. Threat and risk perception

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex (P < 0.008), education (P < 0.022), previous episodes of malaria (P < 0.001) and access to pre‐travel advice (P < 0.001) were all significantly associated with reduced compliance at the significance value set by the study. None of these factors were adjusted for in the analysis

2. Selection of participants into the study: moderate

"The nonresponse rate was about 10% (n = 65), with the main reason being the short transit time"

3. Measurement of interventions: low

Participants were asked to self‐report which prophylactic regimen they were taking while they were still taking it

4. Departures from intended interventions: moderate

No information was provided regarding switches between prophylactic regimens

5. Missing data: low

Adherence information was not available for 4/595 participants

6. Measurement of outcomes: serious

The outcome measure was based on participant self‐reporting; participants and personnel were not blinded.

7. Selection of the reported results: moderate

There was insufficient information provided to know what questions were asked regarding adherence

8. Other: low

"The authors had no financial or other conflicts of interest to disclose"

Laverone 2006

Methods

Design: retrospective cohort study

Study dates: 1 January 2003 to 31 December 2004

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: "An anonymous survey in a post‐travel situation"

Participants

Number enrolled: 1176 agreed to participate, 1237 approached

Inclusion criteria: "travellers who had already completed their journey for which they had undergone immunization prophylaxis and who had returned to complete their vaccination schedule"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "offered health advice following the World Health Organization guidelines for international travel"

Country of recruitment: Italy

Regions of malaria exposure: 97 countries: 39 states in Africa, 25 in Asia, 16 in North and Central America, 8 in South America, 6 in Europe and 3 in Oceania

Duration of exposure to malaria: 1 to 7 days, 8.9%; 8 to 14 days, 30.1%; 15 to 21 days, 34.6%; 22 to 30 days, 16.8%; > 30 days, 8.9%; not available 0.7%

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Atovaquone‐proguanil*

3. Chloroquine*

Not included in the review:

4.Chloroquine‐proguanil*

5. Proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any, visual impairment (blurred vision), nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams (nightmares), insomnia, anxiety (anxiety disorder), depression, psychosis (hallucinations)

2. Adverse effects; other (slight illness, tiredness, restlessness, drowsiness, palpitations, weakness, photosensitization, mental confusion, rash)

Outcomes assessed not included in the review:

3. Adverse effects; other, incidence < 1% (liver pain, aerophagy, rise in transaminase levels, gastrointestinal disturbance, epistaxis, fever)

4. Compliance with vaccinations

5. Side effects from vaccinations

6. Occurrence of health problems and unforeseen events during travel in the countries visited

7. Attention to avoiding potentially risky food and drink

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Demographic information was collected, but provided on aggregate for the entire cohort

2. Selection of participants into the study: low

1176 of 1237 (95.1%) response rate

3. Measurement of interventions: serious

Participants were asked to self‐report which prophylactic regimen they had used, up to over 12 months since travelling

4. Departures from intended interventions: serious

No switches were reported, and this information was not sought in the questionnaire

5. Missing data: low

642/646 (99%) participants were included in the analysis

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

The questionnaire was provided in full, and all outcomes were reported

8. Other: no information

No information was provided regarding the study sponsor

Lobel 2001

Methods

Design: cross‐sectional cohort study

Study dates: 13 July to 9 August 1997

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 6633 respondents, 5626 met inclusion criteria

Inclusion criteria: "travelers departing Nairobi, or Mombasa, Kenya, from July 13 to August 9, 1997, on flights to Europe, including London, Paris, Frankfurt, Amsterdam, and Rome"

Exclusion criteria: residents of African countries, individuals who had remained in Africa for more than 1 year, individuals who visited only non malarious areas, including Nairobi and Lesotho

Factors influencing drug allocation: no information available

Region of recruitment: Nairobi or Mombasa, Kenya

Region of malaria exposure: Nairobi or Mombasa, Kenya

Duration of exposure to malaria: < 5 weeks

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Doxycycline*

3. Chloroquine*

Not included in the review:

4. Chloroquine‐proguanil*

5. Proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any,

2. Serious adverse outcomes

3. Adverse effects; other (neuropsychologic, gastrointestinal, respiratory)

4. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

5. Pre‐travel medical advice

6. Compliance with antimosquito measures

7. Self‐treatment of presumed malaria

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

The number of travellers and country of origin was reported, but was not adjusted for in the analysis. Sex, age and duration of stay were reported on aggregate.

2. Selection of participants into the study: serious

Response rate 6633/15,487 (43%)

3. Measurement of interventions: low

Participants were asked to provide information regarding their prophylactic regimen during their flight home, while they should have still been using it

4. Departures from intended interventions: moderate

No information was available regarding switches between alternative prophylactic regimens

5. Missing data: low

4934/4982 (99%) participants included in adverse event reporting

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

There was insufficient information provided regarding the questions included in the questionnaire. Symptoms were grouped together to report outcomes

8. Other: low

"The authors had no financial or other conflicts of interest to disclose"

Mavrogordato 2012

Methods

Design: retrospective cohort study

Study dates: October to December 2005, with a 2 year follow‐up

Malaria transmission pattern and local antimalarial drug resistance: "Malaria endemic area. Local chloroquine/proguanil resistance"

Adverse event monitoring: Not clear

Participants

Number enrolled: 33

Inclusion criteria: not explicitly stated. Participants were travellers who took part in a scientific survey and rafting expedition in Ethiopia between October and December 2005

Exclusion criteria: none stated

Country of recruitment: various, participants were from "a non‐malarious area, mainly the UK"

Country of malaria exposure: Ethiopia

Duration of exposure to malaria: 3 months

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine, dose not specified, during travel and 4 weeks after return

2. Atovaquone‐proguanil, dose not specified, during travel and for 1 week after return

3. Doxycycline, dose not specified, during travel and 4 weeks after return

Not included in the review:

4. Chloroquine‐proguanil, dose not specified, during travel and 4 weeks after return

Outcomes

Included in the review:

1. Measures of adherence to the drug regimen

Outcomes assessed not included in the review:

2. Clinical cases of malaria

3. Adverse effects (information not provided by drug class)

4. Factors influencing choice of prophylaxis

Notes

Funding sources: Work was supported by the Biomedical Research Centre (Grant RG561620 to AMLL)

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Demographic information is provided for the entire cohort

2. Selection of participants into the study: low

No participants refused to participate in the study. Start of follow‐up began at the start of travel and not at the start of treatment, but this was judged to have a low impact on monitoring self‐reported adherence

3. Measurement of interventions: low

Intervention status was determined by one of the participants on the expedition

4. Departures from intended interventions: low

There are no documented switches between interventions of interest

5. Missing data: low

Two people (6%) were lost to follow‐up in respect to data on efficacy. No participants were lost to follow‐up when monitoring adherence

6. Measurement of outcomes: serious

Adherence was monitored by the medical officer on the trip, and reporting may have been influenced by social desirability bias

7. Selection of the reported results: low

All prespecified outcomes have been reported

8. Other: low

Government funding

Meier 2004

Methods

Design: retrospective cohort study

Study dates: 1 January 1990 and 31 December 1999

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: incident cases of depression, psychoses and panic attacks severe enough to require hospitalisation, referral to a specialist or specific pharmacological treatment within the UK general practice research database

Participants

Number enrolled: 35,370

Inclusion criteria: "men and women aged 17‐79 years who received between one and four prescriptions for mefloquine, proguanil and/or chloroquine, or subjects who received one prescription only for doxycycline... we included only those subjects who medical record contained a code indicating that the person received the drug for malaria prophylaxis within 1 week of the prescription date e.g. ‘travel advice’ or ‘prophylactic drug use’"

Exclusion criteria: "participants who received the study drugs on a longer‐term basis...subjects had to be enrolled in the database for at least 12 months before the date of the first prescription for a study drug and had to have had some recorded activity (diagnoses or drug prescriptions) after the prescription(s) for an antimalarial drug... subjects with a history of alcoholism"

Country of recruitment: UK

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Doxycycline*

Not included in the review:

3. Chloroquine‐proguanil*

4. Proguanil*

5. Chloroquine* (data reported combined with proguanil and chloroquine‐proguanil)

*dosing regimen not specified

Outcomes

1. Serious adverse events

2. Adverse events; psychiatric disorders (depression, psychosis)

3. Adverse events; other (panic attacks, suicide)

Notes

Funding sources: "This study was funded by an unconditional grant by F. Hoffmann‐La Roche Ltd, Basel, Switzerland"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Women and those aged 40 to 49 years were at higher risk of depression but this was not adjusted for in the analysis. Risk ratio estimates for psychoses and panic attacks could not be adjusted for because numbers were too small for the multivariate model. Data on destination and duration of travel were not available

2. Selection of participants into the study: low

Recruitment onto the General Practice Research Database was unlikely to be related to exposure or outcome

3. Measurement of interventions: moderate

"Antimalarial drugs can be used for malaria prophylaxis, for treatment of an acute malaria infection, or as a reserve drug… In order to distinguish these options, we included only those subjects whose medical records contained a code indicating ‘travel advice’ or ‘prophylactic drug use’"

4. Departures from intended interventions: serious

Discontinuations and switches between prophylactic regimens were not recorded in this database

5. Missing data: low

All participants in the research database were included in the analysis

6. Measurement of outcomes: moderate

"...we reviewed all computer records of potential cases and included or excluded cases on the available clinical information, blinded to exposure status"

Comment: general practitioners diagnosing patients would have been aware of their exposure status

7. Selection of the reported results: low

Information on all outcomes prespecified in the methods section were reported for all participants.

8. Other: serious

Funded by Roche pharmaceuticals

Napoletano 2007

Methods

Design: retrospective cohort study

Study dates: 1 October 2005 to 30 June 2006

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: telephone questionnaire to all travellers to tropical countries for whom antimalarial chemoprophylaxis was prescribed

Participants

Number enrolled: 1906 questionnaires returned

Inclusion criteria: participants staying in high risk malarial areas, aged between 18 and 65 years, with no severe underlying disease (e.g. heart disease, diabetes) with an available phone number

Exclusion criteria: immigrants (due to potential difficulty in linguistic communication)

Country of recruitment: Italy

Country of malaria exposure: various: Kenya, Tanzania/Zanzibar, India, Madagascar, Brazil, other countries of South America, South Africa, Senegal, Mali, Myanmar, Ghana, Congo, and others

Duration of exposure to malaria: mean stay 2 weeks

Type of participants: Travellers

Interventions

Included in the review:

1. Mefloquine*

2. Chloroquine*

3. Atovaquone + proguanil*

4. Doxycycline*

Not included in the review:

5. Chloroquine + proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any

2. Serious adverse effects

3. Adverse effects; other (any gastrointestinal, any neuropsychiatric)

4. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

5. Clinical cases of malaria

6. Eating habits during travel

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Demographic information was provided on aggregate for the entire cohort

2. Selection of participants into the study: moderate

Non‐response rates to the questionnaire were not reported

3. Measurement of interventions: moderate

The prescription was provided by several travel clinics which also performed the study. However, it was unclear whether this information was used to determine intervention status or relied on participant self‐reporting

4. Departures from intended interventions: low

Discontinuations were reported, with detailed reasons for discontinuations. No switches to alternative regimens were reported

5. Missing data: low

All participants were included in the analysis

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: low

The methods section makes clear which outcomes were being assessed; all outcomes were reported

8. Other: no information

No information was provided regarding the study sponsor

Nosten 1994

Methods

Design: RCT

Study dates: January 1987 to November 1990

Malaria transmission pattern and local antimalarial drug resistance: "in an area of seasonal malaria transmission... mefloquine and quinine resistance is increasing in this area, and the proportion of recrudescent infections is rising"

Adverse event monitoring: trial occurred over two phases. Phase 1: Weekly basic observations and simple symptom questionnaire. ECG, haematological and biochemical tests were done fortnightly. Children born to women in the trial were assessed at birth and at 3, 6, 12, and 24 months. Phase 2: weekly basic observations and expanded simple symptom questionnaire. ECG and blood tests were performed at baseline, at midstudy and at term. Each delivery was supervised. Additional assessments at 1 week and 2 and 9 months for children born to women in the trial

Participants

Number enrolled: 339

Inclusion criteria: "Women attending the weekly clinic were admitted to the study if they were at > 20 weeks of estimated gestation"

Exclusion criteria: Not mentioned

Region of recruitment: Thai‐Burmese border

Region of malaria exposure: Thai‐Burmese border

Duration of exposure to malaria: ongoing exposure in a semi‐immune population, monitored until delivery

Type of participants: Pregnant Thai residents in malaria‐endemic area (presumed semi‐immune)

Interventions

1. Mefloquine (1 x 250 mg tablet, Lariam; Hoffmann‐La Roche) weekly for 4 weeks, then 125 mg weekly until delivery, with 500 mg base loading dose in phase 1 but not phase 2

2. Placebo (1 tablet) weekly until delivery

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Episodes of parasitaemia

3. Serious adverse events (including childhood deaths)

4. Adverse events; vertigo, visual impairment (visual abnormalities), nausea, vomiting, abdominal pain, headache, dizziness, pruritis

5. Adverse events; other (weakness, anorexia, cough, falls, constipation, unsteadiness)

6. Discontinuation of study drug due to adverse effects

7. Adverse pregnancy outcomes (spontaneous abortions, still births, congenital malformations)

Outcomes assessed not included in the review:

8. Laboratory tests; haematologic (full blood count, haematocrit) and biochemical (creatinine, blood urea, transaminases, alkaline phosphatase, albumin, globulin)

9. Outcomes related to pregnancy; weight gain during follow‐up, complications of labour, mean duration of labour, maternal anaemia

10. Fetal outcomes; mean birth weight, percent premature, fetal distress

11. Infant follow up; mean age at which children could crawl, sit, walk or talk, Romberg test

Notes

Funding sources: United Nations Development Programme/World Bank/World Health Organization Special Programme for Research and Training in Tropical Diseases; Wellcome Trust of Great Britain; Praevention Foundation. The Hague (to FLK)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"...women were randomized to receive either mefloquine…or placebo"

Comment: unclear what method of randomization was used

Allocation concealment (selection bias)

Unclear risk

"...the investigators were unaware of the randomisation"

Comment: no mention of method used to conceal allocation

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"...double blind...women were randomised to receive either mefloquine…or identical placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"...the investigators were unaware of the randomisation"

Incomplete outcome data (attrition bias); efficacy

Low risk

Comment: total number of participants with positive blood smear result at any time during prophylaxis was reported. Clinical cases of malaria were reported"

Incomplete outcome data (attrition bias); safety

High risk

"Ten women (8%) in phase I (3 mefloquine, 7 placebo) and 18 (8%) in phase II (9 in each group) dropped out of the study. The main reason was the discomfort of blood sampling (26 cases) and, in 1 case, pruritus attributed to mefloquine"

Comment: 28 women dropped out but reasons were provided for only 27 women; numbers were not provided across groups

Selective reporting (reporting bias); efficacy

Low risk

Comment: all episodes of parasitaemia and clinical cases of malaria were reported

Selective reporting (reporting bias); safety

High risk

Comment: Data on adverse effects were reported for only participants from phase 2 of the trial (220/339 women). Fifteen symptoms were listed in the comparative table, but the narrative states "twenty questions were asked". Romberg test results were not reported. Biochemical, haematological and ECG parameters were not reported other than "there were no differences"

Other bias

Low risk

Funding: United Nations Development Programme/World Bank/World Health Organization Special Programme for Research and Training in Tropical Diseases; Wellcome Trust of Great Britain; Praevention Foundation. The Hague (to FLK)

Ohrt 1997

Methods

Design: RCT

Duration of study: May to July 1994

Malaria transmission pattern and local drug resistance: "P. falciparum resistant to sulfadoxine‐pyrimethamine and both P falciparum and P vivax resistant to chloroquine"

Adverse event monitoring: symptoms reported in the first week of the study, daily questioning about symptoms, exit questionnaire

Participants

Number enrolled: 204

Inclusion criteria: "All soldiers from military posts that were considered to have high malaria attack rates"

Exclusion criteria: history of frequent travel, allergy to one of the study drugs, glucose‐6‐phosphate dehydrogenase deficiency, history of underlying illness

Country of recruitment: Indonesia

Country of malaria exposure: Indonesia

Duration of exposure to malaria: Study duration was approximately 13 weeks

Type of participants: military, semi‐immune (60% of participants had prior exposure to malaria)

Interventions

1. Mefloquine (1 x 250 mg tablet, containing the equivalent of 228 mg mefloquine base) once weekly (after a loading dose of 250 mg per day for 3 days).*

2. Doxycycline hyclate (1 x 100 mg capsule) once daily*

3. Placebo*

Co‐interventions: All soldiers were given doxycycline tablets for 4 to 6 weeks to enable clearance of sulfadoxine‐pyrimethamine from the blood before study prophylaxis began. All participants received radical treatment for pre‐existing malaria parasites in the blood and liver prior to beginning study prophylaxis.

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Adverse events; any, nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, insomnia, abnormal dreams

3. Serious adverse events

4. Adverse events; other (all gastrointestinal, all neurologic, constipation, anorexia, fever, malaise, skin related, cough, somnolence, palpitations, sexual dysfunction)

5. Discontinuation of study drug due to adverse effect

Outcomes assessed not included in the review:

6. Exit questionnaire (incomplete data reported)

Notes

Funding source: Pfizer Indonesia supplied active and placebo doxycycline; F. Hoffman‐La Roche supplied active and placebo mefloquine, and gave financial support; USA Army Medical Research and Materiel Command gave financial support; USA Naval Medical Research and Development Command gave financial support

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Block randomization was used (block size, 15)"

Comment: Used a randomization code, but it was not stated how it was generated

Allocation concealment (selection bias)

Unclear risk

"The randomization code was stored in individual envelopes in a locked box at the study site...Drugs were packaged into weekly ziplock plastic bags"

Comment: Unclear whether the investigators or participants would foresee assignment. There was no mention of central allocation, sequentially numbered drug containers or sequentially numbers opaque sealed envelopes

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"Drugs were packaged into weekly zipper‐lock plastic bags: each bag contained a mefloquine or mefloquine placebo tablet and a blister pack of seven doxycycline or doxycycline placebo capsules (double‐dummy technique)"

The placebo medication had an "identical appearance"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The randomisation code was stored in individual envelopes in a locked box at the study site. All investigators and study personnel did not have access to or know the randomisation code throughout the study"

Incomplete outcome data (attrition bias); efficacy

Unclear risk

"Sixteen of the 204 participants did not complete the study"

Comment: It was unclear whether the duration of follow up included the post‐prophylaxis period to monitor for relapses

Incomplete outcome data (attrition bias); safety

High risk

Exit questionnaire: "Only data from persons who were still receiving the study drug at the time of the questionnaire were included"

Comment: numbers not reported

Selective reporting (reporting bias); efficacy

Low risk

"The primary end point for efficacy was the first occurrence of malaria, as documented by a positive malaria smear"

Comment: all cases of malaria were reported.

Selective reporting (reporting bias); safety

High risk

Comment: Not all data were reported from the exit questionnaire; the study reports "...the only statistically significant finding". Data on adverse symptoms were not reported for the placebo group

Other bias

Low risk

"Neither of the pharmaceutical companies that provided support played any role in the gathering, analysing or interpreting the data"

Overbosch 2001

Methods

Design: RCT

Duration of study: April to October 1999

Malaria transmission pattern and local drug resistance: not mentioned

Adverse event monitoring: "evaluated 7, 28 and 60 days after return to obtain information about a targeted list of adverse events"

Participants

Number enrolled: 1013

Inclusion criteria: "travellers aged ≥ 3 years and weighing ≥ 11 kg with planned travel of ≤ 28 days to a malaria‐endemic area"

Exclusion criteria: "poor general health; drug hypersensitivity (to atovaquone, chloroquine or proguanil); history of alcoholism, seizures or psychiatric or severe neurological disorders; generalized psoriasis; severe blood disorders; pregnancy/lactation; renal, hepatic or cardiac dysfunction; clinical malaria within previous 12 months; travel to malaria endemic area within previous 60 days"

Countries of recruitment: Canada, Germany, Netherlands, South Africa, UK

Regions of malaria exposure: various malaria‐endemic destinations (79% Africa, 6% South America)

Mean duration of exposure to malaria: 2.5 weeks

Type of participants: travellers, non‐immune

Interventions

1. Mefloquine (1 x 250 mg tablet; or alternatively ¼, ½ or ¾ of a tablet, according to body weight) once weekly, starting 1 to 3 weeks before travel and continuing for 4 weeks after travel*

2. Atovaquone‐proguanil (1 combined tablet containing 250 mg atovaquone and 100 mg proguanil hydrochloride; or alternatively 1 to 3 combined tablets for children according to body weight, each tablet containing 62.5 mg atovaquone and 25 mg proguanil hydrochloride) once daily, starting 1 to 2 days before travel and continuing for 1 week after leaving the malaria‐endemic area*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria (antibody to blood‐stage malaria parasites)

2. Adverse events; any

3. Serious adverse events

4. Adverse effects; any (moderate or severe), visual impairment, nausea, vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia, anxiety, depression, pruritis

5. Adverse effects; other (mouth ulcers)

6. Discontinuation of study drug due to adverse effects

7. Measures of adherence to the drug regimen

Outcomes assessed not included in the review:

8. Laboratory tests; haematology (haemoglobin level, white blood cell count and platelet count) and chemistry (creatinine and alanine aminotransferase)

Notes

Funding source: GlaxoSmithKline

"Subjects were enrolled in study MAL30010"‐ Enrollment criteria and study conduct were described in a separate publication (Høgh 2000) which refers to a different study population (atovaquone‐proguanil versus chloroquine‐proguanil).

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A computer‐generated code was used to randomly assign a treatment number" (Høgh 2000)

Allocation concealment (selection bias)

Low risk

"Treatment codes were provided to investigators in opaque sealed envelopes, to be opened only if knowledge of study drug assignment was required for management of a medical emergency" (Høgh 2000)

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"For each active drug, capsules or film‐coated tablets were identical in appearance to the matching placebo"

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"All subjects and study personnel remained blinded to treatment assignment with 5 exceptions. Two subjects in the atovaquone‐proguanil group and 3 in the mefloquine group lost their study drug during their return trip from a malaria‐endemic area, and the investigator broke the blind to enable completion of postexposure prophylaxis with active drug"

Incomplete outcome data (attrition bias); efficacy

Low risk

"A total of 963 subjects completed the 60‐day follow‐up period and had efficacy information recorded. A total of 915 subjects had paired serum samples available for serological testing"

Comment: 963/976 (randomized and received first dose of study drug) = 98.7%. 915/976 = 93.75%. Reasons for leaving the study early were reported and numbers were balanced across groups

Incomplete outcome data (attrition bias); safety

Unclear risk

Comment: 96.35% of randomized participants were included in adverse event reporting. Reasons for leaving the study early were reported and numbers were balanced across groups

Selective reporting (reporting bias); efficacy

Low risk

Comment: Full clinical details were provided for every episode in which an episode of malaria was considered (4 cases)

Selective reporting (reporting bias); safety

High risk

Comment: Data on adverse symptoms were not reported for the placebo group due to a shorter duration of follow‐up. Data were collected 7, 28 and 60 days after travel. However, data were only presented for 7 days after return

Other bias

High risk

Funding: GlaxoSmithKline

It was not made clear whether the interpretation of the study findings was independent of the study sponsor

Pearlman 1980

Methods

Design: RCT

Study dates: unclear, during 1977

Malaria transmission pattern and local antimalarial drug resistance: "subjects were resident in an area highly endemic for P. vivax and chloroquine resistant P. falciparum"

Adverse event monitoring: "a physician visited the study area each week and conducted a sick call for participating and nonparticipating villagers...Between physician visits, residents were taken to a nearby health centre for serious medical problems"

Participants

Number enrolled: 990

Inclusion criteria: "All eligible and consenting villagers over 10 years of age were included in the study"

Exclusion criteria: "Female villagers of childbearing age (15‐44 years) were not considered for inclusion"

Country of recruitment: The Bhu Phram Valley, Thailand

Country of malaria exposure: The Bhu Phram Valley, Thailand

Duration of exposure to malaria: study duration 26 weeks

Type of participants: Thai residents, semi‐immune

Interventions

1. Mefloquine (1 x 180 mg tablet, children 22 to 35 kg ½ dose) weekly

2. Mefloquine (1 x 360 mg tablet, children 22 to 35 kg ¼ dose) weekly

3. Mefloquine (1 x 360 mg tablet, children 22 to 35 kg ¼ dose) every 2 weeks

4. Placebo (1 x tablet) weekly

Co‐interventions: "Those who had experienced falciparum parasitemias were given a therapeutic dose of sulfadoxine (1,500 mg)‐pyrimethamine (75 mg), and those with vivax or malariae parasitemias were treated with the standard regimen of chloroquine (1,500 mg over a 3‐day period), followed by primaquine, 15 mg daily for 14 days, for those study subjects known to be G‐6‐PD normal"

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Episodes of parasitaemia

3. Adverse events; any

Outcomes assessed not included in the review:

4. Laboratory tests; haematocrit, white cell count, white cell differential, serum glutamic oxaloacetic transaminase, alkaline phosphatase and blood urea nitrogen

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Assignment to one of six treatment groups was made on a stratified random number basis"

Comment: no details of how random numbers were generated

Allocation concealment (selection bias)

Unclear risk

"In the course of this visit, the technician opened a sealed, numbered envelope, gave the enclosed tablets, and observed the subject swallow them"

Comment: no mention of the envelope being opaque

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"Each subject received two tablets each week (medication, placebo or a combination) in order to maintain the double blind nature of the study"

"All tablets were identical in appearance"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but not clear how this was achieved

Incomplete outcome data (attrition bias); efficacy

Unclear risk

"Nine hundred and ninety nine subjects began the 25‐week field trial and 856 completed it (86.5%). 160/189 (85%) of the mefloquine 180 mg weekly group, 169/191 (88%) of the mefloquine 360 mg weekly, 158/184 (86%) of the mefloquine 360 mg fortnightly and 36/44 (82%) of the placebo group completed the trial"

Comment: reasons for losses to follow‐up were not reported

Incomplete outcome data (attrition bias); safety

Low risk

"There was no clinical evidence of drug toxicity in the 990 study participants, nor were there significant changes in the biochemical parameters"

Selective reporting (reporting bias); efficacy

Low risk

"Table 2 shows the number of subjects in each group who completed the study, the number infected with P. falciparum, and the number of episodes of asexual parasitemia"

Selective reporting (reporting bias); safety

High risk

"There was no clinical evidence of drug toxicity in the 990 study participants"

Comment: it was unclear whether all events that occurred during the 6 month trial period were included

Other bias

Unclear risk

Comment: study sponsor not reported

Petersen 2000

Methods

Design: retrospective cohort study

Study dates: 1 May 1996 to 30 April 1998

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 5446 questionnaires mailed, 4158 respondents

Inclusion criteria: "travellers 18 years old or older, who were not pregnant and had no previous adverse reactions to any of the prescribed drugs"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "the standard recommendations to Danish travelers were followed"

Country of recruitment: Denmark

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Chloroquine*

Not included in the review:

3. Chloroquine + proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; any

2. Serious adverse outcomes

3. Adverse effects; visual impairment (blurred vision), nausea, vomiting, abdominal pain, diarrhoea, dizziness, depression

4. Adverse effects; other (loss of appetite, strange thoughts, tingling, altered spatial perception, mouth ulcers)

Outcomes assessed not included in the review:

5. Discontinuation of study drug due to adverse effects (data reported on aggregate)

6. Measure of adherence to the drug regimen (data reported on aggregate)

7. Duration in days of symptoms

Notes

Funding sources: Not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

The questionnaire collected information regarding age, body weight and gender, destination and duration of travel but these were not reported

2. Selection of participants into the study: serious

Response rate 4158/5446 (76.3%)

3. Measurement of interventions: low

The prescription was provided by a travel clinic which also performed the study, and switches and discontinuations have been recorded and reported

4. Departures from intended interventions: moderate

Discontinuations were reported. Although changes in prophylaxis were mentioned, it was unclear whether participants were analysed according to original or subsequent prophylactic grouping

5. Missing data: low

4020/4158 (97%) of participants are included in the analysis for adverse events

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded. It was unclear whether the questionnaire implied causality to the drug regimen

7. Selection of the reported results: moderate

The questionnaire included demographic information, but this was not reported. All results were reported according to short‐term or long‐term users of prophylaxis, which was not specified in the methods section

8. Other: no information

No information is provided regarding the study sponsor

Philips 1996

Methods

Design: cross‐sectional cohort study

Study dates: November 1993 to October 1994

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient questionnaire sent 2 weeks after travellers return

Participants

Number enrolled: 741 respondents, 918 questionnaires sent

Inclusion criteria: "...travelers were asked to participate in the study when they attended TMVC clinics in Adelaide or Melbourne for pretravel consultation. If either doxycycline or mefloquine malaria chemoprophylaxis was recommended for part, or whole, of their itinerary, permission was sought to have them receive a mailed questionnaire"

Exclusion criteria: "...under 18 years old, if doxycycline was recommended at doses other than 100mg daily, if other antimalarials were to be used during the intended journey, or if a traveller was not returning home in under 6 months"

Factors influencing drug allocation: "Unless a contraindication existed for one or the other drug, the choice of which one to take was left to the traveler, the physician having already discussed, at some length, the different regimens, cost, and commonly reported adverse effects"

Country of recruitment: Australia

Region of malaria exposure: various (Southeast Asia, Africa, South Asia (India), Pacific)

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

1. Mefloquine*

2. Doxycycline*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; any, nausea/vomiting, abdominal pain, diarrhoea, headache, dizziness, abnormal dreams, insomnia, anxiety

2. Serious adverse events

3. Adverse events; other (mood change, palpitations, itching, rash, red skin, vaginal itch)

4. Adverse effects; any

5. Adverse effects; abdominal pain, diarrhoea

6. Discontinuation of study drug due to adverse effects

7. Measure of adherence to the drug regimen

Outcomes assessed not included in the review

8. Reasons for choice of antimalarial drug regimen

Notes

Funding sources: "Thanks to Roche and Pfizer pharmaceutical companies for their financial support"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Identified confounders were measured and reported across groups. Mefloquine users were more likely to be female and had longer duration of treatment

2. Selection of participants into the study: serious

Response rate 668 of 918 (73%)

3. Measurement of interventions: low

The prescription was provided by a travel clinic which also performed the study; discontinuations were recorded and reported

4. Departures from intended interventions: moderate

Discontinuations were recorded. It was unclear whether information regarding switches was recorded

5. Missing data: low

All information was collected at one time point and all participants were included in the analysis

6. Measurement of outcomes: serious

Comment: The outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: serious

Information was reported for all adverse events recorded, but participants' assessment of causality to the study drug was only reported for two side effects

8. Other: serious

"Sponsored by Roche and Pfizer pharmaceuticals"

The role of the study sponsor was not made clear

Potasman 2002

Methods

Design: RCT

Study dates: unclear

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: "Two days after drug ingestion, a second EEG was performed, and a blood sample for mefloquine level was obtained...Travelers were given forms on which to record adverse effects that appeared within 48 hours after drug intake"

Participants

Number enrolled: 90

Inclusion criteria: not explicitly mentioned, included travellers from the Bnia Zion medical centre, Haifa, Israel

Exclusion criteria: "Travelers younger than 18 years; with a history of epilepsy or depression, known allergy to mefloquine, cardiac conduction block; using beta‐blockers; or who were pregnant...Travelers with an abnormal baseline EEG (unifocal or repetitive bursts)"

Country of recruitment: Israel

Country of malaria exposure: not applicable

Duration of follow up: 48 hours

Type of participants: non‐travellers

Interventions

1. Mefloquine (1 x Mephaquine 250 mg tablet, Mepha, Aesch, Switzerland) one dose

2. Mefloquine (1 x Larium 250 mg tablet, Roche, Basel, Switzerland) one dose

3. Placebo

Outcomes

1. Adverse events; any

2. Adverse events; other (neuropsychiatric, abnormal EEG 48 hours after ingestion)

Notes

Funding sources: "Partially funded by Mepha Ltd, Aesch, Switzerland"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Eligible travelers were randomly assigned to one of three groups" "Randomization and statistical tests were carried out using Statmate and InStat"

Allocation concealment (selection bias)

Unclear risk

Comment: not mentioned

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

"Participants were unaware of their group assignment until they completed their tests"

Comment: methods used to blind participants not reported

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"EEG pairs (pre‐ and post‐mefloquine) were examined separately by two senior neurologists who were unaware of group allocation"

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Unclear risk

Comment: data were provided for all participants who were not excluded on the basis of abnormal baseline EEG

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

Unclear risk

"Adverse effects, mainly gastrointestinal and neuropsychiatric were noted in 26 travellers"

Comment: specific nature of each adverse effect is not noted per group

Other bias

High risk

Partially funded by Mepha Ltd, Aesch, Switzerland.

Comment: the role of the study sponsor was not clear

Rack 2005

Methods

Design: retrospective cohort study

Study dates: July 2003 to June 2004

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 794

Inclusion criteria: Travellers who were visiting five popular tropical regions or countries.

Exclusion criteria: aged < 18 years, travelling for more than 2 months, and major acute or chronic diseases

Country of recruitment: Germany

Country of malaria exposure: Kenya/Tanzania, Senegal/Gambia, India/Nepal, Thailand, Brazil

Duration of exposure to malaria: various, mean duration of travel 23.9 days

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Doxycycline*

3. Atovaquone‐proguanil*

4. Chloroquine*

Not included in the review:

5. Chloroquine‐proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Narrative description of adverse effects

Outcomes assessed not included in the review:

2. Risk behaviours during travel

3. Illness during travel

4. Seeking medical care owing to illness or accident

5. Accidents during travel

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Demographic information was provided for the entire cohort, not by prophylactic regimen

2. Selection of participants into the study: moderate

Numbers of participants choosing not to participate in the study were not reported

3. Measurement of interventions: serious

Participants were asked to self‐report which prophylaxis they took after return. The time after return was not specified

4. Departures from intended interventions: no information

There was insufficient information provided to determine whether the questionnaire contained information regarding discontinuations or switches

5. Missing data: moderate

Follow up was obtained for 658 (83%) travellers

6. Measurement of outcomes: serious

There was insufficient information on the questionnaire about how adverse effects were sought and if outcome measures were objective. There was no mention of blinding of outcome assessors

7. Selection of the reported results: moderate

There was insufficient information provided regarding the questionnaire to determine if all questions were reported. Side effects were grouped to report symptoms

8. Other: no information

No information was provided regarding the study sponsor

Rieckmann 1993

Methods

Design: cohort study

Study dates: 1989

Malaria transmission pattern and local antimalarial drug resistance: higher levels of P falciparum than P vivax locally. Local chloroquine and primaquine resistance

Adverse event monitoring: unclear

Participants

Number enrolled: 349

Inclusion criteria: Unclear

Exclusion criteria: Unclear

Country of recruitment: Australia

Country of malaria exposure: Papua New Guinea

Duration of exposure to malaria: 3 to 13 week training exercises

Type of participants: Soldiers

Interventions

Included in the review:

1. Mefloquine (1 x 250 mg weekly)

2. Doxycycline (1 x 100 mg tablet, daily, starting one day before deployment and continuing until 3 days after return)

Not included in the review:

3. Doxycycline + primaquine

4. Doxycycline + chloroquine

Outcomes

Included in the review:

1. Narrative description of adverse effects

Outcomes assessed not included in the review::

2. Clinical cases of malaria

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

No demographic information was provided

2. Selection of participants into the study: moderate

Numbers of participants choosing not to participate in the study not reported

3. Measurement of interventions: low

All participants were soldiers who were issued with medication

4. Departures from intended interventions: moderate

No information was provided regarding discontinuations or switches

5. Missing data: moderate

No losses to follow‐up or treatment withdrawals were reported, but the paper does not clearly state that none occurred

6. Measurement of outcomes: serious

There was insufficient information on how adverse effects were sought and if outcome measures were objective. There was no mention of blinding outcome assessors

7. Selection of the reported results: moderate

There was insufficient information provided regarding the questionnaire to determine if all questions were reported. Side effects were grouped to report symptoms.

8. Other: no information

No information is provided regarding the study sponsor

Rietz 2002

Methods

Design: cross‐sectional cohort study

Study dates: June to December 2000

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 491

Inclusion criteria: "visitors over fifteen who were travelling to South or Central America, Africa, India or South‐East Asia, including China, and who were not suffering from any chronic illness"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "After talking to the doctor, the doctor wrote whether malaria prophylaxis had been decided on and if so which kind"

Country of recruitment: Sweden

Region of malaria exposure: various, including South or Central America, Africa, India or Southeast Asia, including China

Duration of exposure to malaria: "most were abroad between two to four weeks"

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Chloroquine*

3. Non‐users

Not included in the review:

4. Chloroquine‐proguanil*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; any, seriously negative effect on the journey

2. Adverse effects; any

3. Adverse effects; other (neuropsychiatric, skin problems)

Outcomes assessed not included in the review:

4. Importance attached to prophylaxis

5. Whether travellers had any anxiety about side effects prior to taking prophylaxis

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex, destination and duration of travel data were collected but not reported across groups. BMI was not measured

2. Selection of participants into the study: serious

Response rate 62%

3. Measurement of interventions: low

The prescription was provided by a travel clinic which also performed the study

4. Departures from intended interventions: moderate

Discontinuations were reported, but not across groups. Switches were not recorded

5. Missing data: low

All participants who completed both questionnaires were included in the analysis

6. Measurement of outcomes: moderate

The outcome measure was subjective; participants and personnel were not blinded. Participants were asked to report all symptoms, and which they felt were due to prophylaxis

7. Selection of the reported results: moderate

Symptoms were grouped to report outcomes

8. Other: low

Source of funding not mentioned. "competing interests: none declared"

Salako 1992

Methods

Design: RCT

Study dates: July 1987 to June 1988

Malaria transmission pattern and local antimalarial drug resistance: "holoendemic for malaria... at the time of the trial, chloroquine resistance was not a problem"

Adverse event monitoring: "study participants were seen weekly up to week 28". Interview with study personnel for events such as "fever, chills, malaise, nausea and vomiting, rashes and other symptoms and signs that could be regarded as adverse events"

Participants

Number enrolled: 567

Inclusion criteria: "...adult males aged 16 to 60 years, judged healthy on clinical grounds (no history of any illness and physical examination revealed no evidence of an acute or chronic illness). The patients were not on any drugs"

Exclusion criteria: "...known hypersensitivity to sulphonamides, antimalarial drug treatment in the preceeding four weeks, presence of chronic debilitating disease and inability to attend regularly for follow up"

Country of recruitment: Nigeria

Country of malaria exposure: Nigeria

Duration of exposure to malaria: study duration 24 weeks

Type of participants: Nigerian residents, semi‐immune.

Interventions

1. Mefloquine (1 x 250 mg tablet, Hoffman‐La Roche) weekly for 4 weeks followed by 1 x 125 mg tablet weekly for 20 weeks, total duration 24 weeks*

2. Chloroquine (1 x 300 mg base tablet, Hoffman‐La Roche) weekly, total duration 24 weeks*

3. Placebo, 1 tablet (Hoffman‐La Roche) weekly, total duration 24 weeks*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Episodes of parasitaemia

3. Adverse events; any, abdominal pain, diarrhoea, headache, dizziness, pruritis, visual impairment (blurred sight)

4. Serious adverse events

5. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

6. Laboratory tests; white blood cell counts, haematocrit, serum glutamic oxaloacetic transaminase and serum glutamic‐pyruvic transaminase

7. Adverse events: rash, muscle stiffness (occurred in < 1% of study participants)

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"...subjects were allocated randomly into five groups on the basis of a pre‐determined randomisation list"

Comment: no mention of how the list was generated

Allocation concealment (selection bias)

Unclear risk

"...blister packs containing a total of 24 tablets were provided for each subject ...The packs and tablets were identical in appearance and were labelled with the appropriate double‐blind number"

Comment: no mention of opaque sealed envelopes or central allocation

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"The packs and tablets were identical in appearance"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no description provided of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Low risk

Comment: numbers lost to follow up were provided across groups, with reasons provided. 107/113 (95%) mefloquine recipients, 103/115 (90%) chloroquine recipients and 101/114 (89%) placebo recipients completed the trial

Incomplete outcome data (attrition bias); safety

Low risk

Comment: reports "number of individuals suffering adverse events during the trial". Numbers lost to follow up were provided across groups, with reasons provided. 107/113 (95%) mefloquine recipients, 103/115 (90%) chloroquine recipients and 101/114 (89%) placebo recipients completed the trial

Selective reporting (reporting bias); efficacy

Low risk

Comment: clinical cases of malaria and episodes of parasitaemia are reported for all participants

Selective reporting (reporting bias); safety

Unclear risk

"No change of clinical relevance occurred in any of the groups in the above laboratory tests"

Comment: there was insufficient information available regarding the collection of adverse events to determine whether the reported list included all events or only a targeted list. Data not fully reported for blood tests

Other bias

Unclear risk

Comment: study sponsor not mentioned, but four of the authors are attributed to F Hoffman‐La Roche

Santos 1993

Methods

Design: RCT

Study dates: August 1982 to January 1983

Malaria transmission pattern and local antimalarial drug resistance: region considered hyperendemic. P falciparum resistant to chloroquine and “high prevalence of multiresistant Plasmodium falciparum transmission”

Adverse event monitoring: during the initial screening visit, weekly visits, and a final visit at study end, participants were asked about illnesses, mainly about signs and symptoms compatible with malaria, and blood tests were done, including haematocrit and leucocyte count

Participants

Number enrolled: 122

Inclusion criteria: "volunteer soldiers and civilians aggregated to the 5th Battalion of Engineering and Construction in a community in Porto Velho"

Exclusion criteria: aged < 12 years and > 55 years, pregnancy, people with debilitating disease, people who took antimalarial drugs in the previous four weeks and people with allergy to sulphonamides

Country of recruitment: Brazil

Country of malaria exposure: Brazil

Duration of exposure to malaria: Mean duration within study (across groups) 16.9 weeks

Type of participants: Brazilian soldiers and civilians, semi‐immune

Interventions

Included in review comparisons:

1. Mefloquine (2 x 250 mg tablets, Roche) every 4 weeks*

2. Mefloquine (1 x 250 mg tablet, Roche) every 2 weeks*

3. Placebo

Not included in review comparisons:

4. Fansidar*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Adverse effects; any, anxiety

Outcomes assessed not included in the review:

3. Laboratory tests; haematocrit, white blood cell counts, serum glutamic oxaloacetic transaminase and serum glutamic‐pyruvic transaminase

Notes

Funding sources: Laboratory Roche provided mefloquine and “support” for conducting the study. Comando do 5o Batalhão de Engenharia e Construção, Porto Velho, RO, provided laboratory and field installations

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: described as a randomized controlled trial, but no details were given on the sequence generation

Allocation concealment (selection bias)

Unclear risk

Comment: no description of allocation concealment provided

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"Each week... participants ingested 4 tablets of equal appearance, contained in sealed envelopes"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"Each week... participants ingested 4 tablets of equal appearance, contained in sealed envelopes, with a code pre‐determined for each individual and not opened after the completion of the study"

Comment: no mention of blinding of outcome assessors

Incomplete outcome data (attrition bias); efficacy

High risk

"120 participants were initially recruited (30 in each group). Six of them were then excluded and were not included in the analysis. 8 participants left the area of study (one after the 10th week and 7 after the 11th week of exposure)"

Outcomes were included in the analysis, and were substituted by eight new participants. With these six excluded participants and eight substituted participants, final sample size was 122.

Comment: participants were not followed up beyond the active phase of treatment for relapses

Incomplete outcome data (attrition bias); safety

Unclear risk

Comment: reasons for losses to follow‐up were not reported

Selective reporting (reporting bias); efficacy

Low risk

Comment: all cases of malaria were reported

Selective reporting (reporting bias); safety

Unclear risk

Comment: there was insufficient information provided regarding the method of adverse effects monitoring to determine whether all outcomes had been reported

Other bias

High risk

Roche provided mefloquine and “support” for conducting the study

Saunders 2015

Methods

Design: retrospective cohort study

Study dates: January to June 2007

Malaria transmission pattern and local antimalarial drug resistance: "malaria risk and transmission patterns have been known to shift rapidly in Afghanistan"

Adverse event monitoring: "A retrospective, anonymous survey was completed by soldiers returning to Fort Drum, NY from Afghanistan"

Participants

Number enrolled: 2601 surveys distributed, 2351 (90%) returned

Inclusion criteria: none mentioned

Exclusion criteria: none mentioned

Factors influencing drug allocation: "oral mefloquine 250 mg per week was the primary alternative to doxycycline... In some cases, mefloquine was chosen as the first‐line therapy based on either perceived advantages in compliance, unit force protection, and/or operational concerns"

Country of recruitment: USA

Country of malaria exposure: Afghanistan

Duration of exposure to malaria: various, not specified

Type of participants: military

Interventions

Included in review comparisons:

1. Mefloquine*

2. Doxycycline*

Not included in review comparisons:

3. Atovaquone‐proguanil* (data on adverse events not collected; data on compliance not reported)

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any, vomiting, diarrhoea

2. Adverse effects; other (heartburn/dyspepsia)

3. Discontinuations of study drug due to adverse effects

4. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

5. Clinical cases of malaria

6. Adverse effects: numbers not reported in both groups (nausea, headache, dizziness, abnormal dreams, insomnia, depression, photosensitivity, rash, loss of appetite, pain and/or difficulty swallowing, vaginitis, lightheadedness, nervousness, ringing in ears, chills)

7. Use of personal protective measures to prevent mosquito bites

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Information was provided on duration of deployment, area of deployment, sex, age group and rank across regimens. Area deployed in Afghanistan and sex were different across groups. No adjustment for confounders was made in the analysis

2. Selection of participants into the study: low

Response rate 2351/2601 surveys (90%)

3. Measurement of interventions: moderate

Participants were asked to self‐report which prophylaxis was used on return to the USA. It is unclear if participants were still receiving the intervention at this time

4. Departures from intended interventions: serious

"There were 520 respondents (25.2%) reporting more than one medication used to prevent malaria over the course of the deployment"

5. Missing data: low

Analysis included 1898/2011 (94.4%) respondents for doxycycline, 564/596 (94.6%) respondents for mefloquine

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded. Different criteria were used to assess adverse effects related to mefloquine and doxycycline

7. Selection of the reported results: serious

There was insufficient information provided regarding the questionnaire to determine whether all included outcomes were reported. Data for doxycycline were provided by severity gradings but not for mefloquine

8. Other: no information

No information is provided regarding the study sponsor

Schlagenhauf 1997

Methods

Design: cross‐over RCT

Study dates: 1993 to 1994

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: "Throughout dosing, the participants were monitored and questioned regarding their general well‐being. The participants were seen 1) prior to taking any medication, 2) at the end of the first week (during which the loading dose was administered, 3) one week before testing, and 4) on the testing day itself when they were asked to report any changes from normal and questioned with regard to any symptoms experienced while taking the drug"

Participants

Number enrolled: 23

Inclusion criteria: "conducted with trainee pilots attending the Swiss Civil Aviation School during the classroom phases of their study"

Exclusion criteria: "history of a seizure disorder; psychosis or severe depression; known allergy or sensitivity to mefloquine or related compounds; concurrent use of cardioactive medication; compromised renal or hepatic function; pregnancy or the intention to become pregnant within three months of mefloquine use; use of mefloquine in the preceding two months, and use of hypnotics or tranquillizers during the two weeks prior to testing and alcohol within 12 hr of testing"

Country of recruitment: Switzerland

Country of malaria exposure: not applicable

Duration of follow up: 4 weeks

Type of participants: Swissair trainee pilots, did not travel

Interventions

1. Mefloquine (1 x 250 mg tablet) given daily on 3 consecutive days followed from day 8 by once a week administration of 1 tablet for three consecutive weeks

2. Placebo (1 tablet) given daily on 3 consecutive days followed from day 8 by once a week administration of one tablet for 3 consecutive weeks

Outcomes

Included in the review:

1. Adverse events; any

2. Discontinuations of study drug due to adverse effects

3. Adverse events; other outcomes (instrument co‐ordination analyser, sleep assessment, sway, neurobehavioural evaluation system, profile of mood states)

Notes

Funding sources: This study was sponsored by the F. Hoffmann La Roche Tropical Medicine Unit (Basel, Switzerland)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

Comment: method of randomization not reported

Allocation concealment (selection bias)

Unclear risk

Comment: no details of allocation concealment reported

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

Comment: described as double blind but no mention of whether placebo was identical to the active formulation

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no description of who was blinded and how

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Unclear risk

"There was one withdrawal due to dizziness, diarrhea, and flu‐like symptoms and three volunteers spontaneously reported minor sleep‐related AEs (adverse events), including insomnia, unpleasant dreams, superficial sleep, and early awakening. These events all occurred in the mefloquine loading dose phase"

Comment: not clear whether this withdrawal was included in the data analysis

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

High risk

"The individual Environmental Symptom Questionnaire (ESQ) symptoms were also analyzed and items selected for their relevance to mefloquine administration were assessed by Cochran's Q test for related samples"

Comment: intra‐individual changes in scores were obtained during the study, but outcomes were presented as means across groups. Data from the ESQ were not reported, only "no significant differences". Data for the Profile of Mood States questionnaire was presented in a graph with no standard deviations

Other bias

High risk

This study was sponsored by the F. Hoffmann La Roche Tropical Medicine Unit (Basel, Switzerland). The role of the study sponsor was not clear

Schlagenhauf 2003

Methods

Design: RCT

Study dates: 1998 to 2001

Malaria transmission pattern and local drug resistance: not mentioned

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 674

Inclusion criteria: adult travellers aged 18 to 70 years, with planned travel of 1 to 3 weeks to a malaria‐endemic area, and consulting at a travel clinic ≥ 17 days before departure

Exclusion criteria: glucose‐6‐phosphate dehydrogenase deficiency, history of severe adverse events with any of the four study drugs or a contra‐indication for their use, pregnancy or unwillingness to adhere to reliable contraception, history of seizures, psychiatric disorders, severely impaired renal or hepatic function, concurrent or recent vaginal infections or bacterial enteric disorders, a history of photosensitivity, or unwillingness to adhere to the study protocol

Countries of recruitment: Switzerland, Germany and Israel

Region of malaria exposure: sub‐Saharan Africa

Duration of exposure to malaria: 1 to 3 weeks

Type of participants: travellers

Interventions

1. Mefloquine (1 capsule containing mefloquine hydrochloride 274.09 mg, equivalent to mefloquine 250 mg base) once weekly, starting 17 days before travel and continuing for 4 weeks after travel*

2. Chloroquine‐proguanil (1 combined capsule containing chloroquine diphosphatase 161.21 mg, equivalent to chloroquine 100 mg base; and 200 mg proguanil hydrochloride) once daily, starting 17 days before travel and continuing for 4 weeks after travel*

3. Doxycycline (1 capsule containing doxycycline monohydrate 100 mg) once daily, starting 17 days before travel and continuing for 4 weeks after travel*

4. Atovaquone‐proguanil (1 combined capsule containing 250 mg atovaquone and 100 mg proguanil hydrochloride) once daily, starting 17 days before travel and continuing for 1 week after travel*

*matched placebo for each treatment arm

Outcomes

Included in the review:

1. Adverse events; any

2. Serious adverse events

3. Adverse events; other ('gastrointestinal', 'skin symptoms', 'neuropsychological') ‐ any severity, mild, moderate, severe

4. Discontinuation of study drug due to adverse effects

5. Adverse events; other outcomes (profile of mood states, quality of life score)

Notes

Funding sources: GlaxoSmithKline supplied atovaquone‐proguanil and gave financial support; Zeneca supplied chloroquine‐proguanil; Pfizer supplied doxycycline; Roche supplied mefloquine and gave financial support

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Randomisation was from a computer generated table of numbers in permuted blocks of five"

Allocation concealment (selection bias)

Unclear risk

"Participants were allocated treatment sequentially in order of study numbers. Allocation concealment was by sealed envelope"

Comment: not reported whether envelopes were opaque

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"The drugs were provided as identical capsule blister packs in weekly cards"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: Described as double blind but no mention of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

High risk

Comment: Method of detection for malaria, frequency and duration of follow up were not reported

Incomplete outcome data (attrition bias); safety

Unclear risk

"Adverse events were analysed in 623 participants who completed questionnaires at recruitment and at least one of the follow up periods"

"Data was collected during recruitment and at follow up 13‐11 days before departure, 6‐4 days before departure and 7‐14 days after departure"

Comment: it was unclear how many participants provided data at each time point

Selective reporting (reporting bias); efficacy

Low risk

"No cases of malaria were reported for any study arm"

Selective reporting (reporting bias); safety

High risk

"Adverse events were analysed in 623 participants who completed questionnaires at recruitment and at least one of the follow up periods"

"Data was collected during recruitment and at follow up 13‐11 days before departure, 6‐4 days before departure and 7‐14 days after departure"

Comment: Data were presented on aggregate across multiple time points

Other bias

High risk

Funding: Pfizer, GlaxoSmithKline, Roche, and Zeneca provided the drugs free of charge. GlaxoSmith Kline and Roche provided research grants.

"Competing interests: PS has received speakers’ honorariums and travel expenses from Roche and GlaxoSmithKline. She acted as a consultant to Roche in a drug safety database evaluation. RS has received speakers’ honorariums and travel expenses from GlaxoSmithKline, Roche, and Pfizer. He is also a member of the advisory board of GlaxoSmithKline for malaria prophylaxis related questions. BB has received a speaker’s honorarium and travel expenses from GlaxoSmithKline. HN has received speakers’ honorariums and travel expenses from GlaxoSmithKline on different occasions. He has been principal or coinvestigator in several vaccine trials sponsored by GlaxoSmithKline"

Schneider 2013

Methods

Design: retrospective cohort study

Study dates: 1 January 2001 and 1 October 2009

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: Incident cases of a neuropsychiatric disorder including anxiety, stress‐related disorders or psychosis, depression, epilepsy or peripheral neuropathies during or after anti‐malarial drug use within the UK general practice research database

Participants

Number enrolled: Not available

Inclusion criteria: "We identified in the general practice research database all patients who had ≥ 1 prescription of mefloquine, chloroquine and/or proguanil or atovaquone/proguanil between January 1, 2001 and October 1, 2009, and who had a pre‐travel consultation within 1 week of the prescription"

Exclusion criteria: "We only included subjects who used anti‐malarial drugs for malaria prophylaxis... Furthermore, individuals had at least 12 months of information on prescribed drugs and medical diagnoses before the first prescription date for a study drug. In addition, subjects had recorded activity (diagnoses or drug prescriptions) at any time after the prescription for an anti‐malarial drug to include only subjects who returned to the UK. We excluded all patients with a diagnosis of malaria prior to the start of anti‐malarial drug use, patients with a history of cancer, alcoholism, rheumatoid arthritis; or with an outcome of interest prior to using anti‐malarial drugs. The date of the first neuropsychiatric disorder was the index date for each case"

Country of recruitment: UK

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine*

2. Atovaquone‐proguanil*

Not included in review comparisons:

3. Chloroquine‐proguanil*

4. Unexposed (case‐control design)

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; psychiatric disorders (anxiety, depression, psychosis)

2. Adverse events; other ('anxiety or stress related disorders or psychosis', epilepsy, neuropathy, phobia, panic attack)

Notes

Funding sources: F. Hoffmann‐La Roche Ltd., Basel, Switzerland

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex and BMI were measured but only reported for people experiencing adverse events

2. Selection of participants into the study: moderate

"We excluded all patients with a personal history of recorded neuropsychiatric disorders from the study population, but family history is not consistently recorded in the database"

3. Measurement of interventions: moderate

"We only included subjects who used anti‐malarial drugs for malaria prophylaxis. We identified prescriptions for which the GP recorded ‐ within a week of the anti‐malarial drug prescription ‐ specific codes indicating that the person received the prescription for malaria prophylaxis, such as 'travel advice' or “prophylactic drug use”

4. Departures from intended interventions: serious

It is possible that participants discontinued or switched medication and this would not have been captured in the study

5. Missing data: moderate

The study did not report the total number of participants, only those who experienced adverse events

6. Measurement of outcomes: moderate

General practitioners diagnosing patients would have been aware of their exposure status

7. Selection of the reported results: moderate

Data for anxiety, stress‐related disorders and psychosis were reported on aggregate

8. Other: serious

Study was sponsored by Roche. The role of the funding source was not made clear

Schwartz 1999

Methods

Design: cross‐sectional cohort study

Study dates: October 1995 to April 1998

Malaria transmission pattern and local antimalarial drug resistance: "both P. falciparum and P. vivax are hyperendemic"

Adverse event monitoring: "...we directly contacted all travelers for complete follow‐up and assessment of compliance. Fifty travelers taking primaquine completed a questionnaire regarding side effects"

Participants

Number enrolled: 158

Inclusion criteria: Israelis participating in rafting trips in Southern Ethiopia

Exclusion criteria: none mentioned

Country of recruitment: Israel

Country of malaria exposure: Ethiopia

Duration of exposure to malaria: 14 to 20 days

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine (1 x 250 mg tablet) weekly, Starting 1 week prior to departure, during travel and for 4 weeks after return

2. Doxycycline (1 x 100 mg tablet) daily

Not included in review comparisons:

3. Primaquine 15 mg daily for travellers with body weight < 70 kg and 30 mg for those weighing > 70 kg, starting 1 day prior to departure and continuing for up to 2 days after departure

4. Hydroxychloroquine*

*dosing regimen not specified

Outcomes

Included in the review:

1. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

2. Clinical cases of malaria

3. Measure of adherence to the drug regimen (not fully reported)

4. Adverse effects; any (methods of detection different for primaquine versus other regimens)

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex and BMI were not reported for any participants. Destination and duration of travel was roughly equivalent across all groups

2. Selection of participants into the study: moderate

Subjects were selected on the basis of their travel destination. Start of follow up and start of intervention coincide. No non‐responses were reported

3. Measurement of interventions: moderate

"Prior to the trip, participants consulted one of a number of travel clinics in Israel, among them our clinic"

Comment: it was unclear how intervention status was ascertained for participants who visited other clinics

4. Departures from intended interventions: low

Two discontinuations (158 participants) were reported

5. Missing data: serious

"In addition, we directly contacted all travelers for complete follow‐up and assessment of compliance. Fifty travelers taking primaquine completed a questionnaire regarding side effects"

It was unclear how information on discontinuations and side effects were obtained for participants who did not take primaquine"

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: serious

"In addition, we directly contacted all travelers for complete follow‐up and assessment of compliance. Fifty travelers taking primaquine completed a questionnaire regarding side effects"

It was unclear how information on discontinuations and side effects was obtained for participants who did not take primaquine

8. Other: no information

No information was provided regarding the study sponsor

Shamiss 1996

Methods

Design: cross‐sectional cohort study

Study dates: not mentioned

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 45

Inclusion criteria: none mentioned

Exclusion criteria: none mentioned

Factors influencing drug allocation: "Prior knowledge about the side effect profile of mefloquine forced us to prescribe doxycycline 100 mg daily for aviators and mefloquine 250 mg weekly for non‐aviator crew"

Country of recruitment: Israel

Country of malaria exposure: Rwanda and Zaire

Duration of exposure to malaria: "biweekly flights to and from Rwanda to Zaire with an average of 4 hours stay in the field over a period of 2 months"

Type of participants: military

Interventions

1. Mefloquine (1 x 250 mg tablet) weekly, starting on the day of travel (< 12 hours before the first flight) and continuing until 4 weeks after return

2. Doxycycline (1 x 100 mg tablet) daily, starting on the day of travel (< 12 hours before the first flight) and continuing until 4 weeks after return

Outcomes

Included in the review:

1. Adverse effects; any, nausea, abdominal pain, dizziness

2. Adverse effects; other (fatigue)

3. Discontinuations of study drug due to adverse effects

4. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

5. Clinical cases of malaria

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex and BMI were not measured. Destination and duration of travel were set by the study design

2. Selection of participants into the study: low

"Prior knowledge about the side effects profile of mefloquine forced us to prescribe doxycycline 100 mg daily for aviators and mefloquine 250 mg weekly for non‐aviator aircrew up to 1 mo after the last return"

All participants completed questionnaires.

3. Measurement of interventions: low

Type of prophylaxis used was set by the job of the included participants

4. Departures from intended interventions: low

"Two non‐aviators were dropped from the study because of receiving the wrong prescription"

5. Missing data: low

"Two non‐aviators were dropped from the study because of receiving the wrong prescription"

Information was provided for the remaining 43 participants.

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

"...the questionnaire included questions about compliance, side effects attributed to chemoprophylaxis, and any illness after return"

No information was provided regarding illness after return.

8. Other: no information

No information is provided regarding the study sponsor

Sharafeldin 2010

Methods

Design: retrospective cohort study

Study dates: July 2006 to December 2008

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: "Participants… were sent an informative email asking them to complete a web‐based questionnaire"

Participants

Number enrolled: 242 students sent questionnaire, 180 respondents

Inclusion criteria: "all medical students who had performed an elective abroad between July 2006 and December 2008, who had visited countries where hepatitis A is endemic, and who had notified the student registrar to obtain study credits"

Exclusion criteria: none mentioned

Factors influencing drug allocation: "...students are free to visit [our occupational health department] or any other travel clinic including the LUMC in‐hospital travel clinic or their general practitioner"

Country of recruitment: Netherlands

Country of malaria exposure: none mentioned

Duration of exposure to malaria: mean duration of stay = 74 days (range 10 to 224 days )

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine*

2. Atovaquone‐proguanil*

3. Doxycycline*

Not included in review comparisons:

4. Primaquine*

5. Proguanil*

6. Chloroquine* (no data reported)

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any

2. Serious adverse outcomes

3. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

4. Clinical cases of malaria

5. Risk of infection with bloodborne viruses

6. Health risks while abroad

7. Health problems experienced whilst abroad

8. Health problems experienced on return

Notes

Funding sources: There was no dedicated funding for this project

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex, destination and duration of travel were measured but information not provided across groups. BMI was not measured

2. Selection of participants into the study: serious

Response rate 180/242 (74.4%)

3. Measurement of interventions: serious

"...six students did not remember which prophylaxis had been prescribed"

Students were asked to self‐report which prophylaxis they took an average of 235 days after completing their trip

4. Departures from intended interventions: moderate

"Eight students who used mefloquine (20%) stopped the drug prematurely as did ten students on atovaquone‐proguanil (16%) and the student on doxycycline. Only two of these students switched to another prophylaxis"

5. Missing data: low

"none of the questionnaires was incomplete"

All participants were included in the analysis

6. Measurement of outcomes: serious

The outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

Insufficient information was provided on how data on adverse effects were sought

8. Other: low

"There was no dedicated funding for this project"

Sonmez 2005

Methods

Design: prospective cohort study

Study dates: April 2002 to October 2003

Malaria transmission pattern and local antimalarial drug resistance: "20% of recent cases were due to P. falciparum' 'chloroquine resistant P. falciparum"

Adverse event monitoring: "common questionnaires were used to investigate the compliance to and side effects of both regimes"

Participants

Number enrolled: 1400 soldiers worked in the region

Inclusion criteria: "...all Turkish soldiers were examined in detail and serum samples were taken before heading for the region"

Exclusion criteria: "...none of the participants had any chronic disease"

Factors influencing drug allocation: "The preference of the preventive regime was related to the availability of the drugs... the prophylaxis was started with doxycycline, which was at hand in March 2002. Then again the soldiers who came after July 2002 were given mefloquine"

Country of recruitment: Afghanistan

Country of malaria exposure: Afghanistan

Duration of exposure to malaria: "The average time of presence for a single soldier in Kabul region was approx. 6 month [sic]"

Type of participants: military

Interventions

1. Mefloquine*

2. Doxycycline*

*dosing regimen not specified

Outcomes

Included in the review:

1. Serious adverse effects

2. Adverse effects; any, nausea, vomiting, abdominal pain, diarrhoea, headache, insomnia, dyspepsia, anorexia

Outcomes assessed not included in the review:

3. Clinical cases of malaria

Notes

Funding sources: Not mentioned

Communications with study author:

Sonmez 2005 no longer had access to the original study data. However, the study authors confirmed that for table 1: "The comparisons of the number of side effects of both regimes" the number of side effects for specific symptoms e.g. nausea was equivalent to the number of soldiers reporting that side effect. In addition, the authors were able to clarify a discrepancy in the original text: the paper states "27 mefloquine takers (41.2%) reported 43 side effects at the 2nd week of prophylaxis". The total number of mefloquine participants was 228; 41.2% equates to 94 participants. The authors confirmed that the correct figure was 27 mefloquine users (11%).

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age of participants was balanced across groups. Destination and duration of travel were set by the study design. Sex and BMI were not reported

2. Selection of participants into the study: serious

734 soldiers returned questionnaires (52.2%)

3. Measurement of interventions: low

All soldiers were issued with prophylaxis

4. Departures from intended interventions: low

Switches between prophylactic regimens were not possible

5. Missing data: low

The data were collected at 2 time points. The reported denominator for each time point was the same

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

There was insufficient information provided to be sure that all outcomes included in the questionnaire were reported

8. Other: no information

No information was provided regarding the study sponsor

Sossouhounto 1995

Methods

Design: RCT

Study dates: January 1989 to June 1989

Malaria transmission pattern and local antimalarial drug resistance: "region endemic for P. falciparum malaria"

Adverse event monitoring: "participants had access to a village health center, where they could notify personnel of any malaise or side effects. Clinical examinations and parasitologic tests were performed every 4 weeks. Blood counts were carried out at the end of weeks 4, 19 and 24"

Participants

Number enrolled: 500

Inclusion criteria: "five‐hundred male volunteers, aged 16‐60 years, who were residents of a local village, were randomly assigned"

Exclusion criteria: none mentioned

Country of recruitment: Adzope region, Ivory Coast

Country of malaria exposure: Adzope region, Ivory Coast

Duration of exposure to malaria: study duration 20 weeks

Type of participants: Ivory Coast residents, semi‐immune

Interventions

Included in review comparisons:

1. Mefloquine (1 x 250 mg tablet) weekly in weeks 1 to 4, (1 x 125 mg tablet) weekly in weeks 5 to 20

2. Chloroquine (1 x 300 mg tablet) weekly for 20 weeks

3. Placebo (1 tablet) weekly for 20 weeks

Not included in review comparisons:

4. Fansidar

5. Fansifem

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Episodes of parasitaemia

3. Serious adverse events

4. Adverse events: any, diarrhoea, headache, pruritis

Outcomes assessed not included in the review:

5. Laboratory tests; haematocrit and white blood cell count

6. Adverse events: other (leukopenia, malaise; did not occur in any study participants)

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Five‐hundred male volunteers… were randomised"

Comment: Method of randomization was not described

Allocation concealment (selection bias)

Unclear risk

Comment: no description of allocation concealment was provided

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"double blind". "The medications and placebo were identical in appearance"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no information was provided on how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Low risk

"Four hundred and ninety‐nine subjects were evaluated for safety (at least one tablet taken and one visit) as well as for efficacy"

Comment: 499/500 (99.8%) participants included in the analysis

Incomplete outcome data (attrition bias); safety

Low risk

"Four hundred and ninety‐nine subjects were evaluated for safety (at least one tablet taken and one visit) as well as for efficacy"

Comment: 499/500 (99.8%) participants included in the analysis

Selective reporting (reporting bias); efficacy

Low risk

Comment: all outcomes prespecified in the methods section were reported

Selective reporting (reporting bias); safety

Unclear risk

"Blood counts were carried out at the end of weeks 4, 19 and 24"

Comment: blood counts were reported only for one participant who developed reversible leukopenia

Other bias

Unclear risk

Comment: no information provided regarding the study sponsor

Steffen 1993

Methods

Design: cohort study

Study dates: Malpro 1‐ April 1985 to July 1988, Malpro 2‐ July 1988 to December 1991

Malaria transmission pattern and local antimalarial drug resistance: various, not stated

Adverse event monitoring: self‐completed questionnaires were distributed and collected by cabin crews to all passengers returning on charter planes

Participants

Number enrolled: 145,003

Inclusion criteria: not explicitly stated. This trial includes two publications, Steffen 1993 states "All passengers returning on charter planes from Mombasa, Kenya, to Europe", whereas Steffen 1990 states "all passengers flying back to Europe from East Africa (Kenya) or West Africa (9 countries)". Data have been included from Steffen 1993

Exclusion criteria: "All travellers who stayed longer than one year in tropical Africa were excluded, as were those who did not spend the main part of their visit in East Africa (Kenya, Tanzania and Uganda)"

Country of recruitment: not applicable

Region of malaria exposure: East Africa (Kenya, Tanzania, Uganda)

Duration of exposure to malaria: various, not stated

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine*

2. Chloroquine (1 x 300 mg tablet) weekly

Not included in review comparisons:

3. Chloroquine (1 x 600 mg tablet) weekly

4. Proguanil*

5. Chloroquine + proguanil*

6. Pyrimethamine + sulfadoxine*

7. Non‐users (this population was asked about side effects (adverse effects) and instead answered regarding adverse events

*dosing regimen not specified

Outcomes

Included in the review:

1. Serious adverse effects

2. Adverse effects; any (mild, moderate or severe), visual impairment, nausea, headache, dizziness, insomnia, depression, pruritis

3. Adverse effects; other ('other skin', medical consultations due to side effects, incapacitation due to side effects, 'cutaneous', 'redness of the skin', consulted a doctor)

4. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

5. Clinical cases of malaria

6. Measures taken against mosquito bites

7. Sources of pre‐travel health information

8. Places visited in tropical Africa

Notes

Funding sources: "This study was sponsored by F. Hoffman‐La Roche Ltd, Basel, Switzerland"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex and BMI were not reported across different prophylactic groups

2. Selection of participants into the study: moderate

"In Malpro 1, 80.1% of all passengers completed the in‐flight questionnaire… in Malpro 2 the response rate [was] 83.9%"

3. Measurement of interventions: low

Passengers were asked to self‐report which malaria prophylaxis was used. Data were collected on the journey home, meaning it was likely that passengers were still taking this medication

4. Departures from intended interventions: low

Handschin 1997: "2.9% of passengers changed the prophylactic regimen during the observation period"

5. Missing data: moderate

Malpro 1 losses to follow‐up 4.1%, Malpro 2 losses to follow‐up 14.1%

6. Measurement of outcomes: moderate

The outcome measure was subjective; participants and personnel were not blinded. Serious adverse events were verified independently

7. Selection of the reported results: serious

Data on non‐serious side effects were not included from Malpro 1‐ 31% of participants (44,667) were not included

8. Other: serious

The study was funded by Roche. The role of the study sponsor was not made clear

Steketee 1996

Methods

Design: quasi‐RCT

Study dates: September 1987 to June 1990

Malaria transmission pattern and local antimalarial drug resistance: "primarily P falciparum (> 90%), some P malariae and minimal P ovale... High levels of Plasmodium falciparum resistance to CQ... sensitivity of P. falciparum to mefloquine was documented"

Adverse event monitoring: "At the time of each dose, a questionnaire was administered to record symptoms including fever and reported drug side effects since the last visit"

Participants

Number enrolled: 4220

Inclusion criteria: "...consecutive attenders at first antenatal clinic visit were enrolled at three sites… At a fourth side, consecutive attenders in their first and second pregnancy were enrolled"

Exclusion criteria: "At this site [fourth site, government district hospital] women with two or more pregnancies were not enrolled because of the large number of patients attending the clinic and the limited number of study staff"

Country of recruitment: Malawi

Country of malaria exposure: Malawi

Duration of exposure to malaria: Ongoing in semi‐immune population ‐ monitored from enrolment for various periods of time

Type of participants: pregnant Malawian residents, semi‐immune

Interventions

1. Mefloquine (1 x 250 mg tablet) weekly, with a single loading dose of 750 mg

2. Chloroquine (1 x 300 mg tablet) weekly, with a loading dose 25 mg of base/kg given as a divided dose over 2 days

3. Chloroquine (1 x 300 mg tablet) weekly

Outcomes

Included in the review:

1. Episodes of parasitaemia

2. Adverse events; any

3. Serious adverse events

4. Discontinuations of study drug due to adverse effects

5. Adverse pregnancy outcomes; still births, abortions

Outcomes assessed not included in the review:

6. Frequency of placental malarial infection

7. Frequency of prematurity or intra‐uterine growth retardation

8. Frequency of maternal febrile illness or anaemia

9. Likelihood of infant acquisition of malarial infection

Notes

Funding sources: "This work was supported and made possible by the Africa Bureau, Office of Operations and New Initiatives and the Office of Analysis, Research and Technical Support, the USAID through the Africa Child Survival initiative… The Global Program on AIDS, World Health Organisation provided support for the HIV testing and evaluation portion of this study"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

"Systematic assignment of regimens was done based on the clinic and day of enrolment… All women making their first antenatal clinic on a given day were assigned to the same regimen; the following day, enrolled women were assigned to the following regimen"

Allocation concealment (selection bias)

High risk

"Systematic assignment of regimens was done based on the clinic and day of enrolment… All women making their first antenatal clinic on a given day were assigned to the same regimen; the following day, enrolled women were assigned to the following regimen"

Blinding of participants and personnel (performance bias)
Adverse effects/events

High risk

Comment: no mention of participants being blinded to which prophylactic regimen they were taking

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

"All blood smear examinations were done with the microscopist blinded to the study subject’s antimalarial regimen"

Comment: No mention of outcome assessors being blinded to the treatment regimen used when assessing safety outcomes

Incomplete outcome data (attrition bias); efficacy

Unclear risk

"Among the 4187 enrolled women, 3380 (81%) [were analysed]… 94 did not have an initial blood smear result for comparison, 89 left the study area before follow up, 397 delivered before the follow up visit, 133 missed their appropriate follow up visit, and 94 did not have documented adherence to the drug regimen"

Comment: numbers lost to follow up were not reported across groups

Incomplete outcome data (attrition bias); safety

High risk

"A total of 4101 women had information available after their first dose and 2976 women had information available after their dose at four weeks"

Comment: reasons for missing data were not reported

Selective reporting (reporting bias); efficacy

Unclear risk

"Only P falciparum infections were of interest for this study… when P malariae alone was identified these infections were excluded from the analysis"

"For the purposes of malaria prevention and infant outcome we analysed the group of women… only if they were enrolled in the study for six or more weeks and had received the appropriate amount of medication during their participation"

"A total of 1,790 women delivered in study health facilities had received proper dosing on their antimalarial regimen, and had their peripheral blood examined"

Comment: women who had reported fever during pregnancy, and during the 2 weeks prior to delivery was reported, but not reported across antimalarial drug regimens

Selective reporting (reporting bias); safety

High risk

"All other complaints e.g. weakness, heart palpitations accounted for less than 15% of reported symptoms"

Comment: Data were collected weekly but only reported after the first and the fourth dose

Other bias

Low risk

"This work was supported and made possible by the Africa Bureau, Office of Operations and New Initiatives and the Office of Analysis, Research and Technical Support, the USAID through the Africa Child Survival initiative… The Global Program on AIDS, World Health Organisation provided support for the HIV testing and evaluation portion of this study"

Stoney 2016

Methods

Design: Prospective cohort study

Study dates: 2009 to 2011

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: "...participants were asked to complete a survey each week during travel and a post‐travel survey within 2–4 weeks after return"

Participants

Number enrolled: 628 participants completed all three surveys, 370 included in the analysis

Inclusion criteria: "Travelers were included from among all those enrolled if they received a prescription for chemoprophylaxis, traveled to at least one malaria‐endemic area, and completed pre‐ and post‐travel surveys and at least one during‐travel survey"

Exclusion criteria: "To complete the study in a reasonable amount of time, only participants with shorter durations of travel (approximately 2 months) were included"

Factors influencing drug allocation: "Several different medications are available for malaria chemoprophylaxis, depending on the traveler’s destination and medical history"

Country of recruitment: USA

Country of malaria exposure: India (13%), Tanzania (8%), Kenya (7%), South Africa (7%), and Haiti (7%)

Duration of exposure to malaria: median travel duration 13 days

Type of participants: travellers

Interventions

Included in the review:

1. Mefloquine*

2. Doxycycline*

3. Atovaquone‐proguanil*

4. Chloroquine*

Not included in the review:

5. Primaquine*

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse effects; any, headache, abnormal dreams 'intense nightmares', any gastrointestinal

2. Discontinuations of study drug due to adverse effects

3. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

4. Clinical cases of malaria

5. Reasons for non‐compliance with chemoprophylaxis (data provided on aggregate),

6. Use of personal protective measures for malaria prevention

Notes

Funding sources: "This work was supported by a cooperative agreement [1 U19CI000508‐01] between the Centers for Disease Control and Prevention and Boston Medical Center"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex, destination and duration of travel were recorded but figures were not reported across prophylactic regimens

2. Selection of participants into the study: moderate

No information was provided regarding travellers who did not wish to participate in the study

3. Measurement of interventions: low

"The type of chemoprophylaxis prescribed were collected from data entered by clinicians into patients’ medical records"

4. Departures from intended interventions: moderate

No switches or discontinuations were reported. It was unclear whether this information was captured in the questionnaire

5. Missing data: low

364/370 (98%) participants were included in the analysis

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective, participants and personnel were not blinded

7. Selection of the reported results: moderate

Insufficient information provided on how data on adverse effects were obtained to determine whether all outcomes had been reported

8. Other: low

Government funding

Tan 2017

Methods

Design: retrospective cohort study

Study dates: 18 July to 16 September 2016

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 8931

Inclusion criteria: Returned Peace Corps volunteers (RPCV) who served between 1995 and 2014 and had an e‐mail address in Peace Corps' RPCV database

Exclusion criteria: None mentioned

Factors influencing drug allocation: none specified

Country of recruitment: USA

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: returned Peace Corps volunteers

Interventions

1. Mefloquine*

2. Doxycycline*

3. Atovaquone‐proguanil*

4. Chloroquine*

*dosing regimen not specified

Outcomes

Included in the review:

1. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

2. "Questions about medications before, during, or after Peace Corps, as well as habits such as drinking"

Notes

Funding source: "this research did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Important confounders were measured but not been reported across groups. Duration and destination of travel were not measured

2. Selection of participants into the study: serious

8931/47,238 potential respondents included (13% response rate)

3. Measurement of interventions: serious

Participants were asked to self‐report which chemoprophylaxis they had taken at least 2 years after they had finished the course

4. Departures from intended interventions: serious

Limited information was provided regarding switches between interventions. Participants were asked to self‐report this information at least 2 years after finishing treatment

5. Missing data: low

Information on adherence was reported for all participants who answered this question (5026 respondents/5055 who reported taking malaria prophylaxis)

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

There was insufficient information provided to be sure that all outcomes included in the questionnaire were reported

8. Other: low

"This research did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors"

Terrell 2015

Methods

Design: cross‐sectional cohort study

Study dates: 2012 and 2013

Malaria transmission pattern and local antimalarial drug resistance: "...high risk of malaria (mainly P. falciparum) in Kenya, although the risk is assessed as very low in Nairobi and in the highlands above 2,500 m... widespread resistance to chloroquine"

Adverse event monitoring: "...questionnaire‐based, two‐arm cohort study"

Participants

Number enrolled: 2032 completed questionnaires available, 220 failed to indicate which drug they were taking

Inclusion criteria: all military personnel on deployment to Kenya who travelled on one of three main body flights on their return to the UK

Exclusion criteria: none mentioned

Factors influencing drug allocation: "...the choice of drugs considered in this study was limited to mefloquine or doxycycline... participants were free to use another drug should they experience unacceptable adverse effects or where there was an occupational reason"

Country of recruitment: UK

Country of malaria exposure: Kenya

Duration of exposure to malaria: "The majority of participants spent approximately 6 weeks in Kenya with a small number spending a few weeks longer if they filled an administrative role"

Type of participants: military

Interventions

Included in review comparisons:

1. Mefloquine*

2. Doxycycline*

Not included in review comparisons:

3. Atovaquone‐proguanil* (results not included in the analysis)

*dosing regimen not specified

Outcomes

Included in the review :

1. Adverse effects; any

2. Measure of adherence to the drug regimen

Outcomes assessed not included in the review:

3. Clinical cases of malaria

4. Impact of adverse effects on self‐reported ability to work

Notes

Funding sources: "The research was not sponsored by any external body"

After we submitted the review for peer referee, the author sent us a spreadsheet containing numbers of events relating to a variety of symptoms after the review had been submitted for publication. These data are not included in the review and will require some clarification over how they were collected to allow us to assess risk of bias. This additional information will be considered in future updates.

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

"Although not formally recorded, each unit can be assumed to be composed of similar populations in terms of number, age, gender, occupation, and general health"

2. Selection of participants into the study: serious

"Completion rates were consistently poor throughout the study period with only 150 to 250 questionnaires returned per tranche of around 1,000 troops"

3. Measurement of interventions: low

Participants were asked to self‐report which medication they were on while still taking the medication"

4. Departures from intended interventions: moderate

"...[participants] were invited to complete the questionnaire for whichever drug they took for the longer period"

5. Missing data: moderate

"2,032 completed questionnaires available for analysis of which 10.8% (220) failed to indicate which drug they were taking"

6. Measurement of outcomes: serious

The outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: serious

"In both arms, some participants indicated that they had experienced an adverse effect but did not report how it had impacted upon their ability to work. They were excluded from the final analysis"

Mefloquine: 71 participants, doxycycline: 67 participants

8. Other: low

"The research was not sponsored by any external body"

Tuck 2016

Methods

Design: cohort study

Study dates: 15 to 22 February 2015

Malaria transmission pattern and local antimalarial drug resistance: not specified

Adverse event monitoring: patient self‐reported questionnaire

Participants

Number enrolled: 115 (337 eligible)

Inclusion criteria: all land‐based members of a UK military expedition to Sierra Leone

Exclusion criteria: none specified

Country of recruitment: Sierra Leone

Country of malaria exposure: Sierra Leone

Duration of exposure to malaria: not specified

Type of participants: military

Interventions

1. Mefloquine

2. Doxycycline

3. Atovaquone‐proguanil

Outcomes

Included in the review:

1. Adverse effects: any, nausea, abdominal pain, diarrhoea, dizziness, insomnia 'disturbed sleep', pruritis, indigestion, mouth ulcers, lethargy

2. Measure of adherence to the drug regime

Notes

Funding source: unfunded

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Age, sex and BMI were not measured. Demographic information not reported across groups

2. Selection of participants into the study: serious

151 (46.3%) returned survey forms

3. Measurement of interventions: low

Participants were asked to self‐report which medication they were taking while taking it

4. Departures from intended interventions: moderate

Switches between groups were recorded. 8/151 recipients had medications switched due to unacceptable adverse effects. It was unclear to which drug adverse effects were attributed.

5. Missing data: low

Data were reported for all survey respondents.

6. Measurement of outcomes: serious

The outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

There was insufficient information provided to be sure that all outcomes included in the questionnaire were reported

8. Other: low

"This audit was unfunded"

van Riemsdijk 1997

Methods

Design: prospective cohort study

Study dates: 24 February to 24 May 1994

Malaria transmission pattern and local antimalarial drug resistance: various, not stated

Adverse event monitoring: participant self‐reporting questionnaire

Participants

Number enrolled: 1791 eligible and willing to co‐operate, data obtained from 1501 participants.

Inclusion criteria: "...persons who visited the Travel Clinic in the period between 24 February and 24 May, 1994, and who had an anticipated date of return to the Netherlands before the end of the study period, and who had given informed consent"

Exclusion criteria: none stated

Country of recruitment: Rotterdam, Netherlands

Region of malaria exposure: various; Africa, South America, Asia or the Middle East

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine (1 x 250 mg tablet) weekly

2. Non‐users of antimalarials

Not included in review comparisons:

3. Proguanil (1 x 200 mg tablet) daily

Outcomes

Included in the review:

1. Adverse events; nausea, diarrhoea, dizziness, abnormal dreams, insomnia, anxiety, depression, visual impairment

2. Adverse events; other (agitation, confusion)

Outcomes assessed not included in the review:

3. Profile of mood states (only reported in comparison with proguanil)

Notes

Funding sources: Not stated

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Counfounding: low

Identified confounders were measured and balanced across groups

2. Selection of participants into the study: moderate

1501/1791 (86% response rate)

3. Measurement of interventions: moderate

Comment: the prescription was provided by a travel clinic which also performed the study but no information regarding switches and discontinuations were recorded or reported

4. Departures from intended interventions: moderate

No information was provided on discontinuations or switches

5. Missing data: moderate

1227/1449 (85%) participants were included in the analysis; chloroquine‐proguanil users were not included. The number of non‐users decreased from 392 to 340 without explanation

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

It was clear what was asked in the questionnaire. Information was sought on the severity of adverse events but this was not reported

8. Other: no information

No information was provided regarding the study sponsor

van Riemsdijk 2002

Methods

Design: RCT

Malaria transmission pattern and local drug resistance: not mentioned

Study dates: unclear

Adverse event monitoring: baseline evaluation prior to travel, and follow up date 7 days after the participant left the endemic area and two scheduled telephone conversations

Participants

Number enrolled: 140

Inclusion criteria: travellers aged ≥ 3 years and weighing ≥ 11 kg with planned travel ≤ 28 days to a malaria‐endemic area (Overbosch 2001)

Exclusion criteria: In the published report "We excluded those who had risk factors for concentration impairment (e.g. use of opioids, hypnotics, or tranquillizers or use of alcohol 4 hours before testing)"

Within Høgh 2000 (unclear if the same exclusion criteria were applied): poor general health; drug hypersensitivity (to atovaquone, chloroquine or proguanil); history of alcoholism, seizures, psychiatric disorders, severe neurological disorders, severe blood disorders; renal, hepatic or cardiac dysfunction; clinical malaria within previous 12 months; travel to malaria‐endemic area within previous 60 days; risk factors for concentration impairment (e.g. use of opioids, hypnotics, or tranquillizers; or use of alcohol 4 hours before testing)

Country of recruitment: Rotterdam Travel Clinic, Netherlands

Regions of malaria exposure: various malaria endemic destinations (66% in Africa, 13% South America, 24% other)

Mean duration of exposure to malaria: 19 days

Type of participants: travellers, non‐immune

Interventions

1. Mefloquine (1 x 250 mg tablet; or ¼, ½ or ¾ of a tablet, according to body weight) once weekly, starting 7 days before travel and continuing for 4 weeks after travel*

2. Atovaquone‐chloroguanil (1 combined tablet containing 250 mg atovaquone and 100 mg proguanil hydrochloride; or alternatively 1 to 3 combined children's tablets according to body weight, each tablet containing 62.5 mg atovaquone and 25 mg proguanil hydrochloride) once daily, starting 1 to 2 days before travel and continuing for 1 week after leaving the malaria‐endemic area*

*matched placebo for each treatment arm

Outcomes

1. Adverse events; other outcomes (profile of mood states, neurobehavioural evaluation system)

2. Measures of adherence to the drug regimen

3. Discontinuations of the study drug due to adverse effects

Notes

Funding source: Netherlands Inspectorate for Healthcare gave financial support

'independently performed in a sample of patients from one center that participated in the MAL30010 multicenter clinical trial'‐ Enrollment criteria and study conduct were described in a separate publication (Høgh 2000) which refers to a different study population (atovaquone‐proguanil versus chloroquine‐proguanil).

'This study was planned and performed independently from the trial by other researchers and without knowledge of its results.'

'Subjects were separately recruited and asked for consent during the initial screening visit of the trial.'

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"A computer‐generated code was used to randomly assign a treatment number to the three bottles of study drug for every individual. At all sites consecutively enrolled individuals who satisfied all entry criteria received the next treatment number" (Høgh 2000)

Allocation concealment (selection bias)

Low risk

"Treatment codes were provided to investigators in opaque sealed envelopes, to be opened only if knowledge of study drug assignment was required for management of a medical emergency" (Høgh 2000)

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

"To mask differences between the dosing regimes, placebo tablets were used... All placebo treatment regimens were identical to the aforementioned scheme for the active ingredient of mefloquine and atovaquone plus chloroguanide"

Comment: did not mention whether the placebo and intervention tablets were identical in appearance

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"The assessments were made by researchers who were unaware of the treatment allocation"

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

High risk

"We enrolled a total of 140 subjects in the cohort, 119 of whom completed the follow up"

Comment: Those who did not complete follow up were not included in the subsequent statistical analysis. The proportion of participants who did not complete the study due to adverse outcomes varied significantly between groups (67% mefloquine and 33% atovaquone plus chloroguanide)

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

Low risk

"Data were collected on concurrent medications, as well as subject’s use of coffee, alcohol and illicit drugs"

"stratification for sex and adjustment for potential confounders such as smoking and the use of coffee and tea did not affect the result"

Comment: these data were not presented

Other bias

Low risk

Funding: "For this study came from the Inspectorate for Health Care. Glaxo Wellcome kindly provided us with the treatment allocation codes after completion of the study. No financial support, however, was received from any pharmaceutical company"

Vuurman 1996

Methods

Design: RCT

Study dates: not mentioned

Malaria transmission pattern and local antimalarial drug resistance: not applicable

Adverse event monitoring: "After each driving test, subjects [described]... the presence and severity of adverse effects ‐ drowsiness, weakness, headache, fatigue, nervousness, nausea, dizziness and memory disturbance"

Participants

Number enrolled: 42

Inclusion criteria: "...[volunteers] were medically screened by routine blood chemistry and haematology tests, a physical examination including an 12‐lead ECG recording, and urine tests for pregnancy and drugs of abuse"

Exclusion criteria: "...clinically relevant abnormalities in any blood test; far‐field, binocular visual acuity that deviated by more than 0.65 dioptres from normal, corrected or uncorrected; known hypersensitivity to any drug; history of any serious gastrointestinal, hepatic, renal neurologic or psychiatric disorder; evidence of drug or alcohol abuse, excessive alcohol or nicotine use; blood donation or participation in a drug trial within the prior 2 months; and for premenopausal females, pregnancy, lactation or failure to exercise reliable birth control"

Country of recruitment: Netherlands

Country of malaria exposure: not applicable

Duration of follow up: 30 days

Type of participants: non‐exposed Dutch nationals

Interventions

1. Mefloquine (1 x 250 mg tablet) weekly, with loading dose of one tablet daily for 3 days in week 1

2. Placebo (1 tablet) weekly, with identical loading regimen of placebo tablets

Outcomes

1. Adverse events; any, nausea, diarrhoea, headache, dizziness

2. Adverse events; other (fatigue)

3. Discontinuations of study drug due to adverse effects

4. Adverse events; other outcome measures (critical flicker/fusion frequency, critical instability tracking test, standardized stabilimetry method of the International Society of Posturography, tests of driving performance)

Notes

Funding sources: "The study was sponsored by F. Hoffmann‐La Roche Ltd"

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"The study followed a randomised, 2‐arm, double‐blind, parallel group design"

Comment: method of sequence generation not described

Allocation concealment (selection bias)

Unclear risk

"The study followed a randomised, 2‐arm, double‐blind, parallel group design"

Comment: method of allocation concealment not described

Blinding of participants and personnel (performance bias)
Adverse effects/events

Low risk

"They received mefloquine 250 mg or placebo in identically appearing tablets"

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Comment: described as double blind but no description of how this was achieved for researchers and outcome assessors

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Low risk

Comment: dropouts were reported. 2/20 participants dropped out of the mefloquine group, one due to adverse effects related to the study drug

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

High risk

"...subjects used 10 cm visual‐analogue scales to describe their mood in three dimensions – 'Alertness', 'Contentedness', and 'Calmness'”

Comment: outcomes relating to these descriptions were not reported. The study reports "events occurring more than once" in each group

Other bias

High risk

"The study was sponsored by F. Hoffmann‐La Roche Ltd"

Waner 1999

Methods

Design: cross‐sectional cohort study

Study dates: April to May 1996

Malaria transmission pattern and local antimalarial drug resistance: "a high risk Malaria area... Chloroquine‐resistant P. falciparum malaria"

Adverse event monitoring: "In‐flight self administered questionnaires were distributed and completed by travelers on flights returning to Johannesburg International Airport"

Participants

Number enrolled: 4035 questionnaires distributed, 3051 returned

Inclusion criteria: All travelers boarding the only commercial airline serving this area during April and May 1996 were included in the survey

Exclusion criteria: None mentioned

Country of recruitment: South Africa

Country of malaria exposure: South Africa

Duration of exposure to malaria: various, not specified

Type of participants: travellers

Interventions

Included in review comparisons:

1. Mefloquine*

2. Doxycycline*

3. Chloroquine*

Not included in review comparisons:

4. Chloroquine‐proguanil*

5. Proguanil*

*dosing regimen not specified

Outcomes

Included in review comparisons:

1. Adverse effects; any

Outcomes assessed not included in the review:

2. Sources of information on malaria prior to visit,

3. Use of personal protective measures against mosquitoes,

4. Measures of adherence to the drug regimen (information provided on aggregate),

5. Travellers knowledge of malaria symptoms

Notes

Funding sources: not mentioned

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Confounding: moderate

Sex of travellers was not provided by prophylactic regimen. Destination of travel was set by the study design. BMI of travellers and duration of travel were not recorded

2. Selection of participants into the study: serious

Response rate 3051/4035 (75%)

3. Measurement of interventions: low

Travellers were asked to self‐report which prophylactic regimen they were taking while still using the drug

4. Departures from intended interventions: moderate

No discontinuations or switches were reported. This information was not included in the questionnaire

5. Missing data: low

Outcome data were available for 973/978 mefloquine recipients and 80/80 doxycycline recipients

6. Measurement of outcomes: serious

Comment: the outcome measure was subjective; participants and personnel were not blinded

7. Selection of the reported results: moderate

Insufficient information provided on how data on adverse effects were obtained to determine whether all outcomes were reported

8. Other: no information

No information was provided regarding the study sponsor.

Weiss 1995

Methods

Design: RCT

Study dates: April to July 1993

Malaria transmission pattern and local antimalarial drug resistance: "Incidence of new cases of falciparum malaria during the rainy seasons has been measured at 90% in adults. P. falciparum accounts for > 95% of all malaria in Saradidi"

Adverse event monitoring: "Each subject was visited daily at home by an assigned field worker, who asked about symptoms of malaria or drug side effects, obtained malaria smears, or administered drug doses if the subject was not at school"

Participants

Number enrolled: 169

Inclusion criteria: aged 9 to 14 years. "Screening consisted of a physical examination, a urine pregnancy test for girls, and blood tests for complete blood cell count; blood urea nitrogen, serum alanine aminotransferase, and glucose‐6 phosphate dehydrogenase (G6PD) levels; and hemoglobin electrophoresis"

Exclusion criteria: none mentioned

Country of recruitment: Saradidi Rural Health Project, Nyanza province, Kenya on the shores of Lake Victoria

Country of malaria exposure: Saradidi Rural Health Project, Nyanza province, Kenya on the shores of Lake Victoria

Duration of exposure to malaria: study duration 4 months

Type of participants: Kenyan residents, semi‐immune

Interventions

1. Melfoquine (1 x 125 mg tablet) weekly, with a second dose given on the third day of the study, equal to their usual weekly medication.

2. Doxycycline (1 x 50 mg tablet) daily

3. Primaquine

4. Multivitamin (1 x tablet containing vitamin A, 2500 IU, thiamine, 1 mg, riboflavin, 0.5 mg, nicotinamide, 7.5 mg, ascorbic acid, 15 mg, vitamin 0 3, 250 IU) daily

Co‐interventions: After baseline malaria smears, all subjects received curative therapy for preexisting malaria: 7 days of quinine bisulfate, 300 mg three times daily, and doxycycline, 50 mg twice daily. The first dose of prophylactic drug was given starting the day after curative therapy finished

Outcomes

Included in the review:

1. Clinical cases of malaria

2. Episodes of parasitaemia

3. Discontinuations of study drug due to adverse effects

Outcomes assessed not included in the review:

4. Laboratory tests; complete blood cell counts, blood urea nitrogen and serum alanine aminotransferase

5. Mean number of symptoms reported per subject: nausea, abdominal pain, diarrhoea, headache, fever

Notes

Funding sources: Financial support: USA Naval Medical Research and Development Command (work unit no. 623002A.81 0.00 J0 I.HFX. J433). Kenya Medical Research Institute. USA Army Medical Research and Materiel Command Provisional (contract no. DAMDI7‐92‐V‐20J2)

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

"Students from each village school were separately randomized, to control for geographic variation in malaria transmission"

Comment: no description of how randomization was performed

Allocation concealment (selection bias)

Unclear risk

"All medications were in brown envelopes and were administered 7 days each week by I field worker at each school"

Comment: no mention of whether envelopes were sealed or if field workers had access to their content

Blinding of participants and personnel (performance bias)
Adverse effects/events

Unclear risk

Comment: no mention of whether participants were blinded

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

"None of the malaria slide readers knew which drugs the subjects were taking. None of the field workers visiting the homes daily to ask about symptoms or clinical staff evaluating and treating subjects at the Saradidi Clinic knew which drugs the subjects were taking. If there was concern about a drug side effect, the clinical staff would consult the medical monitor, who would break the code for that subject. This occurred only four times during the studies"

Incomplete outcome data (attrition bias); efficacy

Unclear risk

N/A

Incomplete outcome data (attrition bias); safety

Unclear risk

Comment: number included in the safety analysis not reported

Selective reporting (reporting bias); efficacy

Unclear risk

N/A

Selective reporting (reporting bias); safety

Unclear risk

Comment: mean number of symptoms reported per subject during 11 weeks of the study were reported. A targeted list of symptoms was reported, with everything else included in ‘all other’. It was unclear what this list included

Other bias

Low risk

Financial support: USA Naval Medical Research and Development Command (work unit no. 623002A.81 0.00 J0 I.HFX. J433). Kenya Medical Research Institute. USA Army Medical Research and Materiel Command Provisional (contract no. DAMDI7‐92‐V‐20J2)

Wells 2006

Methods

Design: retrospective cohort study

Study dates: January 2002 to December 31 2002

Malaria transmission pattern and local antimalarial drug resistance: various, not specified

Adverse event monitoring: "The study cohort was electronically linked to the Standardized Inpatient Data Record (SIDR) and the Health Care Service Record (HCSR) to identify hospitalization... We analyzed any‐cause hospitalization (excluding complications of pregnancy, childbirth, and the puerperium, congenital anomalies, and certain conditions originating in the perinatal period)"

Participants

Number enrolled: 397442

Inclusion criteria: "All active‐duty US service members during the period January 1, 2002, and December 31, 2002, as reported by the Defense Manpower Data Center (DMDC), Monterey, CA. The mefloquine prescribed group was defined as service members who had been prescribed a minimum of seven mefloquine tablets beginning in 2002 and who were identified as having been deployed at some point during the same time period. We used two reference groups. The first reference group was comprised of service members who had duty zip codes for either Europe or Japan at some time during 2002 and had no evidence of having been deployed from October 1, 2001 through the individual’s period of observation... The second reference group consisted of US service members who were identified as having been deployed for a minimum of 1 month during 2002"

Exclusion criteria: "Both reference groups were restricted to individuals who had no evidence of having received a prescription for mefloquine or chloroquine or a doxycycline prescription for more than 14 tablets.’ ‘Individuals who could not be followed a minimum of 2 months were excluded from the study"

Country of recruitment: USA

Country of malaria exposure: various, not specified

Duration of exposure to malaria: various, not specified

Type of participants: military

Interventions

1. Mefloquine*

2. Non‐users of antimalarials

*dosing regimen not specified

Outcomes

Included in the review:

1. Adverse events; serious (any hospitalization, hospitalizations due to vertiginous syndromes, migraine, dizziness and giddiness, anxiety disorders, somatoform disorders, mood disorders, PTSD, substance use disorders, personality disorders, nystagmus or adjustment reaction)

Outcomes assessed not included in the review:

2. Hospitalizations coded according to classification system: infectious/parasitic, neoplasms, endocrine, nutritional, metabolic, blood and blood‐forming organs, mental disorders, nervous system, circulatory system, respiratory system, digestive system, genitourinary system, skin and subcutaneous tissues, musculoskeletal and connective tissue, ill‐defined conditions, injury and poisoning

Notes

Funding sources: "This represents report 05–05, supported by the Department of Defense, under work unit no. 60002"

Risk of bias

Bias

Authors' judgement

Support for judgement

Other bias

Unclear risk

1. Counfounding: moderate

BMI, destination and duration of travel have not been recorded

2. Selection of participants into the study: serious

"Follow‐up time began on return from deployment for mefloquine‐prescribed members, and for the deployed reference group, on assignment to Europe or Japan, or January 1, 2002, whichever occurred last for the Europe/Japan reference group"

Start of follow up began a long time after start of intervention

3. Measurement of interventions: serious

Surrogate measure used for mefloquine exposure. There was a possiblity that some participants in the second deployed reference group took mefloquine

4. Departures from intended interventions: moderate

"Both reference groups were restricted to individuals who had no evidence of having received a prescription for mefloquine or chloroquine or a doxycycline prescription for more than 14 tablets"

5. Missing data: moderate

"Individuals who could not be followed a minimum of 2 months were excluded from the study"

Comment: number of participants in this group not reported

6. Measurement of outcomes: low

The outcome measure (hospitalizations) was objective

7. Selection of the reported results: low

All prespecified outcomes were reported

8. Other: low

Government funding

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Abraham 1999

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Adera 1995

Cohort study. R eported on efficacy but no other relevant outcomes

Adshead 2014

Single arm cohort study

Angelin 2014

No relevant outcomes reported

Anonymous 1991

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Anonymous 1998

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Anonymous 1998a

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Anonymous 2005

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Anonymous 2009

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Artaso 2004

Not a randomiz ed or cohort study e.g. case report or case control study

Arthur 1990a

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Banerjee 2001

No relevant outcomes reported

Barbero Gonzalez 2003

No relevant outcomes reported

Barrett 1996

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Berger 1998

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Berman 2004

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Bernado 1994

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Bijker 2014

This trial evaluated chemoprophylaxis plus sporozoite immunization

Bjorkman 1991

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Black 2007

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Blanke 2003

Cohort study. R eported on efficacy but no other relevant outcomes

Botella de Maglia 1999

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Bourgeade 1990

Not a randomiz ed or cohort study e.g. case report or case control study

Brenier‐Pinchart 2000

Not a randomiz ed or cohort study e.g. case report or case control study

Brisson 2012

No relevant outcomes reported

Bruguera 2007

Not a randomiz ed or cohort study e.g. case report or case control study

Burke 1993

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Caillon 1992

Not a randomiz ed or cohort study e.g. case report or case control study

Carme 1997

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Castot 1988

Not a randomiz ed or cohort study e.g. case report or case control study

Cave 2003

No relevant outcomes reported

Charles 2007

No relevant outcomes reported

Chin 2016

No relevant outcomes reported

Clifford 2009

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Clift 1996

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Clyde 1976

Single‐arm cohort study

Cobelens 1997

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Cohen 1997

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Conget 1993

Not a randomiz ed or cohort study e.g. case report or case control study

Conrad 1997

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Corbett 1996

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Coulaud 1986

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Croft 1996

Not a randomiz ed or cohort study e.g. case report or case control study

Croft 1997

RCT. C ompared mefloquine with a regimen that is no longer used routinely

Del Cacho 2001

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Dia 2010

No relevant outcomes reported

Durrheim 1999

Cohort study. Compare d mefloquine with a regimen that is no longer used routinely

Eamsila 1993

Cohort study. Compare d mefloquine with a regimen that is no longer used routinely

El Jaoudi 2010

Single arm cohort study

Fernando 2016

No relevant outcomes reported

Fujii 2007

Single arm cohort study

Hamer 2008

No relevant outcomes reported

Hellgren 1990

No relevant outcomes reported

Hopperus 1996

Single arm cohort study

Jaspers 1996

Single arm cohort study

Jensen 1998

Not a randomiz ed or cohort study e.g. case report or case control study

Karbwang 1991

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Karbwang 1991a

Mefloquine was used as a combination regimen with sulph adoxine and pyrimethamine

Khaliq 2001

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Kimura 2006

No relevant outcomes reported

Kitchener 2003

No relevant outcomes reported

Kitchener 2005

Cohort study. A llocation to study drug was based on the occurrence of adverse effects

Kok 1997

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Kollaritsch 2000

Single arm cohort study

Kozarsky 1993

Single arm cohort study

Landry 2006

Single arm cohort study

Lapierre 1983

Single arm cohort study

Lim 2005

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Lobel 1993

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely. C hloroquine users we re not clearly separated from users of chloroquine‐proguanil

Looareesuwan 1987

No relevant outcomes reported

MacArthur 2002

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Malvy 2006

Cohort study. R eported on efficacy but no other relevant outcomes

Marcy 1996

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Massey 2007

No relevant outcomes reported

Matsumura 2005

Single arm cohort study

Meszaros 1996

Not a randomiz ed or cohort study e.g. case report or case control study

Michel 2007

Cohort study. R eported on efficacy but no other relevant outcomes

Mimica 1983

No relevant outcomes reported

Mizuno 2006

Single arm cohort study

Mizuno 2010

Single arm cohort study

Moon 2011

No relevant outcomes reported

Morales de Naime 1989

No relevant outcomes reported

Munawar 2012

Single arm cohort study

Mølle 2000

Cohort selected on basis of adverse events

Namikawa 2008

No relevant outcomes reported

Nasveld 2010

RCT. C ompared mefloquine with a regimen which is not used routinely

Nevin 2010

No relevant outcomes reported

Nevin 2012

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Nosten 1990

RCT. Did not include a comparator; compared alternate mefloquine doses

Nosten 1999

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Nwokolo 2001

Cohort study. Compared mefloquine with a regimen that is no longer used routinely

Olanrewaju 2000

Single arm cohort study

Ollivier 2004

Single arm cohort study

Peetermans 2001

Cohort study. Compared mefloquine with a regimen that is no longer used routinely

Peragallo 1999

Cohort study. Compared mefloquine with a regimen that is no longer used routinely

Peragallo 2002

Single arm cohort study

Peragallo 2014

Single arm cohort study

Philips 1994

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Phillips 1996

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Phillips‐Howard 1998

Cohort study. Compared mefloquine with a regimen that is no longer used routinely

Pistone 2007

No relevant outcomes reported

Port 2011

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Potasman 2000

Cohort selected on basis of adverse events

Quinn 2016

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Reisinger 1989

RCT. C ompared mefloquine with a regimen that is no longer use d routinely

Rieckmann 1974

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Rieke 1993

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Ries 1993

Not a randomiz ed or cohort study e.g. case report or case control study

Ringqvist 2015

Cohort selected on basis of adverse events

Rombo 1993

RCT. C ompared mefloquine with a regimen that is no longer used routinely

Rønn 1998

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Sallent 1997

No relevant outcomes reported

Schlagenhauf 1996

Single arm cohort study

Scott 1993

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Smail 1991

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Smoak 1997

Single arm cohort study

Suriyamongkol 1991

Single arm cohort study

Tansley 2010

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

ter Kuile 1993

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Todd 1997

No relevant outcomes reported

Turner 2014

No relevant outcomes reported

Valerio 2005

No relevant outcomes reported

Van Genderen 2007

No participants received mefloquine prophylaxis

Van Grootheest 1999

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

van Riemsdijk 2004

Single arm cohort study

Venturini 2011

Single arm cohort study

Wagner 1986

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Wallace 1996

Field study in which troops switched extensively between mefloquine and doxycycline. Unable to attribute side effects to either prophylactic regimen

Weinke 1991

Cohort selected on basis of adverse events

White 2016

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Win 1985

Mefloquine not used at a prophylactic dose (e.g. treatment dose or i ntermittent preventive treatment of malaria in pregnancy dose)

Winstanley 1999

Not a research study of malaria prophylaxis e.g. letter to the editor or editorial

Wolters 1997

Cohort study. C ompared mefloquine with a regimen that is no longer used routinely

Data and analyses

Open in table viewer
Comparison 1. Mefloquine versus placebo/non users

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria Show forest plot

9

1908

Risk Ratio (M‐H, Random, 95% CI)

0.09 [0.04, 0.19]

Analysis 1.1

Comparison 1 Mefloquine versus placebo/non users, Outcome 1 Clinical cases of malaria.

Comparison 1 Mefloquine versus placebo/non users, Outcome 1 Clinical cases of malaria.

2 Malaria; episodes of parasitaemia in semi‐immune populations Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Mefloquine versus placebo/non users, Outcome 2 Malaria; episodes of parasitaemia in semi‐immune populations.

Comparison 1 Mefloquine versus placebo/non users, Outcome 2 Malaria; episodes of parasitaemia in semi‐immune populations.

2.1 Trials reporting number of participants with parasitaemia

3

414

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.06, 0.55]

2.2 Trials reporting number of episodes of parasitaemia

2

510

Risk Ratio (M‐H, Random, 95% CI)

0.05 [0.00, 5.25]

3 Serious adverse events or effects (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Mefloquine versus placebo/non users, Outcome 3 Serious adverse events or effects (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 3 Serious adverse events or effects (all studies).

3.1 RCTs (adverse events)

6

1221

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.14, 3.53]

3.2 Cohort studies (adverse effects)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

3.08 [0.39, 24.11]

4 Discontinuations due to adverse effects (all studies) Show forest plot

7

1130

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.55, 4.88]

Analysis 1.4

Comparison 1 Mefloquine versus placebo/non users, Outcome 4 Discontinuations due to adverse effects (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 4 Discontinuations due to adverse effects (all studies).

4.1 RCTs (adverse effects)

7

1130

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.55, 4.88]

5 Nausea (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Mefloquine versus placebo/non users, Outcome 5 Nausea (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 5 Nausea (all studies).

5.1 RCTs (adverse events)

2

244

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [1.05, 1.73]

5.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Fixed, 95% CI)

1.85 [1.42, 2.43]

6 Vomiting (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Mefloquine versus placebo/non users, Outcome 6 Vomiting (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 6 Vomiting (all studies).

6.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.50, 1.19]

6.2 Cohort studies (adverse events)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.45, 1.21]

7 Abdominal pain (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Mefloquine versus placebo/non users, Outcome 7 Abdominal pain (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 7 Abdominal pain (all studies).

7.1 RCTs (adverse events)

3

550

Risk Ratio (M‐H, Fixed, 95% CI)

1.09 [0.84, 1.40]

7.2 Cohort studies (adverse events)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.66, 1.42]

8 Diarrhoea (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Mefloquine versus placebo/non users, Outcome 8 Diarrhoea (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 8 Diarrhoea (all studies).

8.1 RCTs (adverse events)

4

589

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.32, 1.62]

8.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.93, 1.68]

9 Headache (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 Mefloquine versus placebo/non users, Outcome 9 Headache (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 9 Headache (all studies).

9.1 RCTs (adverse events)

5

791

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.71, 0.99]

9.2 Cohort studies (adverse events)

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.63, 4.26]

10 Dizziness (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 Mefloquine versus placebo/non users, Outcome 10 Dizziness (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 10 Dizziness (all studies).

10.1 RCTs (adverse events)

3

452

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.90, 1.17]

10.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [1.29, 2.49]

11 Abnormal dreams (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Mefloquine versus placebo/non users, Outcome 11 Abnormal dreams (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 11 Abnormal dreams (all studies).

11.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

2.35 [1.15, 4.80]

12 Insomnia (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.12

Comparison 1 Mefloquine versus placebo/non users, Outcome 12 Insomnia (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 12 Insomnia (all studies).

12.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.06, 2.02]

13 Anxiety (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.13

Comparison 1 Mefloquine versus placebo/non users, Outcome 13 Anxiety (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 13 Anxiety (all studies).

13.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.67, 2.21]

14 Depressed mood (all studies) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.14

Comparison 1 Mefloquine versus placebo/non users, Outcome 14 Depressed mood (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 14 Depressed mood (all studies).

14.1 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Random, 95% CI)

2.43 [0.65, 9.07]

15 Abnormal thoughts and perceptions Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.15

Comparison 1 Mefloquine versus placebo/non users, Outcome 15 Abnormal thoughts and perceptions.

Comparison 1 Mefloquine versus placebo/non users, Outcome 15 Abnormal thoughts and perceptions.

15.1 Cohort studies (adverse events)

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

5.77 [0.79, 42.06]

16 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.16

Comparison 1 Mefloquine versus placebo/non users, Outcome 16 Pruritis (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 16 Pruritis (all studies).

16.1 RCTs (adverse events)

3

609

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.60, 1.24]

16.2 Cohort studies (adverse events)

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

6.71 [1.58, 28.55]

17 Visual impairment (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.17

Comparison 1 Mefloquine versus placebo/non users, Outcome 17 Visual impairment (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 17 Visual impairment (all studies).

17.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.66, 1.46]

17.2 Cohort studies (adverse events)

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.27, 3.19]

18 Vertigo (all studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.18

Comparison 1 Mefloquine versus placebo/non users, Outcome 18 Vertigo (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 18 Vertigo (all studies).

18.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.78, 1.34]

19 Other adverse events (RCTs) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.19

Comparison 1 Mefloquine versus placebo/non users, Outcome 19 Other adverse events (RCTs).

Comparison 1 Mefloquine versus placebo/non users, Outcome 19 Other adverse events (RCTs).

19.1 Arthralgia

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.29 [0.02, 5.48]

19.2 Back pain

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.01, 1.61]

19.3 Blurred vision

1

208

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.01, 3.89]

19.4 Cough

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.71, 1.14]

19.5 Constipation

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.53, 1.11]

19.6 Decreased appetite

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.95, 1.28]

19.7 Falls

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.82, 1.43]

19.8 Fatigue

1

42

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.14, 5.86]

19.9 Gastritis

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.10, 10.98]

19.10 Myalgia

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.36, 6.57]

19.11 Rash

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.29 [0.04, 2.30]

19.12 Respiratory tract infection

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [1.04, 6.61]

19.13 Sore throat

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.04, 2.75]

19.14 Unsteadiness

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.74, 1.52]

19.15 Weakness

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.96, 1.17]

20 Other adverse effects (cohort studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 1.20

Comparison 1 Mefloquine versus placebo/non users, Outcome 20 Other adverse effects (cohort studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 20 Other adverse effects (cohort studies).

20.1 Agitation

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.61, 1.82]

20.2 Altered spatial perception

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

9.4 [0.57, 153.97]

20.3 Confusion

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.25, 1.78]

20.4 Loss of appetite

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.54, 1.50]

20.5 Mouth ulcers

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.39, 2.56]

20.6 Palpitations

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

8.06 [0.44, 147.68]

20.7 Tingling

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

1.92 [0.59, 6.24]

Open in table viewer
Comparison 2. Mefloquine versus doxycycline

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

4

744

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.35, 5.19]

Analysis 2.1

Comparison 2 Mefloquine versus doxycycline, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 2 Mefloquine versus doxycycline, Outcome 1 Clinical cases of malaria (RCTs).

2 Serious adverse events or effects (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Mefloquine versus doxycycline, Outcome 2 Serious adverse events or effects (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 2 Serious adverse events or effects (all studies).

2.1 RCTs (adverse events)

3

682

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.01, 8.16]

2.2 Cohort studies (adverse effects)

3

3722

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.23, 10.24]

3 Discontinuations due to adverse effects (all studies) Show forest plot

14

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.3

Comparison 2 Mefloquine versus doxycycline, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 3 Discontinuations due to adverse effects (all studies).

3.1 RCTs

4

763

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.41, 2.87]

3.2 Cohort studies

10

10165

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.54, 1.55]

4 Nausea (all studies) Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Mefloquine versus doxycycline, Outcome 4 Nausea (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 4 Nausea (all studies).

4.1 Cohort studies (adverse effects)

5

2683

Risk Ratio (M‐H, Fixed, 95% CI)

0.37 [0.30, 0.45]

4.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.71 [0.75, 9.74]

4.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [1.06, 2.43]

5 Vomiting (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Mefloquine versus doxycycline, Outcome 5 Vomiting (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 5 Vomiting (all studies).

5.1 Cohort studies (adverse effects)

4

5071

Risk Ratio (M‐H, Fixed, 95% CI)

0.18 [0.12, 0.27]

5.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

6 Abdominal pain (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 Mefloquine versus doxycycline, Outcome 6 Abdominal pain (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 6 Abdominal pain (all studies).

6.1 Cohort studies (adverse effects)

4

2569

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.09, 1.07]

6.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

1.65 [0.74, 3.70]

6.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

1.34 [0.83, 2.18]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.7

Comparison 2 Mefloquine versus doxycycline, Outcome 7 Diarrhoea (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 7 Diarrhoea (all studies).

7.1 Cohort studies (adverse effects)

5

5104

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.11, 0.73]

7.2 RCTs (adverse events)

2

376

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.78, 1.29]

7.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

3.58 [1.69, 7.59]

8 Dyspepsia (all studies) Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.8

Comparison 2 Mefloquine versus doxycycline, Outcome 8 Dyspepsia (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 8 Dyspepsia (all studies).

8.1 Cohort studies (adverse effects)

5

5104

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.09, 0.74]

9 Headache (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.9

Comparison 2 Mefloquine versus doxycycline, Outcome 9 Headache (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 9 Headache (all studies).

9.1 Cohort studies (adverse effects)

5

3322

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.50, 2.92]

9.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

2.31 [1.25, 4.27]

9.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

2.45 [1.38, 4.34]

10 Dizziness (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.10

Comparison 2 Mefloquine versus doxycycline, Outcome 10 Dizziness (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 10 Dizziness (all studies).

10.1 Cohort studies (adverse effects)

5

2633

Risk Ratio (M‐H, Random, 95% CI)

3.49 [0.88, 13.75]

10.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

3.05 [1.30, 7.16]

10.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

2.40 [1.47, 3.90]

10.4 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.62, 0.73]

11 Abnormal dreams (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.11

Comparison 2 Mefloquine versus doxycycline, Outcome 11 Abnormal dreams (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 11 Abnormal dreams (all studies).

11.1 Cohort studies (adverse effects)

4

2588

Risk Ratio (M‐H, Random, 95% CI)

10.49 [3.79, 29.10]

11.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.07, 15.89]

11.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

4.33 [2.08, 9.00]

12 Insomnia (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.12

Comparison 2 Mefloquine versus doxycycline, Outcome 12 Insomnia (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 12 Insomnia (all studies).

12.1 Cohort studies (adverse effects)

4

3212

Risk Ratio (M‐H, Random, 95% CI)

4.14 [1.19, 14.44]

12.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

2.03 [0.65, 6.40]

12.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

4.54 [2.09, 9.83]

12.4 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.43, 0.49]

13 Anxiety (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.13

Comparison 2 Mefloquine versus doxycycline, Outcome 13 Anxiety (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 13 Anxiety (all studies).

13.1 Cohort studies (adverse effects)

3

2559

Risk Ratio (M‐H, Fixed, 95% CI)

18.04 [9.32, 34.93]

13.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

8.74 [1.99, 38.40]

13.3 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.47, 0.56]

14 Depressed mood (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.14

Comparison 2 Mefloquine versus doxycycline, Outcome 14 Depressed mood (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 14 Depressed mood (all studies).

14.1 Cohort studies (adverse effects)

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

11.43 [5.21, 25.07]

14.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

6.27 [1.82, 21.62]

14.3 Retrospective healthcare record analysis (adverse events)

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

0.55 [0.51, 0.60]

15 Abnormal thoughts and perceptions Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.15

Comparison 2 Mefloquine versus doxycycline, Outcome 15 Abnormal thoughts and perceptions.

Comparison 2 Mefloquine versus doxycycline, Outcome 15 Abnormal thoughts and perceptions.

15.1 Cohort studies (adverse effects)

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

6.60 [0.92, 47.20]

15.2 Retrospective healthcare record analyses (adverse events)

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

0.41 [0.26, 0.66]

16 Pruritis (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.16

Comparison 2 Mefloquine versus doxycycline, Outcome 16 Pruritis (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 16 Pruritis (all studies).

16.1 Cohort studies (adverse effects)

2

1794

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.30, 0.91]

16.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

2.69 [0.93, 7.78]

17 Photosensitivity (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.17

Comparison 2 Mefloquine versus doxycycline, Outcome 17 Photosensitivity (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 17 Photosensitivity (all studies).

17.1 Cohort studies (adverse effects)

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

0.08 [0.05, 0.11]

17.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

0.03 [0.00, 0.49]

18 Yeast infection (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.18

Comparison 2 Mefloquine versus doxycycline, Outcome 18 Yeast infection (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 18 Yeast infection (all studies).

18.1 Cohort studies (adverse effects)

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.06, 0.16]

18.2 Cohort studies (adverse events)

1

354

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.06, 0.63]

19 Visual impairment (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.19

Comparison 2 Mefloquine versus doxycycline, Outcome 19 Visual impairment (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 19 Visual impairment (all studies).

19.1 Cohort studies (adverse effects)

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [1.41, 3.99]

20 Other adverse effects (cohort studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.20

Comparison 2 Mefloquine versus doxycycline, Outcome 20 Other adverse effects (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 20 Other adverse effects (cohort studies).

20.1 Alopecia

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

3.44 [1.96, 6.03]

20.2 Asthenia

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

1.83 [0.89, 3.76]

20.3 Balance disorder

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

2.87 [1.48, 5.59]

20.4 Decreased appetite

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

1.23 [0.42, 3.64]

20.5 Fatigue

2

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.23 [0.03, 1.77]

20.6 Hypoaesthesia

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

11.48 [3.01, 43.70]

20.7 Malaise

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.11, 0.71]

20.8 Mouth ulcers

1

33

Risk Ratio (M‐H, Fixed, 95% CI)

0.5 [0.02, 11.42]

20.9 Palpitations

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

2.76 [0.16, 48.91]

20.10 Tinnitus

1

684

Risk Ratio (M‐H, Fixed, 95% CI)

7.20 [0.39, 133.30]

21 Other adverse events (RCTs) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.21

Comparison 2 Mefloquine versus doxycycline, Outcome 21 Other adverse events (RCTs).

Comparison 2 Mefloquine versus doxycycline, Outcome 21 Other adverse events (RCTs).

21.1 Constipation

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

21.2 Cough

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.28, 1.01]

21.3 Decreased appetite

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [1.24, 10.20]

21.4 Malaise

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.88, 4.69]

21.5 Palpitations

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

21.6 Pyrexia

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.85 [1.09, 7.42]

21.7 Sexual dysfunction

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

3.05 [0.33, 28.51]

21.8 Somnolence

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

22 Other adverse events (cohort studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.22

Comparison 2 Mefloquine versus doxycycline, Outcome 22 Other adverse events (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 22 Other adverse events (cohort studies).

22.1 Adjustment disorder

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.40, 0.45]

22.2 Confusion

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

2.18 [0.24, 19.49]

22.3 Convulsions

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.45, 0.75]

22.4 Hallucinations

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.18 [0.08, 0.45]

22.5 Paranoia

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.10, 1.63]

22.6 Palpitations

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

13.44 [1.73, 104.38]

22.7 Panic attacks

1

21065

Risk Ratio (M‐H, Fixed, 95% CI)

4.16 [0.55, 31.49]

22.8 PTSD

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.53, 0.64]

22.9 Rash

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.50, 2.94]

22.10 Suicidal ideation

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.31, 0.47]

22.11 Suicide

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.32, 4.56]

22.12 Tinnitus

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.65 [0.61, 0.71]

23 Adherence (cohort studies) Show forest plot

14

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 2.23

Comparison 2 Mefloquine versus doxycycline, Outcome 23 Adherence (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 23 Adherence (cohort studies).

23.1 Adherence during travel

13

15583

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.12, 1.18]

23.2 Adherence in the post‐travel period

4

840

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.95, 1.22]

Open in table viewer
Comparison 3. Mefloquine versus atovaquone‐proguanil

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

2

1293

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

Analysis 3.1

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 1 Clinical cases of malaria (RCTs).

2 Serious adverse events or effects (all studies) Show forest plot

3

3591

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.08, 23.22]

Analysis 3.2

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 2 Serious adverse events or effects (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 2 Serious adverse events or effects (all studies).

2.1 Cohort studies

3

3591

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.08, 23.22]

3 Discontinuations due to adverse effects (all studies) Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 3 Discontinuations due to adverse effects (all studies).

3.1 RCTs

3

1438

Risk Ratio (M‐H, Random, 95% CI)

2.86 [1.53, 5.31]

3.2 Cohort studies

9

7785

Risk Ratio (M‐H, Random, 95% CI)

2.73 [1.83, 4.08]

4 Nausea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.4

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 4 Nausea (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 4 Nausea (all studies).

4.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

2.72 [1.52, 4.86]

4.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

2.50 [1.54, 4.06]

5 Vomiting (all studies) Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.5

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 5 Vomiting (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 5 Vomiting (all studies).

5.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.31 [0.49, 3.50]

5.2 Cohort studies (adverse effects)

3

2180

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.08, 4.09]

6 Abdominal pain (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.6

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 6 Abdominal pain (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 6 Abdominal pain (all studies).

6.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.52, 1.56]

6.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

0.64 [0.38, 1.07]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.7

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 7 Diarrhoea (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 7 Diarrhoea (all studies).

7.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.60, 1.47]

7.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.53, 1.35]

8 Mouth ulcers (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.8

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 8 Mouth ulcers (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 8 Mouth ulcers (all studies).

8.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [0.70, 3.00]

8.2 Cohort studies (adverse effects)

2

783

Risk Ratio (M‐H, Fixed, 95% CI)

0.12 [0.04, 0.37]

9 Headache (all studies) Show forest plot

9

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.9

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 9 Headache (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 9 Headache (all studies).

9.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.72 [0.99, 2.99]

9.2 Cohort studies (adverse effects)

8

4163

Risk Ratio (M‐H, Fixed, 95% CI)

3.42 [1.71, 6.82]

10 Dizziness (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.10

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 10 Dizziness (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 10 Dizziness (all studies).

10.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

3.99 [2.08, 7.64]

10.2 Cohort studies (adverse effects)

8

3986

Risk Ratio (M‐H, Fixed, 95% CI)

3.83 [2.23, 6.58]

10.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.23 [1.04, 1.46]

11 Abnormal dreams (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.11

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 11 Abnormal dreams (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 11 Abnormal dreams (all studies).

11.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [1.37, 3.04]

11.2 Cohort studies (adverse effects)

7

3848

Risk Ratio (M‐H, Random, 95% CI)

6.81 [1.65, 28.15]

12 Insomnia (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.12

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 12 Insomnia (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 12 Insomnia (all studies).

12.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

4.42 [2.56, 7.64]

12.2 Cohort studies (adverse effects)

8

3986

Risk Ratio (M‐H, Fixed, 95% CI)

7.29 [4.37, 12.16]

12.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [1.06, 1.44]

13 Anxiety (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.13

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 13 Anxiety (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 13 Anxiety (all studies).

13.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

6.12 [1.82, 20.66]

13.2 Cohort studies (adverse effects)

4

2664

Risk Ratio (M‐H, Fixed, 95% CI)

10.10 [3.48, 29.32]

13.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.54 [1.28, 1.85]

14 Depressed mood (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.14

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 14 Depressed mood (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 14 Depressed mood (all studies).

14.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

5.78 [1.71, 19.61]

14.2 Cohort studies (adverse effects)

6

3624

Risk Ratio (M‐H, Fixed, 95% CI)

8.02 [3.56, 18.07]

14.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.93 [1.56, 2.38]

15 Abnormal thoughts and perceptions (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.15

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 15 Abnormal thoughts and perceptions (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 15 Abnormal thoughts and perceptions (all studies).

15.1 Cohort studies (adverse effects)

3

2433

Risk Ratio (M‐H, Fixed, 95% CI)

1.50 [0.30, 7.42]

15.2 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

3.00 [0.69, 12.97]

16 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.16

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 16 Pruritis (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 16 Pruritis (all studies).

16.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.60, 2.70]

16.2 Cohort studies (adverse effects)

3

1824

Risk Ratio (M‐H, Fixed, 95% CI)

2.07 [0.40, 10.68]

17 Visual impairment (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.17

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 17 Visual impairment (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 17 Visual impairment (all studies).

17.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

2.04 [0.88, 4.73]

17.2 Cohort studies (adverse effects)

2

1956

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.29, 4.72]

18 Other adverse effects (cohort studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.18

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 18 Other adverse effects (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 18 Other adverse effects (cohort studies).

18.1 Allergic reaction

1

316

Risk Ratio (M‐H, Fixed, 95% CI)

0.79 [0.04, 14.48]

18.2 Alopecia

1

1469

Risk Ratio (M‐H, Fixed, 95% CI)

4.55 [0.30, 70.01]

18.3 Asthenia

2

1956

Risk Ratio (M‐H, Fixed, 95% CI)

1.84 [0.26, 13.12]

18.4 Balance disorder

1

1469

Risk Ratio (M‐H, Fixed, 95% CI)

2.86 [0.19, 44.19]

18.5 Cough

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.92]

18.6 Disturbance in attention

3

1363

Risk Ratio (M‐H, Fixed, 95% CI)

4.45 [1.84, 10.77]

18.7 Dyspepsia

2

362

Risk Ratio (M‐H, Fixed, 95% CI)

0.50 [0.17, 1.46]

18.8 Fatigue

2

618

Risk Ratio (M‐H, Fixed, 95% CI)

4.62 [0.47, 45.56]

18.9 Hypoaesthesia

2

1946

Risk Ratio (M‐H, Fixed, 95% CI)

4.45 [0.93, 21.26]

18.10 Loss of appetite

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.33, 1.43]

18.11 Muscle pain

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

7.57 [0.45, 127.80]

18.12 Palpitations

3

2180

Risk Ratio (M‐H, Fixed, 95% CI)

3.34 [0.73, 15.26]

18.13 Photosensitization

2

718

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.10, 4.92]

18.14 Pyrexia

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

4.28 [0.24, 75.57]

18.15 Rash

2

711

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.15, 6.09]

18.16 Restlessness

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

5.24 [0.32, 84.52]

18.17 Slight illness

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

5.83 [0.36, 93.84]

18.18 Somnolence

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

1.55 [0.21, 11.40]

18.19 Tinnitus

1

477

Risk Ratio (M‐H, Fixed, 95% CI)

2.31 [0.13, 42.64]

18.20 Circulatory disorders

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

6.38 [0.36, 114.01]

19 Other adverse events (cohort studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 3.19

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 19 Other adverse events (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 19 Other adverse events (cohort studies).

19.1 Adjustment disorder

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.76 [1.54, 2.02]

19.2 Confusion

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.04, 25.96]

19.3 Convulsions

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.79, 2.30]

19.4 Hallucinations

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

0.25 [0.08, 0.79]

19.5 Paranoia

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.76 [0.08, 36.72]

19.6 PTSD

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

2.51 [1.93, 3.26]

19.7 Suicidal ideation

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.03, 2.77]

19.8 Suicide

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.06, 7.78]

19.9 Tinnitus

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.42 [1.21, 1.68]

20 Adherence (RCTs) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.20

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 20 Adherence (RCTs).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 20 Adherence (RCTs).

20.1 van Riemsdijk 2002

1

119

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.88, 1.02]

20.2 Overbosch 2001; during travel

1

966

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.95, 1.01]

20.3 Overbosch 2001; post‐travel

1

966

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.74, 0.85]

21 Adherence (cohort studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.21

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 21 Adherence (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 21 Adherence (cohort studies).

21.1 During travel

6

5577

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.86, 1.34]

21.2 Post‐travel

2

422

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.64, 1.23]

Open in table viewer
Comparison 4. Mefloquine versus chloroquine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

4

877

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.28, 0.52]

Analysis 4.1

Comparison 4 Mefloquine versus chloroquine, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 1 Clinical cases of malaria (RCTs).

2 Serious adverse events or effects (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 Mefloquine versus chloroquine, Outcome 2 Serious adverse events or effects (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 2 Serious adverse events or effects (all studies).

2.1 RCTs

4

1000

Risk Ratio (M‐H, Fixed, 95% CI)

2.77 [0.32, 23.85]

2.2 Cohort studies

6

79257

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.62, 2.07]

3 Discontinuations due to adverse effects (all studies) Show forest plot

11

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.3

Comparison 4 Mefloquine versus chloroquine, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 3 Discontinuations due to adverse effects (all studies).

3.1 RCTs

3

815

Risk Ratio (M‐H, Fixed, 95% CI)

1.60 [0.61, 4.18]

3.2 Cohort studies in short‐term travellers

6

55397

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.78, 1.26]

3.3 Cohort studies in longer term occupational travellers

2

6085

Risk Ratio (M‐H, Fixed, 95% CI)

2.97 [2.41, 3.66]

4 Nausea (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.4

Comparison 4 Mefloquine versus chloroquine, Outcome 4 Nausea (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 4 Nausea (all studies).

4.1 Cohort studies (adverse effects)

6

58984

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.89, 1.68]

4.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.57, 1.79]

5 Vomiting (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.5

Comparison 4 Mefloquine versus chloroquine, Outcome 5 Vomiting (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 5 Vomiting (all studies).

5.1 Cohort studies (adverse effects)

5

5577

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.78, 1.40]

5.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.36, 3.49]

6 Abdominal pain (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.6

Comparison 4 Mefloquine versus chloroquine, Outcome 6 Abdominal pain (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 6 Abdominal pain (all studies).

6.1 Cohort studies (adverse effects)

4

5440

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.80, 1.22]

6.2 RCTs (adverse events)

2

569

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.37, 1.36]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.7

Comparison 4 Mefloquine versus chloroquine, Outcome 7 Diarrhoea (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 7 Diarrhoea (all studies).

7.1 Cohort studies (adverse effects)

5

5577

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.74, 0.95]

7.2 RCTs (adverse events)

3

772

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.46, 1.50]

8 Headache (all studies) Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.8

Comparison 4 Mefloquine versus chloroquine, Outcome 8 Headache (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 8 Headache (all studies).

8.1 Cohort studies (adverse effects)

6

56998

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.53, 1.34]

8.2 RCTs (adverse events)

3

772

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.61, 1.31]

9 Dizziness (all studies) Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.9

Comparison 4 Mefloquine versus chloroquine, Outcome 9 Dizziness (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 9 Dizziness (all studies).

9.1 Cohort studies (adverse effects)

5

58847

Risk Ratio (M‐H, Fixed, 95% CI)

1.51 [1.34, 1.70]

9.2 RCTs (adverse events)

2

569

Risk Ratio (M‐H, Fixed, 95% CI)

0.72 [0.35, 1.46]

10 Abnormal dreams (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.10

Comparison 4 Mefloquine versus chloroquine, Outcome 10 Abnormal dreams (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 10 Abnormal dreams (all studies).

10.1 Cohort studies (adverse effects)

4

2845

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [1.10, 1.33]

10.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

2.70 [1.05, 6.95]

11 Insomnia (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.11

Comparison 4 Mefloquine versus chloroquine, Outcome 11 Insomnia (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 11 Insomnia (all studies).

11.1 Cohort studies (adverse effects)

5

56952

Risk Ratio (M‐H, Random, 95% CI)

1.81 [0.73, 4.51]

11.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.76, 1.84]

12 Anxiety (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.12

Comparison 4 Mefloquine versus chloroquine, Outcome 12 Anxiety (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 12 Anxiety (all studies).

12.1 Cohort studies (adverse effects)

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

6.30 [4.37, 9.09]

13 Depressed mood (all studies) Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.13

Comparison 4 Mefloquine versus chloroquine, Outcome 13 Depressed mood (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 13 Depressed mood (all studies).

13.1 Cohort studies (adverse effects)

5

58855

Risk Ratio (M‐H, Random, 95% CI)

3.14 [1.15, 8.57]

14 Abnormal thoughts and perceptions Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.14

Comparison 4 Mefloquine versus chloroquine, Outcome 14 Abnormal thoughts and perceptions.

Comparison 4 Mefloquine versus chloroquine, Outcome 14 Abnormal thoughts and perceptions.

14.1 Cohort studies (adverse effects)

4

4831

Risk Ratio (M‐H, Fixed, 95% CI)

5.49 [2.65, 11.35]

15 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.15

Comparison 4 Mefloquine versus chloroquine, Outcome 15 Pruritis (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 15 Pruritis (all studies).

15.1 Cohort studies (adverse effects)

2

55544

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.92, 1.40]

15.2 RCTs (adverse events)

2

413

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.03, 2.93]

16 Visual impairment (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.16

Comparison 4 Mefloquine versus chloroquine, Outcome 16 Visual impairment (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 16 Visual impairment (all studies).

16.1 Cohort studies (adverse effects)

5

58847

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.50, 2.44]

16.2 RCTs (adverse events)

1

210

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.01, 2.63]

17 Vertigo (all studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.17

Comparison 4 Mefloquine versus chloroquine, Outcome 17 Vertigo (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 17 Vertigo (all studies).

17.1 Cohort studies (adverse effects)

1

746

Risk Ratio (M‐H, Fixed, 95% CI)

1.13 [0.05, 23.43]

18 Cohort studies in travellers; prespecified adverse effects Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.18

Comparison 4 Mefloquine versus chloroquine, Outcome 18 Cohort studies in travellers; prespecified adverse effects.

Comparison 4 Mefloquine versus chloroquine, Outcome 18 Cohort studies in travellers; prespecified adverse effects.

18.1 Vertigo

1

746

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.05, 23.43]

18.2 Nausea

5

56847

Risk Ratio (M‐H, Random, 95% CI)

1.42 [0.94, 2.13]

18.3 Vomiting

4

3440

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.55, 1.42]

18.4 Abdominal pain

3

3303

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.74, 1.30]

18.5 Diarrhoea

4

3440

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.57, 2.64]

18.6 Headache

5

54861

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.48, 2.65]

18.7 Dizziness

4

56710

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.10, 2.10]

18.8 Abnormal dreams

3

708

Risk Ratio (M‐H, Random, 95% CI)

4.21 [0.57, 31.33]

18.9 Insomnia

4

54815

Risk Ratio (M‐H, Random, 95% CI)

1.56 [0.40, 6.10]

18.10 Anxiety

2

1271

Risk Ratio (M‐H, Random, 95% CI)

3.94 [0.53, 29.48]

18.11 Depressed mood

4

56710

Risk Ratio (M‐H, Random, 95% CI)

2.49 [0.75, 8.31]

18.12 Abnormal thoughts or perceptions

3

2694

Risk Ratio (M‐H, Random, 95% CI)

4.42 [1.58, 12.40]

18.13 Pruritis

1

53407

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.94, 1.48]

18.14 Visual impairment

4

56710

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.55, 0.79]

19 Other adverse effects (cohort studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.19

Comparison 4 Mefloquine versus chloroquine, Outcome 19 Other adverse effects (cohort studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 19 Other adverse effects (cohort studies).

19.1 Altered spatial perception

1

2032

Risk Ratio (M‐H, Fixed, 95% CI)

3.16 [1.55, 6.45]

19.2 Alopecia

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.27, 2.25]

19.3 Asthenia

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.97, 2.40]

19.4 Balance disorder

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

3.59 [2.15, 6.00]

19.5 Confusion

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.11, 36.31]

19.6 Decreased appetite

1

2032

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.87, 1.57]

19.7 Fatigue

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [0.57, 9.80]

19.8 Hypoaesthesia

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

20.26 [1.23, 333.93]

19.9 Irritability

1

746

Risk Ratio (M‐H, Fixed, 95% CI)

4.75 [0.28, 80.59]

19.10 Mouth ulcers

2

55439

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [1.01, 1.87]

19.11 Paraesthesia

2

2778

Risk Ratio (M‐H, Fixed, 95% CI)

2.22 [1.27, 3.89]

19.12 Palpitations

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

4.71 [0.91, 24.26]

19.13 Photosensitization

2

2662

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.52, 1.53]

19.14 Restlessness

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

4.74 [0.65, 34.46]

19.15 Slight illness

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.65 [0.64, 10.87]

19.16 Somnolence

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

6.08 [0.37, 100.36]

19.17 Yeast infection

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.53, 2.49]

20 Other adverse events (RCTs) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.20

Comparison 4 Mefloquine versus chloroquine, Outcome 20 Other adverse events (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 20 Other adverse events (RCTs).

20.1 Abdominal distension

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

3.13 [0.64, 15.27]

20.2 Anger

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.07, 1.55]

20.3 Disturbance in attention

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

3.16 [0.61, 16.47]

20.4 Irritability

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.45, 2.64]

20.5 Loss of appetite

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.35, 3.25]

20.6 Malaise

1

203

Risk Ratio (M‐H, Fixed, 95% CI)

0.32 [0.01, 7.85]

20.7 Mood altered

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.13 [0.29, 4.34]

21 Pregnancy related outcomes (RCTs) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

Analysis 4.21

Comparison 4 Mefloquine versus chloroquine, Outcome 21 Pregnancy related outcomes (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 21 Pregnancy related outcomes (RCTs).

21.1 Spontaneous abortions

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.36, 1.79]

21.2 Still births

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.67, 1.52]

21.3 Congenital malformations

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22 Adherence (cohort studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.22

Comparison 4 Mefloquine versus chloroquine, Outcome 22 Adherence (cohort studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 22 Adherence (cohort studies).

22.1 Short‐term travellers

3

852

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.90, 1.13]

22.2 Short‐term travellers: after return

1

46

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.54, 1.87]

22.3 Longer‐term occupational travellers

2

5777

Risk Ratio (M‐H, Random, 95% CI)

2.02 [1.80, 2.26]

Open in table viewer
Comparison 5. Mefloquine versus currently used regimens; by study design

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Nausea; effects Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 1 Nausea; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 1 Nausea; effects.

1.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.72 [1.52, 4.86]

1.2 Cohort studies

11

5973

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.78, 3.77]

2 Abdominal pain; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.2

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 2 Abdominal pain; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 2 Abdominal pain; effects.

2.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.52, 1.56]

2.2 Cohort studies

9

4494

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.27, 0.87]

3 Diarrhoea; effects Show forest plot

11

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.3

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 3 Diarrhoea; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 3 Diarrhoea; effects.

3.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.60, 1.47]

3.2 Cohort studies

10

7648

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.28, 1.34]

4 Headache; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.4

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 4 Headache; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 4 Headache; effects.

4.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.99, 2.99]

4.2 Cohort studies

9

5592

Risk Ratio (M‐H, Random, 95% CI)

2.19 [1.22, 3.93]

5 Dizziness; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.5

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 5 Dizziness; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 5 Dizziness; effects.

5.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

3.99 [2.08, 7.64]

5.2 Cohort studies

9

4606

Risk Ratio (M‐H, Random, 95% CI)

3.17 [1.58, 6.35]

6 Abnormal dreams; effects Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.6

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 6 Abnormal dreams; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 6 Abnormal dreams; effects.

6.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [1.37, 3.04]

6.2 Cohort studies

7

4543

Risk Ratio (M‐H, Random, 95% CI)

7.30 [2.51, 21.18]

7 Insomnia; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.7

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 7 Insomnia; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 7 Insomnia; effects.

7.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

4.42 [2.56, 7.64]

7.2 Cohort studies

9

5299

Risk Ratio (M‐H, Random, 95% CI)

5.70 [2.83, 11.47]

8 Anxiety; effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.8

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 8 Anxiety; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 8 Anxiety; effects.

8.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

6.12 [1.82, 20.66]

8.2 Cohort studies

4

3390

Risk Ratio (M‐H, Random, 95% CI)

15.26 [8.66, 26.89]

9 Depressed mood; effects Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.9

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 9 Depressed mood; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 9 Depressed mood; effects.

9.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

5.78 [1.71, 19.61]

9.2 Cohort studies

6

4236

Risk Ratio (M‐H, Random, 95% CI)

7.82 [3.79, 16.12]

10 Abnormal thoughts or perceptions; effects Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.10

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 10 Abnormal thoughts or perceptions; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 10 Abnormal thoughts or perceptions; effects.

10.1 Cohort studies

3

3045

Risk Ratio (M‐H, Random, 95% CI)

4.20 [0.81, 21.87]

11 Pruritis; effects Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.11

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 11 Pruritis; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 11 Pruritis; effects.

11.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.60, 2.70]

11.2 Cohort studies

3

2034

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.16, 4.76]

12 Visual impairment; effects Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.12

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 12 Visual impairment; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 12 Visual impairment; effects.

12.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [0.88, 4.73]

12.2 Cohort studies

3

2560

Risk Ratio (M‐H, Random, 95% CI)

2.06 [1.05, 4.02]

13 Adherence; during travel Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.13

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 13 Adherence; during travel.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 13 Adherence; during travel.

13.1 RCTs

1

119

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.88, 1.02]

13.2 Cohort studies

11

12131

Risk Ratio (M‐H, Random, 95% CI)

1.16 [1.03, 1.30]

14 Adherence; after return Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.14

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 14 Adherence; after return.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 14 Adherence; after return.

14.1 Cohort studies

4

1221

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.92, 1.17]

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

'Risk of bias' summary for RCTs: review authors' judgements about each 'Risk of bias' item for each included study.
Figuras y tablas -
Figure 2

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

'Risk of bias' summary in cohort studies: mefloquine versus placebo/no treatment1Assesses whether our pre‐defined confounders were measured and balanced across groups.
 2Assesses the non‐response rate of prospective participants.
 3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
 4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
 5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
 6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
 7Assesses whether it is clear that all information collected within the study has been reported.
 8Assess the risk of bias due to influence by a corporate study sponsor.
Figuras y tablas -
Figure 3

'Risk of bias' summary in cohort studies: mefloquine versus placebo/no treatment

1Assesses whether our pre‐defined confounders were measured and balanced across groups.
2Assesses the non‐response rate of prospective participants.
3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
7Assesses whether it is clear that all information collected within the study has been reported.
8Assess the risk of bias due to influence by a corporate study sponsor.

'Risk of bias' summary in cohort studies: mefloquine versus doxycycline1Assesses whether our pre‐defined confounders are measured and balanced across groups.
 2Assesses the non‐response rate of prospective participants.
 3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
 4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
 5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
 6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
 7Assesses whether it is clear that all information collected within the study has been reported.
 8Assesses the risk of bias due to influence by a corporate study sponsor.
Figuras y tablas -
Figure 4

'Risk of bias' summary in cohort studies: mefloquine versus doxycycline

1Assesses whether our pre‐defined confounders are measured and balanced across groups.
2Assesses the non‐response rate of prospective participants.
3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
7Assesses whether it is clear that all information collected within the study has been reported.
8Assesses the risk of bias due to influence by a corporate study sponsor.

'Risk of bias' summary in cohort studies: mefloquine versus atovaquone‐proguanil1Assesses whether our pre‐defined confounders are measured and balanced across groups.
 2Assesses the non‐response rate of prospective participants.
 3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
 4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
 5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
 6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
 7Assesses whether it is clear that all information collected within the study has been reported.
 8Assesses the risk of bias due to influence by a corporate study sponsor.
Figuras y tablas -
Figure 5

'Risk of bias' summary in cohort studies: mefloquine versus atovaquone‐proguanil

1Assesses whether our pre‐defined confounders are measured and balanced across groups.
2Assesses the non‐response rate of prospective participants.
3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
7Assesses whether it is clear that all information collected within the study has been reported.
8Assesses the risk of bias due to influence by a corporate study sponsor.

'Risk of bias' summary in cohort studies: mefloquine versus chloroquine1Assesses whether our pre‐defined confounders are measured and balanced across groups.
 2Assesses the non‐response rate of prospective participants.
 3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
 4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
 5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
 6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
 7Assesses whether it is clear that all information collected within the study has been reported.
 8Assesses the risk of bias due to influence by a corporate study sponsor.
Figuras y tablas -
Figure 6

'Risk of bias' summary in cohort studies: mefloquine versus chloroquine

1Assesses whether our pre‐defined confounders are measured and balanced across groups.
2Assesses the non‐response rate of prospective participants.
3Assesses the risk that participants labelled as taking mefloquine (or another antimalarial) actually took something else.
4Assesses the risk that participants whose adverse effects are attributed to mefloquine (or another antimalarial) actually took another drug as well.
5Assesses whether outcome data reasonably complete for most participants and whether intervention status reasonably complete for those in whom it was sought.
6Assesses whether the outcome measure was subjective, and whether participants and outcome assessors were blinded.
7Assesses whether it is clear that all information collected within the study has been reported.
8Assesses the risk of bias due to influence by a corporate study sponsor.

Comparison 1 Mefloquine versus placebo/non users, Outcome 1 Clinical cases of malaria.
Figuras y tablas -
Analysis 1.1

Comparison 1 Mefloquine versus placebo/non users, Outcome 1 Clinical cases of malaria.

Comparison 1 Mefloquine versus placebo/non users, Outcome 2 Malaria; episodes of parasitaemia in semi‐immune populations.
Figuras y tablas -
Analysis 1.2

Comparison 1 Mefloquine versus placebo/non users, Outcome 2 Malaria; episodes of parasitaemia in semi‐immune populations.

Comparison 1 Mefloquine versus placebo/non users, Outcome 3 Serious adverse events or effects (all studies).
Figuras y tablas -
Analysis 1.3

Comparison 1 Mefloquine versus placebo/non users, Outcome 3 Serious adverse events or effects (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 4 Discontinuations due to adverse effects (all studies).
Figuras y tablas -
Analysis 1.4

Comparison 1 Mefloquine versus placebo/non users, Outcome 4 Discontinuations due to adverse effects (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 5 Nausea (all studies).
Figuras y tablas -
Analysis 1.5

Comparison 1 Mefloquine versus placebo/non users, Outcome 5 Nausea (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 6 Vomiting (all studies).
Figuras y tablas -
Analysis 1.6

Comparison 1 Mefloquine versus placebo/non users, Outcome 6 Vomiting (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 7 Abdominal pain (all studies).
Figuras y tablas -
Analysis 1.7

Comparison 1 Mefloquine versus placebo/non users, Outcome 7 Abdominal pain (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 8 Diarrhoea (all studies).
Figuras y tablas -
Analysis 1.8

Comparison 1 Mefloquine versus placebo/non users, Outcome 8 Diarrhoea (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 9 Headache (all studies).
Figuras y tablas -
Analysis 1.9

Comparison 1 Mefloquine versus placebo/non users, Outcome 9 Headache (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 10 Dizziness (all studies).
Figuras y tablas -
Analysis 1.10

Comparison 1 Mefloquine versus placebo/non users, Outcome 10 Dizziness (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 11 Abnormal dreams (all studies).
Figuras y tablas -
Analysis 1.11

Comparison 1 Mefloquine versus placebo/non users, Outcome 11 Abnormal dreams (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 12 Insomnia (all studies).
Figuras y tablas -
Analysis 1.12

Comparison 1 Mefloquine versus placebo/non users, Outcome 12 Insomnia (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 13 Anxiety (all studies).
Figuras y tablas -
Analysis 1.13

Comparison 1 Mefloquine versus placebo/non users, Outcome 13 Anxiety (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 14 Depressed mood (all studies).
Figuras y tablas -
Analysis 1.14

Comparison 1 Mefloquine versus placebo/non users, Outcome 14 Depressed mood (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 15 Abnormal thoughts and perceptions.
Figuras y tablas -
Analysis 1.15

Comparison 1 Mefloquine versus placebo/non users, Outcome 15 Abnormal thoughts and perceptions.

Comparison 1 Mefloquine versus placebo/non users, Outcome 16 Pruritis (all studies).
Figuras y tablas -
Analysis 1.16

Comparison 1 Mefloquine versus placebo/non users, Outcome 16 Pruritis (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 17 Visual impairment (all studies).
Figuras y tablas -
Analysis 1.17

Comparison 1 Mefloquine versus placebo/non users, Outcome 17 Visual impairment (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 18 Vertigo (all studies).
Figuras y tablas -
Analysis 1.18

Comparison 1 Mefloquine versus placebo/non users, Outcome 18 Vertigo (all studies).

Comparison 1 Mefloquine versus placebo/non users, Outcome 19 Other adverse events (RCTs).
Figuras y tablas -
Analysis 1.19

Comparison 1 Mefloquine versus placebo/non users, Outcome 19 Other adverse events (RCTs).

Comparison 1 Mefloquine versus placebo/non users, Outcome 20 Other adverse effects (cohort studies).
Figuras y tablas -
Analysis 1.20

Comparison 1 Mefloquine versus placebo/non users, Outcome 20 Other adverse effects (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 1 Clinical cases of malaria (RCTs).
Figuras y tablas -
Analysis 2.1

Comparison 2 Mefloquine versus doxycycline, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 2 Mefloquine versus doxycycline, Outcome 2 Serious adverse events or effects (all studies).
Figuras y tablas -
Analysis 2.2

Comparison 2 Mefloquine versus doxycycline, Outcome 2 Serious adverse events or effects (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 3 Discontinuations due to adverse effects (all studies).
Figuras y tablas -
Analysis 2.3

Comparison 2 Mefloquine versus doxycycline, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 4 Nausea (all studies).
Figuras y tablas -
Analysis 2.4

Comparison 2 Mefloquine versus doxycycline, Outcome 4 Nausea (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 5 Vomiting (all studies).
Figuras y tablas -
Analysis 2.5

Comparison 2 Mefloquine versus doxycycline, Outcome 5 Vomiting (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 6 Abdominal pain (all studies).
Figuras y tablas -
Analysis 2.6

Comparison 2 Mefloquine versus doxycycline, Outcome 6 Abdominal pain (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 7 Diarrhoea (all studies).
Figuras y tablas -
Analysis 2.7

Comparison 2 Mefloquine versus doxycycline, Outcome 7 Diarrhoea (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 8 Dyspepsia (all studies).
Figuras y tablas -
Analysis 2.8

Comparison 2 Mefloquine versus doxycycline, Outcome 8 Dyspepsia (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 9 Headache (all studies).
Figuras y tablas -
Analysis 2.9

Comparison 2 Mefloquine versus doxycycline, Outcome 9 Headache (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 10 Dizziness (all studies).
Figuras y tablas -
Analysis 2.10

Comparison 2 Mefloquine versus doxycycline, Outcome 10 Dizziness (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 11 Abnormal dreams (all studies).
Figuras y tablas -
Analysis 2.11

Comparison 2 Mefloquine versus doxycycline, Outcome 11 Abnormal dreams (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 12 Insomnia (all studies).
Figuras y tablas -
Analysis 2.12

Comparison 2 Mefloquine versus doxycycline, Outcome 12 Insomnia (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 13 Anxiety (all studies).
Figuras y tablas -
Analysis 2.13

Comparison 2 Mefloquine versus doxycycline, Outcome 13 Anxiety (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 14 Depressed mood (all studies).
Figuras y tablas -
Analysis 2.14

Comparison 2 Mefloquine versus doxycycline, Outcome 14 Depressed mood (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 15 Abnormal thoughts and perceptions.
Figuras y tablas -
Analysis 2.15

Comparison 2 Mefloquine versus doxycycline, Outcome 15 Abnormal thoughts and perceptions.

Comparison 2 Mefloquine versus doxycycline, Outcome 16 Pruritis (all studies).
Figuras y tablas -
Analysis 2.16

Comparison 2 Mefloquine versus doxycycline, Outcome 16 Pruritis (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 17 Photosensitivity (all studies).
Figuras y tablas -
Analysis 2.17

Comparison 2 Mefloquine versus doxycycline, Outcome 17 Photosensitivity (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 18 Yeast infection (all studies).
Figuras y tablas -
Analysis 2.18

Comparison 2 Mefloquine versus doxycycline, Outcome 18 Yeast infection (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 19 Visual impairment (all studies).
Figuras y tablas -
Analysis 2.19

Comparison 2 Mefloquine versus doxycycline, Outcome 19 Visual impairment (all studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 20 Other adverse effects (cohort studies).
Figuras y tablas -
Analysis 2.20

Comparison 2 Mefloquine versus doxycycline, Outcome 20 Other adverse effects (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 21 Other adverse events (RCTs).
Figuras y tablas -
Analysis 2.21

Comparison 2 Mefloquine versus doxycycline, Outcome 21 Other adverse events (RCTs).

Comparison 2 Mefloquine versus doxycycline, Outcome 22 Other adverse events (cohort studies).
Figuras y tablas -
Analysis 2.22

Comparison 2 Mefloquine versus doxycycline, Outcome 22 Other adverse events (cohort studies).

Comparison 2 Mefloquine versus doxycycline, Outcome 23 Adherence (cohort studies).
Figuras y tablas -
Analysis 2.23

Comparison 2 Mefloquine versus doxycycline, Outcome 23 Adherence (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 1 Clinical cases of malaria (RCTs).
Figuras y tablas -
Analysis 3.1

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 2 Serious adverse events or effects (all studies).
Figuras y tablas -
Analysis 3.2

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 2 Serious adverse events or effects (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 3 Discontinuations due to adverse effects (all studies).
Figuras y tablas -
Analysis 3.3

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 4 Nausea (all studies).
Figuras y tablas -
Analysis 3.4

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 4 Nausea (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 5 Vomiting (all studies).
Figuras y tablas -
Analysis 3.5

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 5 Vomiting (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 6 Abdominal pain (all studies).
Figuras y tablas -
Analysis 3.6

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 6 Abdominal pain (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 7 Diarrhoea (all studies).
Figuras y tablas -
Analysis 3.7

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 7 Diarrhoea (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 8 Mouth ulcers (all studies).
Figuras y tablas -
Analysis 3.8

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 8 Mouth ulcers (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 9 Headache (all studies).
Figuras y tablas -
Analysis 3.9

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 9 Headache (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 10 Dizziness (all studies).
Figuras y tablas -
Analysis 3.10

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 10 Dizziness (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 11 Abnormal dreams (all studies).
Figuras y tablas -
Analysis 3.11

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 11 Abnormal dreams (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 12 Insomnia (all studies).
Figuras y tablas -
Analysis 3.12

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 12 Insomnia (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 13 Anxiety (all studies).
Figuras y tablas -
Analysis 3.13

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 13 Anxiety (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 14 Depressed mood (all studies).
Figuras y tablas -
Analysis 3.14

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 14 Depressed mood (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 15 Abnormal thoughts and perceptions (all studies).
Figuras y tablas -
Analysis 3.15

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 15 Abnormal thoughts and perceptions (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 16 Pruritis (all studies).
Figuras y tablas -
Analysis 3.16

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 16 Pruritis (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 17 Visual impairment (all studies).
Figuras y tablas -
Analysis 3.17

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 17 Visual impairment (all studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 18 Other adverse effects (cohort studies).
Figuras y tablas -
Analysis 3.18

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 18 Other adverse effects (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 19 Other adverse events (cohort studies).
Figuras y tablas -
Analysis 3.19

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 19 Other adverse events (cohort studies).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 20 Adherence (RCTs).
Figuras y tablas -
Analysis 3.20

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 20 Adherence (RCTs).

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 21 Adherence (cohort studies).
Figuras y tablas -
Analysis 3.21

Comparison 3 Mefloquine versus atovaquone‐proguanil, Outcome 21 Adherence (cohort studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 1 Clinical cases of malaria (RCTs).
Figuras y tablas -
Analysis 4.1

Comparison 4 Mefloquine versus chloroquine, Outcome 1 Clinical cases of malaria (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 2 Serious adverse events or effects (all studies).
Figuras y tablas -
Analysis 4.2

Comparison 4 Mefloquine versus chloroquine, Outcome 2 Serious adverse events or effects (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 3 Discontinuations due to adverse effects (all studies).
Figuras y tablas -
Analysis 4.3

Comparison 4 Mefloquine versus chloroquine, Outcome 3 Discontinuations due to adverse effects (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 4 Nausea (all studies).
Figuras y tablas -
Analysis 4.4

Comparison 4 Mefloquine versus chloroquine, Outcome 4 Nausea (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 5 Vomiting (all studies).
Figuras y tablas -
Analysis 4.5

Comparison 4 Mefloquine versus chloroquine, Outcome 5 Vomiting (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 6 Abdominal pain (all studies).
Figuras y tablas -
Analysis 4.6

Comparison 4 Mefloquine versus chloroquine, Outcome 6 Abdominal pain (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 7 Diarrhoea (all studies).
Figuras y tablas -
Analysis 4.7

Comparison 4 Mefloquine versus chloroquine, Outcome 7 Diarrhoea (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 8 Headache (all studies).
Figuras y tablas -
Analysis 4.8

Comparison 4 Mefloquine versus chloroquine, Outcome 8 Headache (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 9 Dizziness (all studies).
Figuras y tablas -
Analysis 4.9

Comparison 4 Mefloquine versus chloroquine, Outcome 9 Dizziness (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 10 Abnormal dreams (all studies).
Figuras y tablas -
Analysis 4.10

Comparison 4 Mefloquine versus chloroquine, Outcome 10 Abnormal dreams (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 11 Insomnia (all studies).
Figuras y tablas -
Analysis 4.11

Comparison 4 Mefloquine versus chloroquine, Outcome 11 Insomnia (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 12 Anxiety (all studies).
Figuras y tablas -
Analysis 4.12

Comparison 4 Mefloquine versus chloroquine, Outcome 12 Anxiety (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 13 Depressed mood (all studies).
Figuras y tablas -
Analysis 4.13

Comparison 4 Mefloquine versus chloroquine, Outcome 13 Depressed mood (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 14 Abnormal thoughts and perceptions.
Figuras y tablas -
Analysis 4.14

Comparison 4 Mefloquine versus chloroquine, Outcome 14 Abnormal thoughts and perceptions.

Comparison 4 Mefloquine versus chloroquine, Outcome 15 Pruritis (all studies).
Figuras y tablas -
Analysis 4.15

Comparison 4 Mefloquine versus chloroquine, Outcome 15 Pruritis (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 16 Visual impairment (all studies).
Figuras y tablas -
Analysis 4.16

Comparison 4 Mefloquine versus chloroquine, Outcome 16 Visual impairment (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 17 Vertigo (all studies).
Figuras y tablas -
Analysis 4.17

Comparison 4 Mefloquine versus chloroquine, Outcome 17 Vertigo (all studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 18 Cohort studies in travellers; prespecified adverse effects.
Figuras y tablas -
Analysis 4.18

Comparison 4 Mefloquine versus chloroquine, Outcome 18 Cohort studies in travellers; prespecified adverse effects.

Comparison 4 Mefloquine versus chloroquine, Outcome 19 Other adverse effects (cohort studies).
Figuras y tablas -
Analysis 4.19

Comparison 4 Mefloquine versus chloroquine, Outcome 19 Other adverse effects (cohort studies).

Comparison 4 Mefloquine versus chloroquine, Outcome 20 Other adverse events (RCTs).
Figuras y tablas -
Analysis 4.20

Comparison 4 Mefloquine versus chloroquine, Outcome 20 Other adverse events (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 21 Pregnancy related outcomes (RCTs).
Figuras y tablas -
Analysis 4.21

Comparison 4 Mefloquine versus chloroquine, Outcome 21 Pregnancy related outcomes (RCTs).

Comparison 4 Mefloquine versus chloroquine, Outcome 22 Adherence (cohort studies).
Figuras y tablas -
Analysis 4.22

Comparison 4 Mefloquine versus chloroquine, Outcome 22 Adherence (cohort studies).

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 1 Nausea; effects.
Figuras y tablas -
Analysis 5.1

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 1 Nausea; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 2 Abdominal pain; effects.
Figuras y tablas -
Analysis 5.2

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 2 Abdominal pain; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 3 Diarrhoea; effects.
Figuras y tablas -
Analysis 5.3

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 3 Diarrhoea; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 4 Headache; effects.
Figuras y tablas -
Analysis 5.4

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 4 Headache; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 5 Dizziness; effects.
Figuras y tablas -
Analysis 5.5

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 5 Dizziness; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 6 Abnormal dreams; effects.
Figuras y tablas -
Analysis 5.6

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 6 Abnormal dreams; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 7 Insomnia; effects.
Figuras y tablas -
Analysis 5.7

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 7 Insomnia; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 8 Anxiety; effects.
Figuras y tablas -
Analysis 5.8

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 8 Anxiety; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 9 Depressed mood; effects.
Figuras y tablas -
Analysis 5.9

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 9 Depressed mood; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 10 Abnormal thoughts or perceptions; effects.
Figuras y tablas -
Analysis 5.10

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 10 Abnormal thoughts or perceptions; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 11 Pruritis; effects.
Figuras y tablas -
Analysis 5.11

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 11 Pruritis; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 12 Visual impairment; effects.
Figuras y tablas -
Analysis 5.12

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 12 Visual impairment; effects.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 13 Adherence; during travel.
Figuras y tablas -
Analysis 5.13

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 13 Adherence; during travel.

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 14 Adherence; after return.
Figuras y tablas -
Analysis 5.14

Comparison 5 Mefloquine versus currently used regimens; by study design, Outcome 14 Adherence; after return.

Summary of findings for the main comparison. Mefloquine versus atovaquone‐proguanil for preventing malaria in travellers

Mefloquine compared with atovaquone‐proguanil for preventing malaria in travellers

Population: non‐immune adults and children travelling to or living in malaria‐endemic settings

Intervention: mefloquine 250 mg weekly

Comparison: atovaquone‐proguanil (250 mg atovaquone and 100 mg proguanil hydrochloride) daily

Outcome data collection: physicians performed blinded assessment of whether reported symptoms could be related to the study drug

Outcomes

Anticipated absolute effects*
(95% CI)

Relative effect
(95% CI)

Studies contributing to effect estimate
(participants)

Additional studies considered in GRADE assessment
(participants)

Certainty of the evidence
(GRADE)

Atovoquone‐proguanil

Mefloquine

Clinical malaria

2 RCTs

(1293)

⊕⊕⊝⊝
low1,2,3

Serious adverse effects

0 per 100

1 in 100

(0 to 12)

RR 1.40

(0.08 to 23.22)

4 cohort studies

(3693)

1 RCT

(976)

⊕⊕⊝⊝
low1,2,4,5

Discontinuation of drug due to adverse effects

2 per 100

6 per 100

(3 to 11)

RR 2.86

(1.53 to 5.31)

3 RCTs

(1438)

7 cohort studies

(4498)

⊕⊕⊕⊕
high1,2,4,6

Abnormal dreams

7 per 100

14 per 100

(10 to 21)

RR 2.04

(1.37 to 3.04)

1 RCT

(976)

7 cohort studies

(3848)

⊕⊕⊕⊕
high1,2,4,6

Insomnia

3 per 100

13 per 100

(8 to 23)

RR 4.42

(2.56 to 7.64)

1 RCT

(976)

8 cohort studies

(3986)

⊕⊕⊕⊕
high1,2,4,6

Anxiety

1 per 100

6 per 100

(2 to 21)

RR 6.12

(1.82 to 20.66)

1 RCT

(976)

4 cohort studies

(2664)

⊕⊕⊕⊝
moderate1,2,4,7

Depressed mood

1 per 100

6 per 100

(2 to 20)

RR 5.78

(1.71 to 19.61)

1 RCT

(976)

6 cohort studies

(3624)

⊕⊕⊕⊝
moderate1,2,4,7

Abnormal thoughts or perceptions

0 per 100

1 per 100

(0 to 4)

RR 1.50

(0.30 to 7.42)

3 cohort studies

(2433)

⊕⊝⊝⊝
very low1,2,8

Nausea

3 per 100

8 per 100

(5 to 15)

RR 2.72

(1.52 to 4.86)

1 RCT

(976)

7 cohort studies

(3509)

⊕⊕⊕⊕
high1,2,4,6

Vomiting

1 per 100

1 per 100

(0 to 4)

RR 1.31 (0.49 to 3.50)

1 RCT

(976)

3 cohort studies

(2180)

⊕⊕⊕⊝
moderate1,2,4,7

Abdominal pain

5 per 100

5 per 100

(3 to 8)

RR 0.90

(0.52 to 1.56)

1 RCT

(976)

7 cohort studies

(3509)

⊕⊕⊝⊝
moderate1,2,4,8

Diarrhoea

8 per 100

8 per 100

(5 to 12)

RR 0.94

(0.60 to 1.47)

1 RCT

(976)

7 cohort studies

(3509)

⊕⊕⊕⊝
moderate1,2,4,8

Headache

4 per 100

7 per 100

(4 to 12)

RR 1.72

(0.99 to 2.99)

1 RCT

(976)

8 cohort studies

(4163)

⊕⊕⊕⊝
moderate1,2,4,8

Dizziness

2 per 100

8 per 100

(4 to 15)

RR 3.99

(2.08 to 7.64)

1 RCT

(976)

8 cohort studies

(3986)

⊕⊕⊕⊕
high1,2,4,6

Pruritis

2 per 100

3 per 100

(1 to 5)

RR 1.28

(0.60 to 2.70)

1 RCT

(976)

3 cohort studies

(1824)

⊕⊕⊕⊝
moderate1,2,4,8

Visual impairment

2 per 100

4 per 100

(2 to 9)

RR 2.04

(0.88 to 4.73)

1 RCT

(976)

2 cohort studies

(1956)

⊕⊕⊕⊝
moderate1,2,4,8

Mouth ulcers

2 per 100

3 per 100

(1 to 6)

RR 1.45 (0.70 to 3.00)

1 RCT

(976)

2 cohort studies

(783)

⊕⊕⊕⊝
moderate1,2,4,8

*The assumed risk is the median control group risk across studies unless stated in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI). Where the control group risk was 0, we used a value of 0.5 to calculate the corresponding risk in the intervention group. Data from cohort studies were used when data from RCTs were unavailable.
Abbreviations: CI: confidence interval; RR: risk ratio

'Summary of findings' tables are usually limited to seven outcomes. For adverse effects this problematic, as there are many, and to include some and not others risks selective reporting. We have therefore included all prespecified outcomes in the table.

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

1No serious risk of bias: the RCTs were generally at low risk of bias but two of three were sponsored by the manufacturer of one of the study drugs. All cohort studies had methodological problems which could introduce confounding or bias. However, as the GRADE approach automatically downgrades certainty by two levels for non‐randomized studies, we did not downgrade further.
2No serious indirectness: the RCTs were conducted in short‐term international travellers to malaria‐endemic areas in Africa or South America for less than 28 days. The cohort studies were from a variety of populations including short‐term travellers (8 studies), longer‐term occupational travellers (3 studies) and military personnel (1 study).
3Downgraded by two levels for serious imprecision: no episodes of malaria were recorded in either trial.
4No serious inconsistency: the findings of the cohort studies were consistent with the effects seen in the RCTs.
5No serious imprecision: serious adverse effects were rare in all studies.
6No serious imprecision. The effect was statistically significant and the overall data (RCTs and cohort studies) were adequately powered to detect this effect.
7Downgraded by one level for serious imprecision: although the direction of the effect was consistent across all trials, there was substantial heterogeneity in the size of the effect.
8Downgraded by one level for serious imprecision: the 95% CI is wide and includes important effects and no effect.

Figuras y tablas -
Summary of findings for the main comparison. Mefloquine versus atovaquone‐proguanil for preventing malaria in travellers
Summary of findings 2. Mefloquine versus doxycycline for preventing malaria in travellers

Mefloquine compared with doxycycline for preventing malaria in travellers

Population: Non‐immune adults and children travelling to malaria‐endemic settings

Intervention: Mefloquine 250 mg weekly

Comparison: Doxycycline 100 mg daily

Outcome data collection: Self‐reported symptoms experienced whilst taking prophylaxis (adverse events)

Outcomes

Anticipated absolute effects* (95% CI)

Relative effect
(95% CI)

Studies contributing to effect estimate
(participants)

Additional studies considered in GRADE assessment
(participants)

Certainty of the evidence
(GRADE)

Doxycycline

Mefloquine

Clinical malaria

1 per 100

1 per 100

(0 to 5)

RR 1.35
(0.35 to 5.19)

4 RCTs

(744)

⊕⊕⊝⊝

low1,2,3,4

Serious adverse effects

6 per 10005

9 per 1000

(1 to 61)

RR 1.53

(0.23 to 10.24)

3 cohort studies

(3722)

3 RCTs, 1 cohort study

(682; 3772)

⊕⊝⊝⊝
very low2,3,6,7

Discontinuations

due to adverse effects

2 per 100

2 per 100

(1 to 6)

RR 1.08

(0.41 to 2.87)

4 RCTs

(763)

10 cohort studies

(10,165)

⊕⊕⊝⊝

low1,3,7,8

Abnormal dreams

3 per 100

31 per 100

(11 to 87)

RR 10.49

(3.79 to 29.10)

4 cohort studies

(2588)

1 RCT, 1 cohort study

(123; 688)

⊕⊝⊝⊝

very low2,6,9,10

Insomnia

3 per 100

12 per 100

(4 to 43)

RR 4.14 (1.19 to 14.44)

4 cohort studies

(3212)

1 RCT, 2 cohort studies

(123; 355,627)

⊕⊝⊝⊝

very low6,9,10,11

Anxiety

1 per 100

18 per 100

(9 to 35)

RR 18.04

(9.32 to 34.93)

3 cohort studies

(2559)

2 cohort studies

(355,627)

⊕⊝⊝⊝

very low6,9,10,11

Depressed mood

1 per 100

11 per 100

(5 to 25)

RR 11.43

(5.21 to 25.07)

2 cohort studies

(2445)

3 cohort studies

(430,006)

⊕⊝⊝⊝

very low6,9,10,11

Abnormal thoughts or perceptions

0 per 100

3 per 100

(0 to 24)

RR 6.60

(0.92 to 47.20)

2 cohort studies

(2445)

2 cohort studies

(376,024)

⊕⊝⊝⊝

very low6,9,10,11

Nausea

8 per 100

3 per 100

(2 to 4)

RR 0.37

(0.30 to 0.45)

5 cohort studies

(2683)

1 RCT, 1 cohort study

(123; 668)

⊕⊝⊝⊝

very low3,6,10,11

Vomiting

5 per 100

1 per 100

(1 to 1)

RR 0.18

(0.12 to 0.27)

4 cohort studies

(5071)

1 RCT

(123)

⊕⊝⊝⊝

very low3,6,10,11

Abdominal pain

15 per 100

5 per 100

(1 to 16)

RR 0.30

(0.09 to 1.07)

3 cohort studies

(2536)

1 RCT, 1 cohort

(123; 668)

⊕⊝⊝⊝

very low6,7,9,11

Diarrhoea

5 per 100

1 per 100

(1 to 4)

RR 0.28

(0.11 to 0.73)

5 cohort studies

(5104)

2 RCTs; 1 cohort study

(376; 668)

⊕⊝⊝⊝

very low3,6,10,11

Dyspepsia

14 per 100

4 per 100

(1 to 10)

RR 0.26

(0.09 to 0.74)

5 cohort studies

(5104)

⊕⊝⊝⊝

low2,3,6,10

Headache

2 per 100

2 per 100

(1 to 6)

RR 1.21

(0.50 to 2.92)

5 cohort studies

(3320)

1 RCT, 1 cohort study

(123; 688)

⊕⊝⊝⊝

very low3,6,7,11

Dizziness

1 per 100

3 per 100

(1 to 14)

RR 3.49

(0.88 to 13.75)

5 cohort studies

(2633)

1 RCT, 2 cohort studies

(123; 355,627)

⊕⊝⊝⊝

very low3,6,7,11

Visual impairment

3 per 100

7 per 100

(4 to 12)

RR 2.37

(1.41 to 3.99)

2 cohort studies

(1875)

⊕⊝⊝⊝

very low2,6,7,9

Pruritis

3 per 100

2 per 100

(1 to 3)

RR 0.52

(0.30 to 0.91)

2 cohort studies

(1794)

1 cohort study

(688)

⊕⊝⊝⊝

very low6,9,10,11

Photosensitivity

19 per 100

2 per 100

(1 to 2)

RR 0.08

(0.05 to 0.11)

2 cohort studies

(1875)

1 cohort study

(688)

⊕⊝⊝⊝

very low2,6,9,10

Vaginal thrush

16 per 100

2 per 100

(1 to 3)

RR 0.10

(0.06 to 0.16)

1 cohort study

(1761)

1 cohort study

(354)

⊕⊝⊝⊝

very low2,6,9,10

*The assumed risk is the median control group risk across cohort studies unless stated in footnotes. The corresponding risk (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI). Where the control group risk was 0, we used a value of 0.5 to calculate the corresponding risk in the intervention group. Where no RCTs including short‐term travellers reported on our prespecified adverse outcomes, we included information from cohort studies as our primary analysis.

'Summary of findings' tables are usually limited to seven outcomes. For adverse effects this problematic, as there are many, and to include some and not others risks selective reporting. We have therefore included all prespecified outcomes in the table.

GRADE Working Group grades of evidence
High certainty: further research is very unlikely to change our confidence in the estimate of effect.
Moderate certainty: further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low certainty: further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low certainty: we are very uncertain about the estimate.

1No serious risk of bias: none of the RCTs adequately described methods of random sequence generation or allocation concealment, However, given that so few events occurred in these trials, it is unlikely to have introduced bias.
2No serious inconsistency: the direction of the effect is consistent across study designs, or there in consistency in the finding of no effect.
3No serious indirectness: the primary analysis included studies in short‐term international travellers, longer‐term occupational travellers, and military personnel.
4Downgraded by two levels for imprecision: only seven episodes of clinical malaria occurred in the four trials, and consequently, the analysis was substantially underpowered to exclude important differences.
5For serious adverse outcomes we expressed the control group risk as the overall risk in the control group.
6No serious risk of bias: all cohort studies had methodological problems which could introduce confounding or bias. However, as the GRADE approach automatically downgrades certainty by two levels for non‐randomized studies, we did not downgrade further.
7Downgraded by one level for serious imprecision: the 95% confidence interval includes both clinically important effects and no effect.
8Downgrade by one level for serious inconsistency: although there was no substantial difference between drugs in the cohort studies, the proportion of discontinuations was higher with both drugs: 14% for mefloquine and 9% for doxycycline.
9Downgraded by one level for indirectness: the primary analysis included only cohort studies in longer‐term occupation travellers (USA Peace Corps volunteers) and military personnel. Adverse effects in shorter‐term international travellers may be lower.
10No serious imprecision: the effect was statistically significant and the overall data (RCTs and cohort studies) were adequately powered to detect this effect.
11Downgraded by one level for serious inconsistency: there was heterogeneity between trials in the direction of effect.

Figuras y tablas -
Summary of findings 2. Mefloquine versus doxycycline for preventing malaria in travellers
Table 1. Risk of bias assessment methods for cohort studies

Bias

Authors' judgement

Support for judgement

Confounding

Low risk

Moderate risk

Serious risk

Critical risk

No information

We used the following criteria:

Low risk: identified confounders were measured and were balanced across groups (age, sex, destination and duration of travel)

Moderate risk: identified confounders were measured and not balanced across groups, or several confounders had not been measured or not reported across groups

Serious risk: a critical confounder has been measured and is not balanced across groups

Selection of participants into the study

Low risk

Moderate risk

Serious risk

Critical risk

No information

We assessed whether selection into the study was unrelated to intervention or unrelated to outcome, and whether start of intervention and start of follow up coincided for most subjects. Non‐responder bias at the point of selection was considered here for cohort studies. We used the following cut offs for non‐response rate: low risk < 10%, moderate risk 10% to 20%, serious risk > 20%.

Measurement of interventions

Low risk

Moderate risk

Serious risk

Critical risk

No information

We used the following criteria:

Low risk: the prescription was provided by a travel clinic which also performed the study, and discontinuations were recorded and reported, or all participants were issued with their medication e.g. soldiers or participants were asked to self‐report which medication they took whilst they were taking it.

Moderate risk: the prescription was provided by a travel clinic which also performed the study but no information regarding switches and discontinuations was available or patients are asked to self‐report which prophylaxis they took shortly after they finished taking it.

Serious risk: Participants were asked to self‐report which prophylaxis they took a long time after they finished taking it.

Departures from intended interventions

Low risk

Moderate risk

Serious risk

Critical risk

No information

We assessed whether switches between interventions of interest were available. We assessed whether discontinuations and switches between prophylactic regimens had been recorded and reported.

Missing data

Low risk

Moderate risk

Serious risk

Critical risk

No information

We assessed whether outcome data was reasonably complete for most participants. We recorded missing data for included participants e.g. loss to follow up rates and treatment withdrawals.

Measurement of outcomes

Low risk

Moderate risk

Serious risk

Critical risk

No information

We assessed whether the outcome measure was objective or subjective. We assessed whether participants or study personnel were blinded to the intervention received. We assessed whether the methods of outcome assessment were comparable across intervention groups.

Selection of the reported result

Low risk

Moderate risk

Serious risk

Critical risk

No information

We used the following criteria:

Low risk: If the questionnaire was provided in full, or it was clear what was asked within it.

Moderate risk: If it is unclear which questions are asked, or information was provided on aggregate.

Serious risk: If data captured within the questionnaire was clearly missing.

Other

Low risk

Moderate risk

Serious risk

Critical risk

No information

We reported the study sponsor. We classified the analysis of studies sponsored by pharmaceutical companies as independent of the sponsor when it was clearly stated that the sponsor had no input to the trial analysis.

Figuras y tablas -
Table 1. Risk of bias assessment methods for cohort studies
Table 2. Adverse events and adverse effects risk of bias assessment methods

Criterion

Assessment

Explanation

On conduct

Were harms pre‐defined using standardised or precise definitions?

Adequate

Inadequate

Unclear

We classified as 'adequate' if the study reported explicit definitions for adverse events and effects that allow for reproducible ascertainment e.g. what adverse events were being investigated and what constituted an “event”, what was defined as a serious or severe adverse event.

Was ascertainment technique adequately described?

Adequate

Inadequate

Unclear

We classified as 'adequate' if the study reported methods used to ascertain complications, including who ascertained, timing, and methods used.

Was monitoring active or passive?

Active

Passive

Unclear

We classified monitoring as 'active' when authors reviewed participants at set time points during treatment and enquired about symptoms.

Was data collection prospective or retrospective?

Prospective

Retrospective

Unclear

We classified as ‘prospective’ if data collection occurred during treatment, or ‘retrospective’ if data collection occurred following treatment.

For laboratory investigations or other tests

Was the number and timing of tests adequate?

Adequate

Inadequate

Unclear

We classified the number and timing of tests as 'adequate', when tests were taken at baseline and at least one time point during prophylaxis.

Adapted from Bukiwra 2014

Figuras y tablas -
Table 2. Adverse events and adverse effects risk of bias assessment methods
Table 3. Characteristics of included studies for efficacy

Study ID

Participants (immune status)

Number of randomised participants

Mefloquine dose

Drug comparisons of interest

Duration of exposure to malaria

Country of malaria exposure

Local drug resistance

Bunnag 1992

Thai male adults (presumed semi‐immune)

605

250 mg weekly for first 4 weeks, then 125 mg weekly

Placebo

24 weeks (trial duration)

Thailand

Chloroquine, sulphadoxine‐pyrimethamine and quinine resistance

Nosten 1994

Pregnant women from the Thai‐Burma border (presumed semi‐immune)

339

250 mg weekly for first 4 weeks, then 125 mg weekly until delivery

Placebo

Various in endemic area (monitored until delivery)

Thai‐Burma border

Not mentioned

Pearlman 1980

Thai residents aged 10 to 60 years (semi‐immune)

990

180 mg tablet weekly, 360 mg tablet weekly, 360 mg every 2 weeks with appropriate adjustments for children

Placebo

26 weeks

Thailand

Chloroquine resistant Plasmodium falciparum

Santos 1993

Brazilian civilians and soldiers aged 12 to 55 years (semi‐immune)

128

500 mg every 4 weeks, 250mg every 2 weeks

Placebo

17 weeks

Brazil

P falciparum resistant to chloroquine and “high prevalence of multiresistant Plasmodium falciparum transmission”

Sossouhounto 1995

Ivory Coast adult males (semi‐immune)

500

250 mg weekly for first 4 weeks, then 125 mg weekly

Placebo

20 weeks

Ivory C oast

Not mentioned

Ohrt 1997

Indonesian soldiers ('largely' non‐immune)

204

250 mg weekly

Placebo, doxycycline

'approximately 13 weeks'

Indonesia

Sulfadoxine‐pyrimethamine and chloroquine resistance

Weiss 1995

Kenyan children (semi‐immune)

169

125 mg weekly

Placebo (multivitamin), doxycycline, primaquine

11 weeks

Kenya

Not mentioned

Salako 1992

Nigerian adult males (semi‐immune)

567

250 mg weekly for first 4 weeks, then 125 mg weekly

Placebo, chloroquine

24 weeks (trial duration)

Nigeria

"...at the time of the trial, chloroquine resistance was not a problem"

Hale 2003

Ghanain adults (semi‐immune)

530

250 mg weekly

Placebo

12 weeks

Ghana

Not mentioned

Arthur 1990

USA soldiers (non‐immune)

270

250 mg weekly

Doxycycline

8 weeks

Thailand

Local chloroquine resistance

Boudreau 1991

Thai adult males (semi‐immune)

501

500 mg fortnightly

Chloroquine

14 weeks (trial duration)

Cambodia

Local chloroquine resistance

Steketee 1996

Pregnant Malawian residents (semi‐immune)

4220

250 mg weekly

Chloroquine

Various in endemic area (monitored until delivery)

Malawi

P falciparum resistant to chloroquine, documented sensitivity of P falciparum to mefloquine

Figuras y tablas -
Table 3. Characteristics of included studies for efficacy
Table 4. Mefloquine versus placebo/no treatment; characteristics of included studies for safety

Study ID

Participants

Number enrolled

Method of adverse event monitoring

Exclusions for psychiatric adverse effects

Trial duration

Source of funding

RCTs

Bunnag 1992

Thai male adults

605

Interview with study personnel

None

24 weeks

Roche

Davis 1996

Australian adults who did not travel

106

Daily self‐reported diary

Past history of psychiatric conditions

7 weeks

Roche

Hale 2003

Ghanain adults

530

Interview with study personnel

History of neuropsychiatric illness

12 weeks

USA Army

Nosten 1994

Pregnant women, Thai‐Burma border

339

Phase 1: weekly symptom questionnaire. Babies were assessed at birth and at 3, 6, 12, and 24 months.

Phase 2: weekly symptom questionnaire. Babies were assessed at birth and at 2 and 9 months

None

Various

Government funding

Ohrt 1997

Indonesian soldiers

204

Two symptom questionnaires. Daily interview with study personnel

History of underlying illness

13 weeks

Roche, Pfizer, USA Army

Pearlman 1980

Thai residents aged 10 to 60 years

990

Weekly sick call by study personnel

None

26 weeks

Not mentioned

Potasman 2002

Israeli adults who did not travel

90

Self‐reporting diary

History of depression

48 hours

Mepha Ltd

Salako 1992

Nigerian adult males

567

Interview with study personnel

None

24 weeks

Not mentioned

Santos 1993

Brazilian civilians and soldiers aged 12 to 55

128

Interview w ith study personnel

None

17 weeks

Roche

Schlagenhauf 1997

Swissair trainee pilots who did not travel

23

Interview with study personnel

Psychosis or severe depression

4 weeks

Roche

Sossouhounto 1995

Ivory C oast adult males

500

Access to the village health centre

None

20 weeks

Not mentioned

Vuurman 1996

Dutch adult who did not travel

42

Interview with study personnel

H istory of any serious psychiatric disorder; evidence of drug or alcohol abuse

30 days

Roche

Weiss 1995

Kenyan children

169

Interview with study personnel

None

4 months

USA Army

Cohort studies

Participants

Number enrolled

Method of adverse event monitoring

Factors influencing drug allocation

Duration of travel

Source of funding

Hoebe 1997

Danish travellers

300

Telephone interview

Allocation based on guidelines and patient preference

Mean 3 weeks, range 1 to 9 weeks

Not mentioned

Petersen 2000

Danish travellers

4154

Participant self‐reported questionnaire

Allocation based on guidelines and patient preference

Various, not specified

Not mentioned

Rietz 2002

Swedish travellers

491

Participant self‐reported questionnaire

Allocation based on guidelines and patient preference

" Most", range 2 to 4 weeks

Not mentioned

van Riemsdijk 1997

Danish travellers

1501

Participant self‐reported questionnaire

Allocation based on guidelines and patient preference

Mean = 23 days

Not mentioned

Wells 2006

USA soldiers

397,442

Restrospective analysis of hospital records

No information available

Minimum 1 month

Government funding

Figuras y tablas -
Table 4. Mefloquine versus placebo/no treatment; characteristics of included studies for safety
Table 5. Mefloquine versus placebo/no treatment; quality of adverse events reporting

Study ID

Description of how adverse outcomes were defined and recorded¹

Description of ascertainment technique²

Active or passive monitoring?

Prospective or retrospective data collection?

Bunnag 1992

Inadequate

Comment: No definition of adverse events or effects was provided, it is unclear whether or how causality was assessed

Adequate

Active

Prospective

Davis 1996

Adequate

Adequate

Active

Prospective

Hale 2003

Inadequate

Comment: ‘serious’ adverse events were not defined, and methods for determining causality not described

Adequate

Active

Prospective

Nosten 1994

Inadequate

Comment: It is unclear what questions were included within the questionnaire and whether and how causality was assessed. ‘Serious’ adverse effects not defined

Adequate

Active

Prospective

Ohrt 1997

Inadequate

Comment: No definition of adverse events or effects provided, it was unclear whether or how causality was assessed

Adequate

Active

Prospective

Pearlman 1980

Inadequate

Comment: No definition of adverse events or effects was provided, it was unclear whether or how causality was assessed

Inadequate

Comment: Weekly sick call for all villagers

Passive

Prospective

Potasman 2002

Inadequate

Comment: No definition of adverse events or effects was provided, it was unclear whether or how causality was assessed

Adequate

Active

Prospective

Salako 1992

Inadequate

Comment: No definition of adverse events or effects was provided, it was unclear whether or how causality was assessed

Adequate

Active

Prospective

Santos 1993

Inadequate

Comment: No information given in the methods section on definition of adverse outcomes

Inadequate

Comment: No description of ascertainment method

Active

Prospective

Schlagenhauf 1997

Inadequate

Comment: No definition of adverse events or effects was provided, it was unclear whether or how causality was assessed

Adequate

Active

Prospective

Sossouhounto 1995

Inadequate

Comment: No definitions of adverse events or effects were provided, it was unclear whether or how causality was assessed

Unclear

Passive

Prospective

Vuurman 1996

Adequate

Unclear

Active

Prospective

Weiss 1995

Inadequate

Comment: No definitions of adverse events or effects were provided, it was unclear whether or how causality was assessed.

Adequate

Active

Prospective

Cohort studies

Hoebe 1997

Adequate

Adequate

Active

Retrospective

Petersen 2000

Adequate

Adequate

Active

Retrospective

Rietz 2002

Adequate

Adequate

Active

Unclear

'Filled in after their return'

Steffen 1993

Adequate

Adequate

Passive

Unclear

Comment: information was collected during the flight home, when travellers should still have been taking their prophylactic regimen

van Riemsdijk 1997

Adequate

Adequate

Active

Prospective

Wells 2006

Adequate

Adequate

Passive

Retrospective

1. Were harms pre‐defined using standardised or precise definitions?

2. Was ascertainment technique adequately described?

Figuras y tablas -
Table 5. Mefloquine versus placebo/no treatment; quality of adverse events reporting
Table 6. Serious adverse events; mefloquine versus comparators

Study ID

Study design

Mefloquine users

Drug comparators

Events/ participants

Description

Drug

Events/ participants

Description

Events (not attributed by study authors or participants to the drug regimen)

Bunnag 1992

RCT

0/116

Placebo

1/121

None provided

Nosten 1994

RCT

1/159 (women)

One death

  • Septic shock after an emergency caesarean section

Four congenital malformations:

  • Limb dysplasia (1 case), ventricular septal defect (2 cases), amniotic bands (1 case)

Placebo

0/152 (women)

One congenital malformation:

  • anencephaly

Sossouhounto 1995

RCT

0/103

Placebo

1/96

One death (not described)

Ohrt 1997

RCT

0/61

Placebo

0/65

Doxycycline

1/62

Acute hysteria¹

Lobel 2001

Cohort study

8/3703

8 hospitalisations

  • for "fainting, gastrointestinal symptoms, rashes, headaches, ophthalmologic symptoms, and fever"

Doxycycline

0/69

Chloroquine

0/119

Overbosch 2001

RCT

10/483

"...infectious illnesses in 7 subjects and breast cancer, anaphylaxis, or fractured femur in 1 subject each"

Atovaquone‐proguanil

4/493

"...infectious illnesses in 3 subjects and cerebral ischemia in 1 subject"

Studies reporting no serious events or effects

Salako 1992

RCT

0/107

"Adverse events were all mild and there were no deaths"

Placebo

Chloroquine

0/101

0/103

Arthur 1990

RCT

0/134

"No serious side effects occurred with either drug regimen"

Doxycycline

0/119

Schlagenhauf 2003

RCT

0/153

"Although a large number of adverse events were reported, none were serious"

Doxycycline

Atovaquone‐proguanil

0/153

0/164

Sonmez 2005

Cohort study

0/228

"No drug induced side effects necessitating emergency care were observed"

Doxycycline

0/506

Andersson 2008

Cohort study

0/491

"No serious adverse events were recorded"

Atovaquone‐proguanil

0/161

Napoletano 2007

Cohort study

0/548

Records hospitalisations, and reports that none occurred in either group of participants

Atovaquone‐proguanil

Chloroquine

0/707

0/37

Sossouhounto 1995

RCT

0/103

"All side effects were transient (and)... mild"

Chloroquine

0/100

1 This trial described a potentially serious adverse event, but did not provide enough detail to meet our definition.

Figuras y tablas -
Table 6. Serious adverse events; mefloquine versus comparators
Table 7. Serious adverse effects; mefloquine versus comparators

Study ID

Study design

Mefloquine users

Drug comparators

Events/ participants

Description

Drug

Events/ participants

Description

Effects (attributed by study authors or participants to the drug regimen)

Hoebe 1997

Cohort study

2/104

Two "serious acute adverse reactions"¹

  • Depressed mood

  • Dizziness

No treatment

0/93

Petersen 2000

Cohort study

5/809

5 hospitalisations:

  • Depressed mood

  • Depressed mood

  • Depressed mood, "strange thoughts"

  • Depressed mood, "strange thoughts", itching, vertigo

  • Vertigo, fever, mouth ulcers, diarrhoea

Chloroquine

6/1223

2 hospitalisations:

  • Blurred vision, nausea, headache, general skin itching, paraesthesia

  • Depressed mood

No treatment

0/161

Korhonen 2007

Cohort study

15/1612

15 hospitalisations:

  • Dizziness (3)

  • Heart palpitations (2)

  • Limb numbness (1)

  • Abdominal pain (1)

  • Yeast infection (1)

  • Anxiety and depression (1)

  • Visual disturbance, photosensitivity (1)

  • Passing out, extreme fatigue (1)

  • "Went crazy", anxiety, nausea, vomiting (1)

  • "Psychotic reaction", anxiety, abnormal dreams (1)

  • Anxiety, abnormal dreams, insomnia, unsteadiness (1)

  • Nausea, dizziness, blackout (1)

Doxycycline

9/708

9 hospitalisations:

  • Gastrointestinal disturbance (6)

  • Photosensitivity (1),

  • Coughing (1)

  • Anaemia (1)

Atovaquone‐proguanil

0/72

Chloroquine

4/832

4 hospitalisations:

  • Nausea, dizziness, visual disturbance, insomnia, abnormal dreams, unsteadiness, weakness

  • Abnormal dreams

  • Seizures

  • Abdominal pain, diarrhoea

Philips 1996

Cohortstudy

4/285

3 hospitalisations with "either gastrointestinal or neurologic symptoms" and one seizure

Doxycycline

1/383

Severe oesophagitis

Steketee 1996

RCT

1/?

One "neuropsychiatric side effect"

  • Disorientation to time and place¹

Chloroquine

0/?

Albright 2002

Cohort study

1/115

One "serious side effect"¹

  • Hallucinations

Chloroquine

0/22

Corominas 1997

Cohort study

1/609

One hospitalisation:

  • Heart palpitations, convulsions, paraesthesia and vertigo

Chloroquine

0/137

Steffen 1993

Cohort study

7/52981

7 hospitalisations, including:

  • Seizures (2)

  • Psychosis (2)

  • Vertigo (1)

  • 2 not characterised

Chloroquine

7/20332

7 hospitalisations. 'Includes':

  • Seizures (2)

  • Psychosis (1)

  • 4 not characterised

Studies reporting no serious events or effects

Hale 2003

RCT

0/46

Nine serious adverse events in the trial (trial arm not specified) "none of which were considered by study physicians to be related to the study drug"

Placebo

0/94

Salako 1992

RCT

0/107

"Adverse events were all mild and there were no deaths"

Placebo

Chloroquine

0/101

0/103

Arthur 1990

RCT

0/134

"No serious side effects occurred with either drug regimen"

Doxycycline

0/119

Schlagenhauf 2003

RCT

0/153

"Although a large number of adverse events were reported, none were serious"

Doxycycline

Atovaquone‐proguanil

0/153

0/164

Sonmez 2005

Cohort study

0/228

"No drug induced side effects necessitating emergency care were observed"

Doxycycline

0/506

Andersson 2008

Cohort study

0/491

"No serious adverse events were recorded"

Atovaquone‐proguanil

0/161

Napoletano 2007

Cohort study

0/548

Records hospitalisations, and reports that none occurred in either group of participants

Atovaquone‐proguanil

Chloroquine

0/707

0/37

Sossouhounto 1995

RCT

0/103

"All side effects were transient (and)... mild"

Chloroquine

0/100

¹ This trial described a potentially serious adverse effect, but did not provide enough detail to meet our strict definition.

Figuras y tablas -
Table 7. Serious adverse effects; mefloquine versus comparators
Table 8. Mefloquine versus doxycycline; characteristics of included studies for safety

Study ID

Participants

Number enrolled

Method of adverse event monitoring

Significant exclusions for psychiatric adverse effects

Duration of travel

Source of funding

Randomized controlled trials

Arthur 1990

USA soldiers

270

Blood tests, stool samples. Interview with study personnel

None

5 weeks

Not mentioned

Ohrt 1997

Indonesian soldiers

204

Interview with study personnel. Exit questionnaire

" History of underlying illness"

13 weeks

Pfizer and Roche

Schlagenhauf 2003

Non‐immune adult short‐term travellers

674

Participant self‐reported questionnaire

History of seizures or psychiatric disorders

4 to 6 weeks

GlaxoSmithKline and Roche

Weiss 1995

Kenyan children

169

Interview with study personnel

None

4 months

Government funding

Non‐randomized studies

Participants

Number enrolled

Method of adverse event monitoring

Factors influencing drug allocation

Duration of travel

Source of funding

Cunningham 2014

UK Foreign and Commonwealth Office staff

327

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

0 to 36 months

Not mentioned

Eick‐Cost 2017

USA s oldiers

367,840

Data from the Defense Medical Surveillance System, the Pharmacy Data Transaction Service and the Theater Medical Data Store

No information available

Various, not specified

Not mentioned

Goodyer 2011

UK adult short‐term travellers

185

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

< 28 days

GlaxoSmithKline

Korhonen 2007

Peace Corps volunteers

2701

Participant self‐reported questionnaire

Allocation based on guidelines and participan t preference

≥ 6 months

Two staff employed by Peace Corps

Landman 2015

Peace Corps volunteers

1184

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, not specified

Not mentioned

Laver 2001

Adult short‐term travellers

660

Participant self‐reported questionnaire

No information available

93% < 4 weeks

" No financial interests to disclose"

Lobel 2001

Adult short‐term travellers

5626

Participant self‐reported questionnaire

No information available

< 5 weeks

" No financial interests to disclose"

Meier 2004

UK adults enrolled in UK g eneral p ractice research database

35,370

Incident cases of depression, psychoses and panic attacks within the UK general practice research database

No information available

Various, not specified

Roche

Napoletano 2007

Italian short‐term travellers

1906

Telephone interview

Allocation based on guidelines and participant preference

Mean 2 weeks, range 0 to > 35 days

Not mentioned

Philips 1996

Australian short‐term travellers

741

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, mean 3 weeks, maximum 3 months

Roche and Pfizer

Saunders 2015

USA soldiers

2351

Participant self‐reported questionnaire

Primarily doxycycline, soldiers with contra‐indications received mefloquine

> 90% for 10 months or more

Not mentioned

Schwartz 1999

Israeli short‐term travellers

158

Participant self‐reported questionnaire

"... daily doxycycline or daily primaquine... was recommended"

14 to 20 days

Not mentioned

Shamiss 1996

Israeli soldiers

45

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

"... an average of 4 hours stay in the field over a period of 2 months"

Not mentioned

Sharafeldin 2010

Dutch medical students

180

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Mean 74 days (range 10 to 224 days)

No dedicated funding

Sonmez 2005

Turkish soldiers

1400

Participant self‐reported questionnaire

Prior to March 2002: doxycyline

After July 2002: mefloquine

A pprox. 6 months

Not mentioned

Stoney 2016

USA short‐term travellers

370

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Median duration 13 days

Government funding

Tan 2017

Peace Corps volunteers

8931

Participant self‐reported questionnaire

No information available

Various, not specified

No dedicated funding

Terrell 2015

UK soldiers

2032

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Median duration 13 days

"... not funded by an external body"

Tuck 2016

UK soldiers

151

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, not specified

No dedicated funding

Waner 1999

Adult short‐term travellers

3051

Participant self‐reported questionnaire

No information available

A pprox. 6 weeks

Not mentioned

Figuras y tablas -
Table 8. Mefloquine versus doxycycline; characteristics of included studies for safety
Table 9. Mefloquine versus doxycycline; quality of adverse event reporting

Study ID

Harms predefined¹

Description of ascertainment technique²

Active or passive monitoring?³

Prospective or retrospective data collection?

RCTs

Arthur 1990

Inadequate:

No definitions provided for serious side effects

Unclear: it is not reported who conducted the interviews

Active

Prospective

Ohrt 1997

Inadequate

Comment: No definitions of adverse events or effects were provided, it wa s unclear whether or how causality was assessed

Adequate

Active

Prospective

Schlagenhauf 2003

Adequate

Adequate

Active

Prospective

Weiss 1995

Inadequate

" Each subject was visited daily at home by an assigned field worker, who asked about symptoms of malaria or drug side effects"

Adequate

Active

Prospective

Cohort studies

Cunningham 2014

Inadequate

Comment: questionnaire included a targeted list of side effects, including " other psychological problems" . What was included within this was not defined

Adequate

Passive

Unclear

Comment: questionnaire was performed while participants were still taking chemoprophylaxis medication, although 75% were non‐compliant

Eick‐Cost 2017

Adequate

Adequate

Passive

Prospective

Goodyer 2011

Inadequate

" Also included on the questionnaire was a single free‐text question asking travellers to describe any side effects of antimalarial medication"

Adequate

Active

Retrospective

Korhonen 2007

Adequate

Adequate

Passive

Unclear

Comment: n o information wa s provided regarding the timing of the questionnaire during treatment

Landman 2015

Adequate

Adequate

Passive

Unclear

Comment: all participants were emailed the questionnaire at one time point, which occurred at varying points during the prophylactic regimen

Lobel 2001

Inadequate

"Travellers… were given a questionnaire that asked for... adverse health events attributed to those drugs"

Adequate

Passive

Unclear

Comment: information was collected at the airport, when travellers should still have been taking the prophylactic regimen

Meier 2004

Adequate

Adequate

Passive

Retrospective

Napoletano 2007

Unclear

Comment: adverse events were categorised on a scale of one to four, but it is unclear whether and how causality was assessed

Adequate

Active

Retrospective

Philips 1996

Inadequate

Comment: it wa s unclear what constituted a serious or severe event and insufficient information on the questions that travellers were asked

Inadequate

"... a mailed questionnaire approximately 2 weeks after their anticipated return home date’ ‘if a reply had not been received within 4 weeks an abbreviated questionnaire was sent out."

Comment: no details provided regarding abbreviated questionnaire

Active

Retrospective

Saunders 2015

Inadequate

Comment: insufficient information of the questions that travellers were asked

Adequate

Passive

Retrospective

Schwartz 1999

Inadequate

"... we directly contacted all travelers for complete follow‐up and assessment of compliance. Fifty travelers taking primaquine completed a questionnaire regarding side effects"

Inadequate

Comment: see quote. Different methods of follow up for different forms of prophylaxis

Unclear

Unclear

Shamiss 1996

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Inadequate

" Questionnaires were distributed and collected by the flight surgeon to 45 aircrew…questionnaires were immediately evaluated and further data collection was done by telephone, if necessary"

Passive

Unclear

Comment: it wa s unclear at which time point data collection occurred

Sharafeldin 2010

Inadequate

Comment: n o information wa s provided on how information on adverse effects was sought

Inadequate

Comment: n o mention of how adverse events were recorded in the questionnaire

Passive

Retrospective

Sonmez 2005

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Adequate

Active

Prospective

Stoney 2016

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Inadequate

Comment: n o information is reported on how adverse events were ascertained

Active

Prospective

Tan 2017

Adequate

Adequate

Active

Retrospective

Terrell 2015

Inadequate

" The questionnaire approved by the MODREC included the 19 commonest adverse effects described in the manufacturers’ product documentation"

Comment: Adverse events listed in the questionnaire are not reported

Adequate

Passive

Unclear

Comment: information obtained during transit through Nairobi back to the UK. It wa s unclear whether participants were still taking prophylaxis at this time point

Tuck 2016

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Adequate

Active

Unclear

Comment: i t wa s not specified at which point during treatment the questionnaire was administered

Waner 1999

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Adequate

Passive

Unclear

Comment: information was collected during the flight home, when travellers should still have been taking their prophylactic regimen

1. Were harms pre‐defined using standardised or precise definitions?

2. Was ascertainment technique adequately described?

3. Monitoring classed as 'active' if it occurred at set time points during treatment.

For full description of analysis methods, see Table 2.

Figuras y tablas -
Table 9. Mefloquine versus doxycycline; quality of adverse event reporting
Table 10. Mefloquine versus atovaquone‐proguanil; characteristics of included studies for safety

Study ID

Participants

Number enrolled

Method of adverse event monitoring

Significant exclusions for psychiatric adverse effects

Duration of travel

Source of funding

Randomized controlled trials

Overbosch 2001

Travellers from Canada, Germany, Netherlands, South Africa, UK

1013

Interview with study personnel

"... history of alcoholism, seizures or psychiatric or severe neurological disorders"

Mean 2.5 weeks

GlaxoSmithKline

Schlagenhauf 2003

Non‐immune adult short‐term travellers

674

Participant self‐reported questionnaire

" History of seizures or psychiatric disorders"

4 to 6 weeks

GlaxoSmithKline and Roche

van Riemsdijk 2002

Dutch short‐term travellers

140

Interview and testing with study personnel

"H istory of alcoholism, seizures, psychiatric disorders, severe neurological disorders"

Mean 19 days

Government funding

Non‐randomis ed studies

Participants

Number enrolled

Method of adverse event monitoring

Factors influencing drug allocation

Duration of travel

Source of funding

Andersson 2008

Swedish soldiers

609

Participant self‐reported questionnaire

Mainly mefloquine, soldiers with contra‐indications received atovaquone‐proguanil

6 months

Not mentioned

Belderok 2013

Dutch short‐term travellers

945

Participant self‐reported questionnaire (measured adherence)

Allocation based on guidelines and participant preference

84% < 29 days

Government funding

Cunningham 2014

UK Foreign and Commonwealth Office staff

327

Participant self‐reported questionnaire

Allocation based on guidelines and p articipant preference

0‐36 months

Not mentioned

Eick‐Cost 2017

USA s oldiers

367,840

Data from the Defense Medical Surveillance System, the Pharmacy Data Transaction Service and the Theater Medical Data Store

No information available

Various, not specified

Not mentioned

Goodyer 2011

UK adult short‐term travellers

185

Participant self‐reported questionnaire

Allocation based on guidelines and p articipant preference

< 28 days

GlaxoSmithKline

Kato 2013

Japanese short‐term travellers

316

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Mean 20.0 ± 9.6 days in the atovaquone‐proguanil group and 59.0 ± 15.9 days in the mefloquine group

Not mentioned

Korhonen 2007

Peace Corps volunteers

2701

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

≥ 6 months

Two staff employed by Peace Corps

Kuhner 2005

German short‐term travellers

495

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

A tovaquone‐proguanil mean 2.6 weeks, mefloquine mean 7 weeks

Not mentioned

Landman 2015

Peace Corps volunteers

1184

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, not specified

Not mentioned

Laverone 2006

Italian short‐term travellers

1176

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

> 90% 0 to 30 days

Not mentioned

Napoletano 2007

Italian short‐term travellers

1906

Telephone interview

Allocation based on guidelines and participant preference

Mean 2 weeks, range 0 to > 35 days

Not mentioned

Schneider 2013

UK adults enrolled in UK g eneral p ractice research database

Not available

Incident cases of a neuropsychiatric disorders during or after antimalarial drug use

No information available

Various, not specified

Roche

Sharafeldin 2010

Dutch medical students

180

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Mean duration of stay 74 days (range 10 to 224 days)

" N o dedicated funding for this project"

Stoney 2016

USA short‐term travellers

370

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Median duration 13 days

Government funding

Tan 2017

Peace Corps volunteers

8931

Participant self‐reported questionnaire

No information available

Various, not specified

No dedicated funding

Tuck 2016

UK soldiers

151

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, not specified

No dedicated funding

Figuras y tablas -
Table 10. Mefloquine versus atovaquone‐proguanil; characteristics of included studies for safety
Table 11. Mefloquine versus atovaquone‐proguanil; quality of adverse event reporting

Study ID

Harms predefined¹

Description of ascertainment technique²

Active or passive monitoring?³

Prospective or retrospective data collection?

RCTs

Overbosch 2001

Adequate

Adequate

Active

Prospective

Schlagenhauf 2003

Adequate

Adequate

Active

Prospective

van Riemsdijk 2002

Adequate

Adequate

Active

Prospective

Cohort studies

Andersson 2008

Inadequate

Comment: insufficient information provided on the questions which soldiers were asked

Inadequate

Comment: different ascertainment technique used for one of the three groups, which is inadequately described

Active

Unclear

Comment: d ata collection was prospective for 448/609 participants (LA04 and LA05), but retrospective for 161 participants (LA02)

Cunningham 2014

Inadequate

Comment: questionnaire included a targeted list of side effects, including " other psychological problems" . What was included within this was not defined

Adequate

Passive

Unclear

Comment: questionnaire was performed while participants were still taking chemoprophylaxis medication, although 75% were non‐compliant

Eick‐Cost 2017

Adequate

Adequate

Passive

Prospective

Goodyer 2011

Inadequate

" Also included on the questionnaire was a single free‐text question asking travelers to describe any side effects of antimalarial medication"

Adequate

Active

Retrospective

Kato 2013

Adequate

Adequate

Passive

Unclear

Comment: the timing of this questionnaire has not been made clear

Korhonen 2007

Adequate

Adequate

Passive

Unclear

Comment: n o information wa s provided regarding the timing of the questionnaire during treatment

Kuhner 2005

Inadequate

Comment: insufficient information provided on the questions that participants were asked

Adequate

Active

Retrospective

Landman 2015

Adequate

Adequate

Passive

Unclear

Comment: all participants were emailed the questionnaire at one time point, which occurred at varying points during the prophylactic regimen

Laverone 2006

Adequate

Adequate

Passive

Retrospective

Napoletano 2007

Unclear

Comment: adverse events were categorised on a scale of one to four, but it is unclear whether and how causality was assessed

Adequate

Active

Retrospective

Schneider 2013

Adequate

Adequate

Passive

Retrospective

Sharafeldin 2010

Inadequate

Comment: n o information is provided on how information on adverse effects was sought

Inadequate

Comment: n o mention of how adverse events were recorded in the questionnaire.

Passive

Retrospective

Stoney 2016

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Inadequate

Comment: n o information is reported on how adverse events were ascertained

Active

Prospective

Tan 2017

Adequate

Adequate

Active

Retrospective

Tuck 2016

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Adequate

Active

Unclear

Comment: i t wa s not specified at which point during treatment the questionnaire was administered

1. Were harms pre‐defined using standardised or precise definitions?

2. Was ascertainment technique adequately described?

3. Monitoring classed as 'active' if it occurred at set time points during treatment.

For full description of analysis methods, see Table 2.

Figuras y tablas -
Table 11. Mefloquine versus atovaquone‐proguanil; quality of adverse event reporting
Table 12. Mefloquine versus chloroquine; characteristics of included studies for safety

Study ID

Participants

Number enrolled

Method of adverse event monitoring

Significant exclusions for psychiatric side effects

Trial duration

Source of funding

RCT s

Boudreau 1991

Thai gem miners

501

Interview with study personnel

None

14 weeks

USA Army

Boudreau 1993

USA soldiers

359

Interview with study personnel and computerised questionnaire

"M edical history of psychiatric or neurological problems within the last 5 years"

13 weeks

Not mentioned

Bunnag 1992

Thai adult mal es

605

Interview with study personnel

None

24 weeks

Roche

Salako 1992

Nigerian adult males

567

Interview with study personnel

None

24 weeks

Not mentioned

Sossouhounto 1995

Ivory C oast adult males

500

" Access to the village health centre. Clinical examination with study personnel"

None

20 weeks

Not mentioned

Steketee 1996

Pregnant Malawian women

4220

Interview with study personnel

None

Monitored from enrolment to delivery

Government funding

Non‐randomised studies

Participants

Number enrolled

Method of adverse event monitoring

Factors influencing drug allocation

Duration of travel

Source of funding

Albright 2002

USA travelling children aged < 13 years

177

Interview with study personnel

Allocation based on guidelines and participant preference

Various, not specified

Not mentioned

Corominas 1997

Spanish short‐term adult travellers

1054

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Maximum 6 weeks

Not mentioned

Cunningham 2014

UK Foreign and Commonwealth Office staff

327

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

0 to 36 months

Not mentioned

Hill 2000

USA short‐term travellers

822

Interview with study personnel

Allocation based on guidelines and participant preference

Median 19 days, up to 90 days

Not mentioned

Korhonen 2007

Peace Corps volunteers

2701

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

≥ 6 months

Two staff employed by Peace Corps

Laver 2001

Adult short‐term travellers

660

Participant self‐reported questionnaire

No information available

93% < 4 weeks

" No financial interests to disclose"

Laverone 2006

Italian short‐term travellers

1176

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

> 90% 0 to 30 days

Not mentioned

Lobel 2001

Adult short‐term travellers

5626

Participant self‐reported questionnaire

No information available

M ost < 5 weeks

" No financial interests to disclose"

Napoletano 2007

Italian short‐term travellers

1906

Telephone interview

Allocation based on guidelines and participant preference

Mean 2 weeks, range 0 to > 35 days

Not mentioned

Petersen 2000

Danish travellers

4154

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Various, 65% < 3 weeks

Not mentioned

Rietz 2002

Swedish short‐term travellers

491

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

" Most" 2 to 4 weeks

Not mentioned

Steffen 1993

Adult short‐term travellers

145,003

Participant self‐reported questionnaire

No information available

98% stayed between 1 and 4 weeks

Roche

Stoney 2016

USA short‐term travellers

370

Participant self‐reported questionnaire

Allocation based on guidelines and participant preference

Median duration 13 days

Government funding

Tan 2017

Peace Corps volunteers

8931

Participant self‐reported questionnaire

No information available

Various, not specified

No dedicated funding

Waner 1999

Adult short‐term travellers

3051

Participant self‐reported questionnaire

No information available

A pprox. 6 weeks

" not funded by an external body"

Figuras y tablas -
Table 12. Mefloquine versus chloroquine; characteristics of included studies for safety
Table 13. Mefloquine versus chloroquine; quality of adverse events reporting

Study ID

Harms predefined¹

Description of ascertainment technique²

Active or passive monitoring?³

Prospective or retrospective data collection?

RCTs

Boudreau 1991

Adequate

Adequate

Active

Prospective

Boudreau 1993

Adequate

Adequate

Active

Prospective

Bunnag 1992

Inadequate

" Adverse events were defined clinically, and starting week 14, volunteers reporting adverse events were interviewed by members of the hospital team"

Adequate

Active

Prospective

Salako 1992

Inadequate

" Particular attention was paid to complaints such as fever, chills, malaise, nausea and vomiting, rashes and other symptoms and signs that could be regarded as adverse events."

Comment: no clear definition of adverse events wa s provided

Adequate

Active

Prospective

Sossouhounto 1995

Inadequate

" Participants had access to a village health center, where they could notify personnel of any malaise or side effects"

Unclear

" Clinical examinations and parasitologic tests were performed every 4 weeks"

Passive

Prospective

Steketee 1996

Adequate

Adequate

Active

Prospective

Cohort studies

Albright 2002

Adequate

Adequate

Passive

Retrospective

Corominas 1997

Inadequate

Comment: insufficient information wa s provided about the questions that travellers were asked

Adequate

Active

Retrospective

Cunningham 2014

Inadequate

Comment: questionnaire included a targeted list of side effects, including " other psychological problems" . What was included within this was not defined

Adequate

Passive

Unclear

Comment: questionnaire was performed while participants were still taking chemoprophylaxis medication, although 75% were non‐compliant

Hill 2000

Inadequate

Comment: insufficient information wa s provided about the questions that travellers were asked

Adequate

Active

Retrospective

Korhonen 2007

Adequate

Adequate

Passive

Unclear

Comment: No information wa s provided regarding the timing of the questionnaire during treatment

Laverone 2006

Adequate

Adequate

Passive

Retrospective

Lobel 2001

Inadequate

"Travellers… were given a questionnaire that asked for... adverse health events attributed to those drugs"

Adequate

Passive

Unclear

Comment: information was collected at the airport, when travellers should still have been taking the prophylactic regimen

Napoletano 2007

Unclear

Comment: adverse events were categorised on a scale of one to four, but it is unclear whether and how causality was assessed

Adequate

Active

Retrospective

Petersen 2000

Inadequate

Comment: i t wa s unclear whether the questionnaire implied causality to the drug regimen

Adequate

Active

Retrospective

Rietz 2002

Adequate

Adequate

Active

Retrospective

Steffen 1993

Adequate

Adequate

Passive

Unclear

Comment: information was collected during the flight home, when travellers should still have been taking the prophylactic regimen

Stoney 2016

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Inadequate

Comment: n o information wa s reported on how adverse events were ascertained

Active

Prospective

Tan 2017

Adequate

Adequate

Active

Retrospective

Waner 1999

Inadequate

Comment: insufficient information provided on the questions that travellers were asked

Adequate

Passive

Unclear

Comment: information was collected during the flight home, when travellers should still have been taking the prophylactic regimen

1. Were harms pre‐defined using standardised or precise definitions?

2. Was ascertainment technique adequately described?

3. Monitoring classed as 'active' if it occurred at set time points during treatment.

For full description of analysis methods, see Table 2.

Figuras y tablas -
Table 13. Mefloquine versus chloroquine; quality of adverse events reporting
Table 14. Mefloquine versus currently used regimens; by duration of travel

Mefloquine versus atovaquone‐proguanil and doxycycline

Outcome

Short‐ term travellers¹

Longer‐ term travellers²

Test for subgroup
differences

Relative effect (RR)
(95% CI)
Studies (participants)

Relative effect (RR)
(95% CI)
Studies (participants)

Serious adverse effects

RR 5.38

(0.60 to 47.84)

3 cohort studies (2657)

RR 0.93

(0.43 to 2.01)

3 cohort studies (3147)

P = 0.14

Discontinuations due to adverse effects (RCTs)

RR 2.64

(1.51 to 4.62)

5 RCTs (2048)

Discontinuations due to adverse effects (cohort studies)

RR 1.81

(0.86 to 3.80)

7 cohort studies (2907)

RR 1.19

(0.45 to 3.17)

4 cohort studies (5711)

P = 0.50

Nausea

RR 2.02

(0.87 to 4.68)

6 cohort studies (2469)

RR 0.96

(0.22 to 4.18)

3 cohort studies (2725)

P = 0.39

Abdominal pain

RR 0.66

(0.22 to 1.98)

5 cohort studies (1801)

RR 0.30

(0.22 to 0.42)

3 cohort studies (2725)

P = 0.18

Diarrhoea

RR 0.64

(0.15 to 2.71)

5 cohort studies (2428)

RR 0.57

(0.22 to 1.49)

4 cohort studies (5187)

P = 0.89

Headache

RR 2.39

(0.69 to 8.22)

5 cohort studies (2086)

RR 2.09

(1.10 to 3.95)

4 cohort studies (3506)

P = 0.85

Dizziness

RR 3.05

(1.15 to 8.12)

4 cohort studies (1067)

RR 3.84

(1.34 to 11.00)

4 cohort studies (3506)

P = 0.76

Abnormal dreams

RR 6.25

(1.16 to 33.67)

3 cohort studies (1037)

RR 7.62

(2.06 to 28.18)

4 cohort studies (3506)

P = 0.86

Insomnia

RR 3.09

(0.30 to 32.21)

4 cohort studies (1760)

RR 8.67

(4.73 to 15.89)

4 cohort studies (3506)

P = 0.40

Anxiety

RR 3.26

(0.20 to 53.46)

1 cohort study (487)

RR 18.05

(9.75 to 33.42)

3 cohort studies (2854)

P = 0.24

Depressed mood

RR 2.52

(0.76 to 8.29)

3 cohort studies (1026)

RR 12.59

(6.47 to 24.49)

3 cohort studies (3210)

P = 0.02

Abnormal thoughts and behaviours

RR 1.29

(0.07 to 22.44)

1 cohort study (487)

RR 7.78

(1.12 to 54.06)

2 cohort studies (2558)

P = 0.31

Adherence: during travel

RR 1.10

(1.03 to 1.18)

7 cohort studies (7241)

RR 1.20

(0.88 to 1.62)

4 cohort studies (4890)

P = 0.61

Adherence: after return

RR 1.04

(0.92 to 1.17)

4 cohort studies (1221)

1 Short‐ term travellers: Approximately 3 weeks (range 1 day to 3 months). References: Goodyer 2011; Kato 2013; Kuhner 2005; Napoletano 2007; Laver 2001; Laverone 2006; Lobel 2001; Philips 1996; Schwartz 1999; Shamiss 1996; Sonmez 2005; Stoney 2016; Terrell 2015
2 Longer‐ term travellers: Approximately 6 months (range 0 to 36 months in Cunningham 2014 . Otherwise 3 months or longer). References Andersson 2008; Cunningham 2014; Korhonen 2007; Landman 2015; Saunders 2015; Sharafeldin 2010

Figuras y tablas -
Table 14. Mefloquine versus currently used regimens; by duration of travel
Table 15. Mefloquine versus currently used regimens; by military or non‐military participants

Mefloquine versus atovaquone‐proguanil and doxycycline

Outcome

Military¹

Non‐military²

Test for subgroup
differences

Relative effect (RR)
(95% CI)
Studies (participants)

Relative effect (RR)
(95% CI)
Studies (participants)

Serious adverse effects

0 events in 1386 participants

RR 1.21

(0.60 to 2.44)

4 cohort studies (4418)

Discontinuations due to adverse effects (RCTs)

RR 2.08

(0.13 to 32.73)

2 RCTs (441)

RR 2.22

(1.17 to 4.21)

4 RCTs (1669)

P = 0.96

Discontinuations due to adverse effects (cohorts)

RR 1.24

(0.32 to 4.88)

4 cohort studies (3408)

RR 1.89

(1.35 to 2.64)

8 cohort studies (8938)

P = 0.56

Nausea

RR 1.39

(0.36 to 5.36)

4 cohort studies (1578)

RR 1.70

(0.60 to 4.81)

6 cohort studies (3767)

P = 0.26

Abdominal pain

RR 0.43

(0.14 to 1.29)

4 cohort studies (1578)

RR 0.56

(0.23 to 1.35)

5 cohort studies (3099)

P = 0.72

Diarrhoea

RR 0.30

(0.09 to 0.96)

4 cohort studies (3999)

RR 1.05

(0.54 to 2.06)

6 cohort studies (3767)

P = 0.07

Headache

RR 1.19

(0.14 to 9.79)

2 cohort studies (1386)

RR 2.48

(1.40 to 4.40)

7 cohort studies (4206)

P = 0.51

Dizziness

RR 2.95

(1.37 to 6.36)

3 cohort studies (844)

RR 3.58

(1.39 to 9.25)

6 cohort studies (3880)

P = 0.76

Abnormal dreams

RR 11.02

(4.61 to 26.34)

1 cohort study (652)

RR 6.59

(1.74 to 25.00)

6 cohort studies (3891)

P = 0.53

Insomnia

RR 2.34

(0.41 to 13.35)

3 cohort studies (1537)

RR 10.24

(6.26 to 16.76)

6 cohort studies (3880)

P = 0.11

Anxiety

RR 16.94

(9.36 to 30.64)

4 cohort studies (3390)

Depressed mood

RR 13.44

(3.34 to 54.05)

1 cohort study (652)

RR 6.49

(2.66 to 15.85)

5 cohort studies (3584)

P = 0.39

Abnormal thoughts and behaviours

RR 5.11

(1.11 to 23.53)

3 cohort studies (3045)

Adherence: during travel

RR 1.18

(1.00 to 1.40)

5 cohort studies (4652)

RR 1.16

(0.99 to 1.35)

8 cohort studies (10785)

P = 0.85

Adherence: after return

RR 1.16

(0.86 to 1.55)

1 cohort study (43)

RR 1.02

(0.89 to 1.16)

3 cohort studies (1178)

P = 0.44

Figuras y tablas -
Table 15. Mefloquine versus currently used regimens; by military or non‐military participants
Comparison 1. Mefloquine versus placebo/non users

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria Show forest plot

9

1908

Risk Ratio (M‐H, Random, 95% CI)

0.09 [0.04, 0.19]

2 Malaria; episodes of parasitaemia in semi‐immune populations Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Trials reporting number of participants with parasitaemia

3

414

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.06, 0.55]

2.2 Trials reporting number of episodes of parasitaemia

2

510

Risk Ratio (M‐H, Random, 95% CI)

0.05 [0.00, 5.25]

3 Serious adverse events or effects (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 RCTs (adverse events)

6

1221

Risk Ratio (M‐H, Fixed, 95% CI)

0.70 [0.14, 3.53]

3.2 Cohort studies (adverse effects)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

3.08 [0.39, 24.11]

4 Discontinuations due to adverse effects (all studies) Show forest plot

7

1130

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.55, 4.88]

4.1 RCTs (adverse effects)

7

1130

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.55, 4.88]

5 Nausea (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 RCTs (adverse events)

2

244

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [1.05, 1.73]

5.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Fixed, 95% CI)

1.85 [1.42, 2.43]

6 Vomiting (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.50, 1.19]

6.2 Cohort studies (adverse events)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.74 [0.45, 1.21]

7 Abdominal pain (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 RCTs (adverse events)

3

550

Risk Ratio (M‐H, Fixed, 95% CI)

1.09 [0.84, 1.40]

7.2 Cohort studies (adverse events)

2

1167

Risk Ratio (M‐H, Fixed, 95% CI)

0.97 [0.66, 1.42]

8 Diarrhoea (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 RCTs (adverse events)

4

589

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.32, 1.62]

8.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Random, 95% CI)

1.25 [0.93, 1.68]

9 Headache (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

9.1 RCTs (adverse events)

5

791

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.71, 0.99]

9.2 Cohort studies (adverse events)

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

1.64 [0.63, 4.26]

10 Dizziness (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

10.1 RCTs (adverse events)

3

452

Risk Ratio (M‐H, Fixed, 95% CI)

1.03 [0.90, 1.17]

10.2 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Fixed, 95% CI)

1.80 [1.29, 2.49]

11 Abnormal dreams (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

11.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

2.35 [1.15, 4.80]

12 Insomnia (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

12.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

1.46 [1.06, 2.02]

13 Anxiety (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

13.1 Cohort studies (adverse events)

2

931

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.67, 2.21]

14 Depressed mood (all studies) Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

14.1 Cohort studies (adverse events)

3

1901

Risk Ratio (M‐H, Random, 95% CI)

2.43 [0.65, 9.07]

15 Abnormal thoughts and perceptions Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

15.1 Cohort studies (adverse events)

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

5.77 [0.79, 42.06]

16 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

16.1 RCTs (adverse events)

3

609

Risk Ratio (M‐H, Fixed, 95% CI)

0.86 [0.60, 1.24]

16.2 Cohort studies (adverse events)

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

6.71 [1.58, 28.55]

17 Visual impairment (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

17.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.98 [0.66, 1.46]

17.2 Cohort studies (adverse events)

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

0.93 [0.27, 3.19]

18 Vertigo (all studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

18.1 RCTs (adverse events)

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.78, 1.34]

19 Other adverse events (RCTs) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

19.1 Arthralgia

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.29 [0.02, 5.48]

19.2 Back pain

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.01, 1.61]

19.3 Blurred vision

1

208

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.01, 3.89]

19.4 Cough

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.71, 1.14]

19.5 Constipation

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

0.77 [0.53, 1.11]

19.6 Decreased appetite

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.10 [0.95, 1.28]

19.7 Falls

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.82, 1.43]

19.8 Fatigue

1

42

Risk Ratio (M‐H, Fixed, 95% CI)

0.91 [0.14, 5.86]

19.9 Gastritis

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.02 [0.10, 10.98]

19.10 Myalgia

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

1.53 [0.36, 6.57]

19.11 Rash

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.29 [0.04, 2.30]

19.12 Respiratory tract infection

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

2.63 [1.04, 6.61]

19.13 Sore throat

1

140

Risk Ratio (M‐H, Fixed, 95% CI)

0.34 [0.04, 2.75]

19.14 Unsteadiness

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.74, 1.52]

19.15 Weakness

1

202

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.96, 1.17]

20 Other adverse effects (cohort studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

20.1 Agitation

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.61, 1.82]

20.2 Altered spatial perception

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

9.4 [0.57, 153.97]

20.3 Confusion

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

0.67 [0.25, 1.78]

20.4 Loss of appetite

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.54, 1.50]

20.5 Mouth ulcers

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

1.00 [0.39, 2.56]

20.6 Palpitations

1

197

Risk Ratio (M‐H, Fixed, 95% CI)

8.06 [0.44, 147.68]

20.7 Tingling

1

970

Risk Ratio (M‐H, Fixed, 95% CI)

1.92 [0.59, 6.24]

Figuras y tablas -
Comparison 1. Mefloquine versus placebo/non users
Comparison 2. Mefloquine versus doxycycline

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

4

744

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.35, 5.19]

2 Serious adverse events or effects (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 RCTs (adverse events)

3

682

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.01, 8.16]

2.2 Cohort studies (adverse effects)

3

3722

Risk Ratio (M‐H, Random, 95% CI)

1.53 [0.23, 10.24]

3 Discontinuations due to adverse effects (all studies) Show forest plot

14

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RCTs

4

763

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.41, 2.87]

3.2 Cohort studies

10

10165

Risk Ratio (M‐H, Random, 95% CI)

0.92 [0.54, 1.55]

4 Nausea (all studies) Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 Cohort studies (adverse effects)

5

2683

Risk Ratio (M‐H, Fixed, 95% CI)

0.37 [0.30, 0.45]

4.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.71 [0.75, 9.74]

4.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

1.61 [1.06, 2.43]

5 Vomiting (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Cohort studies (adverse effects)

4

5071

Risk Ratio (M‐H, Fixed, 95% CI)

0.18 [0.12, 0.27]

5.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

6 Abdominal pain (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 Cohort studies (adverse effects)

4

2569

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.09, 1.07]

6.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

1.65 [0.74, 3.70]

6.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

1.34 [0.83, 2.18]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 Cohort studies (adverse effects)

5

5104

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.11, 0.73]

7.2 RCTs (adverse events)

2

376

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.78, 1.29]

7.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

3.58 [1.69, 7.59]

8 Dyspepsia (all studies) Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 Cohort studies (adverse effects)

5

5104

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.09, 0.74]

9 Headache (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

9.1 Cohort studies (adverse effects)

5

3322

Risk Ratio (M‐H, Random, 95% CI)

1.21 [0.50, 2.92]

9.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

2.31 [1.25, 4.27]

9.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

2.45 [1.38, 4.34]

10 Dizziness (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

10.1 Cohort studies (adverse effects)

5

2633

Risk Ratio (M‐H, Random, 95% CI)

3.49 [0.88, 13.75]

10.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

3.05 [1.30, 7.16]

10.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

2.40 [1.47, 3.90]

10.4 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.62, 0.73]

11 Abnormal dreams (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

11.1 Cohort studies (adverse effects)

4

2588

Risk Ratio (M‐H, Random, 95% CI)

10.49 [3.79, 29.10]

11.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

1.02 [0.07, 15.89]

11.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

4.33 [2.08, 9.00]

12 Insomnia (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

12.1 Cohort studies (adverse effects)

4

3212

Risk Ratio (M‐H, Random, 95% CI)

4.14 [1.19, 14.44]

12.2 RCTs (adverse events)

1

123

Risk Ratio (M‐H, Random, 95% CI)

2.03 [0.65, 6.40]

12.3 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Random, 95% CI)

4.54 [2.09, 9.83]

12.4 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.43, 0.49]

13 Anxiety (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

13.1 Cohort studies (adverse effects)

3

2559

Risk Ratio (M‐H, Fixed, 95% CI)

18.04 [9.32, 34.93]

13.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

8.74 [1.99, 38.40]

13.3 Retrospective healthcare record analysis (adverse events)

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.51 [0.47, 0.56]

14 Depressed mood (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

14.1 Cohort studies (adverse effects)

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

11.43 [5.21, 25.07]

14.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

6.27 [1.82, 21.62]

14.3 Retrospective healthcare record analysis (adverse events)

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

0.55 [0.51, 0.60]

15 Abnormal thoughts and perceptions Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

15.1 Cohort studies (adverse effects)

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

6.60 [0.92, 47.20]

15.2 Retrospective healthcare record analyses (adverse events)

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

0.41 [0.26, 0.66]

16 Pruritis (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

16.1 Cohort studies (adverse effects)

2

1794

Risk Ratio (M‐H, Fixed, 95% CI)

0.52 [0.30, 0.91]

16.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

2.69 [0.93, 7.78]

17 Photosensitivity (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

17.1 Cohort studies (adverse effects)

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

0.08 [0.05, 0.11]

17.2 Cohort studies (adverse events)

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

0.03 [0.00, 0.49]

18 Yeast infection (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

18.1 Cohort studies (adverse effects)

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

0.10 [0.06, 0.16]

18.2 Cohort studies (adverse events)

1

354

Risk Ratio (M‐H, Fixed, 95% CI)

0.19 [0.06, 0.63]

19 Visual impairment (all studies) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

19.1 Cohort studies (adverse effects)

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [1.41, 3.99]

20 Other adverse effects (cohort studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

20.1 Alopecia

2

1875

Risk Ratio (M‐H, Fixed, 95% CI)

3.44 [1.96, 6.03]

20.2 Asthenia

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

1.83 [0.89, 3.76]

20.3 Balance disorder

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

2.87 [1.48, 5.59]

20.4 Decreased appetite

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

1.23 [0.42, 3.64]

20.5 Fatigue

2

74

Risk Ratio (M‐H, Fixed, 95% CI)

0.23 [0.03, 1.77]

20.6 Hypoaesthesia

2

2445

Risk Ratio (M‐H, Fixed, 95% CI)

11.48 [3.01, 43.70]

20.7 Malaise

1

734

Risk Ratio (M‐H, Fixed, 95% CI)

0.28 [0.11, 0.71]

20.8 Mouth ulcers

1

33

Risk Ratio (M‐H, Fixed, 95% CI)

0.5 [0.02, 11.42]

20.9 Palpitations

1

1761

Risk Ratio (M‐H, Fixed, 95% CI)

2.76 [0.16, 48.91]

20.10 Tinnitus

1

684

Risk Ratio (M‐H, Fixed, 95% CI)

7.20 [0.39, 133.30]

21 Other adverse events (RCTs) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

21.1 Constipation

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

21.2 Cough

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

0.53 [0.28, 1.01]

21.3 Decreased appetite

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

3.56 [1.24, 10.20]

21.4 Malaise

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.88, 4.69]

21.5 Palpitations

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

21.6 Pyrexia

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.85 [1.09, 7.42]

21.7 Sexual dysfunction

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

3.05 [0.33, 28.51]

21.8 Somnolence

1

123

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.19, 21.84]

22 Other adverse events (cohort studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

22.1 Adjustment disorder

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.43 [0.40, 0.45]

22.2 Confusion

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

2.18 [0.24, 19.49]

22.3 Convulsions

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.45, 0.75]

22.4 Hallucinations

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.18 [0.08, 0.45]

22.5 Paranoia

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.40 [0.10, 1.63]

22.6 Palpitations

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

13.44 [1.73, 104.38]

22.7 Panic attacks

1

21065

Risk Ratio (M‐H, Fixed, 95% CI)

4.16 [0.55, 31.49]

22.8 PTSD

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.58 [0.53, 0.64]

22.9 Rash

1

668

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.50, 2.94]

22.10 Suicidal ideation

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.31, 0.47]

22.11 Suicide

2

376024

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [0.32, 4.56]

22.12 Tinnitus

1

354959

Risk Ratio (M‐H, Fixed, 95% CI)

0.65 [0.61, 0.71]

23 Adherence (cohort studies) Show forest plot

14

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

23.1 Adherence during travel

13

15583

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [1.12, 1.18]

23.2 Adherence in the post‐travel period

4

840

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.95, 1.22]

Figuras y tablas -
Comparison 2. Mefloquine versus doxycycline
Comparison 3. Mefloquine versus atovaquone‐proguanil

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

2

1293

Odds Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

2 Serious adverse events or effects (all studies) Show forest plot

3

3591

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.08, 23.22]

2.1 Cohort studies

3

3591

Risk Ratio (M‐H, Fixed, 95% CI)

1.40 [0.08, 23.22]

3 Discontinuations due to adverse effects (all studies) Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RCTs

3

1438

Risk Ratio (M‐H, Random, 95% CI)

2.86 [1.53, 5.31]

3.2 Cohort studies

9

7785

Risk Ratio (M‐H, Random, 95% CI)

2.73 [1.83, 4.08]

4 Nausea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

4.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

2.72 [1.52, 4.86]

4.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

2.50 [1.54, 4.06]

5 Vomiting (all studies) Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.31 [0.49, 3.50]

5.2 Cohort studies (adverse effects)

3

2180

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.08, 4.09]

6 Abdominal pain (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

0.90 [0.52, 1.56]

6.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

0.64 [0.38, 1.07]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

0.94 [0.60, 1.47]

7.2 Cohort studies (adverse effects)

7

3509

Risk Ratio (M‐H, Fixed, 95% CI)

0.85 [0.53, 1.35]

8 Mouth ulcers (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

8.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.45 [0.70, 3.00]

8.2 Cohort studies (adverse effects)

2

783

Risk Ratio (M‐H, Fixed, 95% CI)

0.12 [0.04, 0.37]

9 Headache (all studies) Show forest plot

9

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

9.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.72 [0.99, 2.99]

9.2 Cohort studies (adverse effects)

8

4163

Risk Ratio (M‐H, Fixed, 95% CI)

3.42 [1.71, 6.82]

10 Dizziness (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

10.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

3.99 [2.08, 7.64]

10.2 Cohort studies (adverse effects)

8

3986

Risk Ratio (M‐H, Fixed, 95% CI)

3.83 [2.23, 6.58]

10.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.23 [1.04, 1.46]

11 Abnormal dreams (all studies) Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

11.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [1.37, 3.04]

11.2 Cohort studies (adverse effects)

7

3848

Risk Ratio (M‐H, Random, 95% CI)

6.81 [1.65, 28.15]

12 Insomnia (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

12.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

4.42 [2.56, 7.64]

12.2 Cohort studies (adverse effects)

8

3986

Risk Ratio (M‐H, Fixed, 95% CI)

7.29 [4.37, 12.16]

12.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.24 [1.06, 1.44]

13 Anxiety (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

13.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

6.12 [1.82, 20.66]

13.2 Cohort studies (adverse effects)

4

2664

Risk Ratio (M‐H, Fixed, 95% CI)

10.10 [3.48, 29.32]

13.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.54 [1.28, 1.85]

14 Depressed mood (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

14.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

5.78 [1.71, 19.61]

14.2 Cohort studies (adverse effects)

6

3624

Risk Ratio (M‐H, Fixed, 95% CI)

8.02 [3.56, 18.07]

14.3 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.93 [1.56, 2.38]

15 Abnormal thoughts and perceptions (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

15.1 Cohort studies (adverse effects)

3

2433

Risk Ratio (M‐H, Fixed, 95% CI)

1.50 [0.30, 7.42]

15.2 Retrospective healthcare record analysis (adverse events)

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

3.00 [0.69, 12.97]

16 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

16.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

1.28 [0.60, 2.70]

16.2 Cohort studies (adverse effects)

3

1824

Risk Ratio (M‐H, Fixed, 95% CI)

2.07 [0.40, 10.68]

17 Visual impairment (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

17.1 RCTs (adverse effects)

1

976

Risk Ratio (M‐H, Fixed, 95% CI)

2.04 [0.88, 4.73]

17.2 Cohort studies (adverse effects)

2

1956

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.29, 4.72]

18 Other adverse effects (cohort studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

18.1 Allergic reaction

1

316

Risk Ratio (M‐H, Fixed, 95% CI)

0.79 [0.04, 14.48]

18.2 Alopecia

1

1469

Risk Ratio (M‐H, Fixed, 95% CI)

4.55 [0.30, 70.01]

18.3 Asthenia

2

1956

Risk Ratio (M‐H, Fixed, 95% CI)

1.84 [0.26, 13.12]

18.4 Balance disorder

1

1469

Risk Ratio (M‐H, Fixed, 95% CI)

2.86 [0.19, 44.19]

18.5 Cough

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

0.49 [0.08, 2.92]

18.6 Disturbance in attention

3

1363

Risk Ratio (M‐H, Fixed, 95% CI)

4.45 [1.84, 10.77]

18.7 Dyspepsia

2

362

Risk Ratio (M‐H, Fixed, 95% CI)

0.50 [0.17, 1.46]

18.8 Fatigue

2

618

Risk Ratio (M‐H, Fixed, 95% CI)

4.62 [0.47, 45.56]

18.9 Hypoaesthesia

2

1946

Risk Ratio (M‐H, Fixed, 95% CI)

4.45 [0.93, 21.26]

18.10 Loss of appetite

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.33, 1.43]

18.11 Muscle pain

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

7.57 [0.45, 127.80]

18.12 Palpitations

3

2180

Risk Ratio (M‐H, Fixed, 95% CI)

3.34 [0.73, 15.26]

18.13 Photosensitization

2

718

Risk Ratio (M‐H, Fixed, 95% CI)

0.69 [0.10, 4.92]

18.14 Pyrexia

1

652

Risk Ratio (M‐H, Fixed, 95% CI)

4.28 [0.24, 75.57]

18.15 Rash

2

711

Risk Ratio (M‐H, Fixed, 95% CI)

0.96 [0.15, 6.09]

18.16 Restlessness

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

5.24 [0.32, 84.52]

18.17 Slight illness

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

5.83 [0.36, 93.84]

18.18 Somnolence

1

487

Risk Ratio (M‐H, Fixed, 95% CI)

1.55 [0.21, 11.40]

18.19 Tinnitus

1

477

Risk Ratio (M‐H, Fixed, 95% CI)

2.31 [0.13, 42.64]

18.20 Circulatory disorders

1

224

Risk Ratio (M‐H, Fixed, 95% CI)

6.38 [0.36, 114.01]

19 Other adverse events (cohort studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

19.1 Adjustment disorder

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.76 [1.54, 2.02]

19.2 Confusion

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.04, 25.96]

19.3 Convulsions

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.35 [0.79, 2.30]

19.4 Hallucinations

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

0.25 [0.08, 0.79]

19.5 Paranoia

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.76 [0.08, 36.72]

19.6 PTSD

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

2.51 [1.93, 3.26]

19.7 Suicidal ideation

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.03, 2.77]

19.8 Suicide

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.06, 7.78]

19.9 Tinnitus

1

49419

Risk Ratio (M‐H, Fixed, 95% CI)

1.42 [1.21, 1.68]

20 Adherence (RCTs) Show forest plot

2

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

20.1 van Riemsdijk 2002

1

119

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.88, 1.02]

20.2 Overbosch 2001; during travel

1

966

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.95, 1.01]

20.3 Overbosch 2001; post‐travel

1

966

Risk Ratio (M‐H, Random, 95% CI)

0.80 [0.74, 0.85]

21 Adherence (cohort studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

21.1 During travel

6

5577

Risk Ratio (M‐H, Random, 95% CI)

1.08 [0.86, 1.34]

21.2 Post‐travel

2

422

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.64, 1.23]

Figuras y tablas -
Comparison 3. Mefloquine versus atovaquone‐proguanil
Comparison 4. Mefloquine versus chloroquine

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Clinical cases of malaria (RCTs) Show forest plot

4

877

Risk Ratio (M‐H, Fixed, 95% CI)

0.38 [0.28, 0.52]

2 Serious adverse events or effects (all studies) Show forest plot

10

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

2.1 RCTs

4

1000

Risk Ratio (M‐H, Fixed, 95% CI)

2.77 [0.32, 23.85]

2.2 Cohort studies

6

79257

Risk Ratio (M‐H, Fixed, 95% CI)

1.14 [0.62, 2.07]

3 Discontinuations due to adverse effects (all studies) Show forest plot

11

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

3.1 RCTs

3

815

Risk Ratio (M‐H, Fixed, 95% CI)

1.60 [0.61, 4.18]

3.2 Cohort studies in short‐term travellers

6

55397

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.78, 1.26]

3.3 Cohort studies in longer term occupational travellers

2

6085

Risk Ratio (M‐H, Fixed, 95% CI)

2.97 [2.41, 3.66]

4 Nausea (all studies) Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Cohort studies (adverse effects)

6

58984

Risk Ratio (M‐H, Random, 95% CI)

1.23 [0.89, 1.68]

4.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Random, 95% CI)

1.01 [0.57, 1.79]

5 Vomiting (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

5.1 Cohort studies (adverse effects)

5

5577

Risk Ratio (M‐H, Fixed, 95% CI)

1.05 [0.78, 1.40]

5.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.12 [0.36, 3.49]

6 Abdominal pain (all studies) Show forest plot

6

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

6.1 Cohort studies (adverse effects)

4

5440

Risk Ratio (M‐H, Fixed, 95% CI)

0.99 [0.80, 1.22]

6.2 RCTs (adverse events)

2

569

Risk Ratio (M‐H, Fixed, 95% CI)

0.71 [0.37, 1.36]

7 Diarrhoea (all studies) Show forest plot

8

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

7.1 Cohort studies (adverse effects)

5

5577

Risk Ratio (M‐H, Fixed, 95% CI)

0.84 [0.74, 0.95]

7.2 RCTs (adverse events)

3

772

Risk Ratio (M‐H, Fixed, 95% CI)

0.83 [0.46, 1.50]

8 Headache (all studies) Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 Cohort studies (adverse effects)

6

56998

Risk Ratio (M‐H, Random, 95% CI)

0.84 [0.53, 1.34]

8.2 RCTs (adverse events)

3

772

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.61, 1.31]

9 Dizziness (all studies) Show forest plot

7

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

9.1 Cohort studies (adverse effects)

5

58847

Risk Ratio (M‐H, Fixed, 95% CI)

1.51 [1.34, 1.70]

9.2 RCTs (adverse events)

2

569

Risk Ratio (M‐H, Fixed, 95% CI)

0.72 [0.35, 1.46]

10 Abnormal dreams (all studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

10.1 Cohort studies (adverse effects)

4

2845

Risk Ratio (M‐H, Fixed, 95% CI)

1.21 [1.10, 1.33]

10.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

2.70 [1.05, 6.95]

11 Insomnia (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

11.1 Cohort studies (adverse effects)

5

56952

Risk Ratio (M‐H, Random, 95% CI)

1.81 [0.73, 4.51]

11.2 RCTs (adverse events)

1

359

Risk Ratio (M‐H, Random, 95% CI)

1.19 [0.76, 1.84]

12 Anxiety (all studies) Show forest plot

3

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

12.1 Cohort studies (adverse effects)

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

6.30 [4.37, 9.09]

13 Depressed mood (all studies) Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

13.1 Cohort studies (adverse effects)

5

58855

Risk Ratio (M‐H, Random, 95% CI)

3.14 [1.15, 8.57]

14 Abnormal thoughts and perceptions Show forest plot

4

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

14.1 Cohort studies (adverse effects)

4

4831

Risk Ratio (M‐H, Fixed, 95% CI)

5.49 [2.65, 11.35]

15 Pruritis (all studies) Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

15.1 Cohort studies (adverse effects)

2

55544

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.92, 1.40]

15.2 RCTs (adverse events)

2

413

Risk Ratio (M‐H, Random, 95% CI)

0.28 [0.03, 2.93]

16 Visual impairment (all studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

16.1 Cohort studies (adverse effects)

5

58847

Risk Ratio (M‐H, Random, 95% CI)

1.10 [0.50, 2.44]

16.2 RCTs (adverse events)

1

210

Risk Ratio (M‐H, Random, 95% CI)

0.14 [0.01, 2.63]

17 Vertigo (all studies) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

17.1 Cohort studies (adverse effects)

1

746

Risk Ratio (M‐H, Fixed, 95% CI)

1.13 [0.05, 23.43]

18 Cohort studies in travellers; prespecified adverse effects Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

18.1 Vertigo

1

746

Risk Ratio (M‐H, Random, 95% CI)

1.13 [0.05, 23.43]

18.2 Nausea

5

56847

Risk Ratio (M‐H, Random, 95% CI)

1.42 [0.94, 2.13]

18.3 Vomiting

4

3440

Risk Ratio (M‐H, Random, 95% CI)

0.89 [0.55, 1.42]

18.4 Abdominal pain

3

3303

Risk Ratio (M‐H, Random, 95% CI)

0.98 [0.74, 1.30]

18.5 Diarrhoea

4

3440

Risk Ratio (M‐H, Random, 95% CI)

1.22 [0.57, 2.64]

18.6 Headache

5

54861

Risk Ratio (M‐H, Random, 95% CI)

1.12 [0.48, 2.65]

18.7 Dizziness

4

56710

Risk Ratio (M‐H, Random, 95% CI)

1.52 [1.10, 2.10]

18.8 Abnormal dreams

3

708

Risk Ratio (M‐H, Random, 95% CI)

4.21 [0.57, 31.33]

18.9 Insomnia

4

54815

Risk Ratio (M‐H, Random, 95% CI)

1.56 [0.40, 6.10]

18.10 Anxiety

2

1271

Risk Ratio (M‐H, Random, 95% CI)

3.94 [0.53, 29.48]

18.11 Depressed mood

4

56710

Risk Ratio (M‐H, Random, 95% CI)

2.49 [0.75, 8.31]

18.12 Abnormal thoughts or perceptions

3

2694

Risk Ratio (M‐H, Random, 95% CI)

4.42 [1.58, 12.40]

18.13 Pruritis

1

53407

Risk Ratio (M‐H, Random, 95% CI)

1.18 [0.94, 1.48]

18.14 Visual impairment

4

56710

Risk Ratio (M‐H, Random, 95% CI)

0.66 [0.55, 0.79]

19 Other adverse effects (cohort studies) Show forest plot

5

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

19.1 Altered spatial perception

1

2032

Risk Ratio (M‐H, Fixed, 95% CI)

3.16 [1.55, 6.45]

19.2 Alopecia

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

1.69 [1.27, 2.25]

19.3 Asthenia

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

1.52 [0.97, 2.40]

19.4 Balance disorder

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

3.59 [2.15, 6.00]

19.5 Confusion

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.03 [0.11, 36.31]

19.6 Decreased appetite

1

2032

Risk Ratio (M‐H, Fixed, 95% CI)

1.17 [0.87, 1.57]

19.7 Fatigue

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.37 [0.57, 9.80]

19.8 Hypoaesthesia

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

20.26 [1.23, 333.93]

19.9 Irritability

1

746

Risk Ratio (M‐H, Fixed, 95% CI)

4.75 [0.28, 80.59]

19.10 Mouth ulcers

2

55439

Risk Ratio (M‐H, Fixed, 95% CI)

1.37 [1.01, 1.87]

19.11 Paraesthesia

2

2778

Risk Ratio (M‐H, Fixed, 95% CI)

2.22 [1.27, 3.89]

19.12 Palpitations

3

3408

Risk Ratio (M‐H, Fixed, 95% CI)

4.71 [0.91, 24.26]

19.13 Photosensitization

2

2662

Risk Ratio (M‐H, Fixed, 95% CI)

0.89 [0.52, 1.53]

19.14 Restlessness

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

4.74 [0.65, 34.46]

19.15 Slight illness

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

2.65 [0.64, 10.87]

19.16 Somnolence

1

525

Risk Ratio (M‐H, Fixed, 95% CI)

6.08 [0.37, 100.36]

19.17 Yeast infection

1

2137

Risk Ratio (M‐H, Fixed, 95% CI)

1.15 [0.53, 2.49]

20 Other adverse events (RCTs) Show forest plot

2

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

20.1 Abdominal distension

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

3.13 [0.64, 15.27]

20.2 Anger

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

0.33 [0.07, 1.55]

20.3 Disturbance in attention

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

3.16 [0.61, 16.47]

20.4 Irritability

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.08 [0.45, 2.64]

20.5 Loss of appetite

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.06 [0.35, 3.25]

20.6 Malaise

1

203

Risk Ratio (M‐H, Fixed, 95% CI)

0.32 [0.01, 7.85]

20.7 Mood altered

1

359

Risk Ratio (M‐H, Fixed, 95% CI)

1.13 [0.29, 4.34]

21 Pregnancy related outcomes (RCTs) Show forest plot

1

Risk Ratio (M‐H, Fixed, 95% CI)

Subtotals only

21.1 Spontaneous abortions

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

0.80 [0.36, 1.79]

21.2 Still births

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

1.01 [0.67, 1.52]

21.3 Congenital malformations

1

2334

Risk Ratio (M‐H, Fixed, 95% CI)

0.0 [0.0, 0.0]

22 Adherence (cohort studies) Show forest plot

6

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

22.1 Short‐term travellers

3

852

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.90, 1.13]

22.2 Short‐term travellers: after return

1

46

Risk Ratio (M‐H, Random, 95% CI)

1.00 [0.54, 1.87]

22.3 Longer‐term occupational travellers

2

5777

Risk Ratio (M‐H, Random, 95% CI)

2.02 [1.80, 2.26]

Figuras y tablas -
Comparison 4. Mefloquine versus chloroquine
Comparison 5. Mefloquine versus currently used regimens; by study design

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Nausea; effects Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.72 [1.52, 4.86]

1.2 Cohort studies

11

5973

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.78, 3.77]

2 Abdominal pain; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

0.90 [0.52, 1.56]

2.2 Cohort studies

9

4494

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.27, 0.87]

3 Diarrhoea; effects Show forest plot

11

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

0.94 [0.60, 1.47]

3.2 Cohort studies

10

7648

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.28, 1.34]

4 Headache; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.72 [0.99, 2.99]

4.2 Cohort studies

9

5592

Risk Ratio (M‐H, Random, 95% CI)

2.19 [1.22, 3.93]

5 Dizziness; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

3.99 [2.08, 7.64]

5.2 Cohort studies

9

4606

Risk Ratio (M‐H, Random, 95% CI)

3.17 [1.58, 6.35]

6 Abnormal dreams; effects Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

6.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [1.37, 3.04]

6.2 Cohort studies

7

4543

Risk Ratio (M‐H, Random, 95% CI)

7.30 [2.51, 21.18]

7 Insomnia; effects Show forest plot

10

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

7.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

4.42 [2.56, 7.64]

7.2 Cohort studies

9

5299

Risk Ratio (M‐H, Random, 95% CI)

5.70 [2.83, 11.47]

8 Anxiety; effects Show forest plot

5

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

8.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

6.12 [1.82, 20.66]

8.2 Cohort studies

4

3390

Risk Ratio (M‐H, Random, 95% CI)

15.26 [8.66, 26.89]

9 Depressed mood; effects Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

9.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

5.78 [1.71, 19.61]

9.2 Cohort studies

6

4236

Risk Ratio (M‐H, Random, 95% CI)

7.82 [3.79, 16.12]

10 Abnormal thoughts or perceptions; effects Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

10.1 Cohort studies

3

3045

Risk Ratio (M‐H, Random, 95% CI)

4.20 [0.81, 21.87]

11 Pruritis; effects Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

11.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

1.28 [0.60, 2.70]

11.2 Cohort studies

3

2034

Risk Ratio (M‐H, Random, 95% CI)

0.88 [0.16, 4.76]

12 Visual impairment; effects Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

12.1 RCTs

1

976

Risk Ratio (M‐H, Random, 95% CI)

2.04 [0.88, 4.73]

12.2 Cohort studies

3

2560

Risk Ratio (M‐H, Random, 95% CI)

2.06 [1.05, 4.02]

13 Adherence; during travel Show forest plot

12

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

13.1 RCTs

1

119

Risk Ratio (M‐H, Random, 95% CI)

0.95 [0.88, 1.02]

13.2 Cohort studies

11

12131

Risk Ratio (M‐H, Random, 95% CI)

1.16 [1.03, 1.30]

14 Adherence; after return Show forest plot

4

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

14.1 Cohort studies

4

1221

Risk Ratio (M‐H, Random, 95% CI)

1.04 [0.92, 1.17]

Figuras y tablas -
Comparison 5. Mefloquine versus currently used regimens; by study design