Efficacy and Safety of Azithromycin-Chloroquine versus Sulfadoxine-Pyrimethamine for Intermittent Preventive Treatment of Plasmodium falciparum Malaria Infection in Pregnant Women in Africa: An Open-Label, Randomized Trial

Joshua Kimani, Kamija Phiri, Steve Kamiza, Stephan Duparc, Ayman Ayoub, Ricardo Rojo, Jeffery Robbins, Russell Orrico, Pol Vandenbroucke, Joshua Kimani, Kamija Phiri, Steve Kamiza, Stephan Duparc, Ayman Ayoub, Ricardo Rojo, Jeffery Robbins, Russell Orrico, Pol Vandenbroucke

Abstract

Background: The World Health Organization recommends intermittent preventive treatment in pregnancy (IPTp) with sulfadoxine-pyrimethamine (SP) in African regions with moderate to high malaria transmission. However, growing resistance to SP threatens the effectiveness of IPTp-SP, and alternative drugs are needed. This study tested the efficacy, tolerability, and safety of a fixed-dose combination azithromycin-chloroquine (AZCQ; 250 mg AZ/155 mg CQ base) for IPTp relative to IPTp-SP.

Methods and findings: A randomized, Phase 3, open-label, multi-center study was conducted in sub-Saharan Africa (Benin, Kenya, Malawi, Tanzania, and Uganda) between October 2010 and November 2013. Pregnant women received 3 IPTp courses with AZCQ (each course: 1,000/620 mg AZCQ QD for 3 days) or SP (each course 1,500/75 mg SP QD for 1 day) at 4- to 8-week intervals during the second and third trimester. Long-lasting insecticide-treated bednets were also provided at enrollment. Study participants were followed up until day 28 post delivery (time window: day 28-42). The primary endpoint was the proportion of participants with sub-optimal pregnancy outcomes (a composite endpoint comprising live-borne neonates with low birth weight [LBW, <2,500 g], premature birth [<37 weeks], still birth [>28 weeks], abortion [≤28 weeks], lost to follow-up prior to observation of pregnancy outcome, or missing birth weight). The study was terminated early after recruitment of 2,891 of the planned 5,044 participants, due to futility observed in a pre-specified 35% interim analysis. In the final intent-to-treat dataset, 378/1,445 (26.2%) participants in the AZCQ and 342/1,445 (23.7%) in the SP group had sub-optimal pregnancy outcomes, with an estimated risk ratio (RR) of 1.11 (95% CI: 0.97, 1.25; p = 0.12). There was no significant difference in the incidence of LBW between treatment groups (57/1138 [5.0%] in the AZCQ group, 68/1188 [5.7%] in the SP group, RR 0.87 [95% CI: 0.62, 1.23]; p = 0.44). IPTp-AZCQ was less well-tolerated in mothers than IPTp-SP. Occurrences of congenital anomalies, deaths, and serious adverse events were comparable in neonates for both groups. Limitations included the open-label design and early study termination.

Conclusions: IPTp-AZCQ was not superior to IPTp-SP in this study and alternatives for IPTp-SP remain to be identified. The proportions of sub-optimal pregnancy outcomes and LBW were lower than expected, which may be linked to insecticide-treated bednet use throughout the study. Reduced incidences of symptomatic malaria infection and peripheral parasitemia in the AZCQ group relative to SP suggest that AZCQ warrants further investigation as an alternative treatment of uncomplicated malaria.

Trial registration: ClinicalTrials.gov (NCT01103063).

Conflict of interest statement

Competing Interests: Drs. A. Ayoub, R. Rojo, J. Robbins, P. Vandenbroucke, and Mr. R. Orrico are employees of the sponsor, Pfizer. Dr. S. Duparc is an employee of Medicines for Malaria Venture, who co-funded this study. Dr. J. Kimani was a principal investigator on this study. This does not alter the authors' adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Participant flow chart.
Fig 1. Participant flow chart.
aThe reasons why study participants were no longer willing to participate included: no specific reason provided, n = 17; family, social, or personal issues, n = 17; experience of AEs, n = 16; no longer willing to take study drug, n = 6; relocation, n = 3; stillbirth, n = 1. bThe reasons why study participants were no longer willing to participate included: family, social, or personal issues, n = 10; relocation, n = 3; no specific reason provided, n = 2. cThe AEs leading to discontinuation were combinations of nausea, vomiting, asthenia, spontaneous abortion, imminent abortion, and restlessness. dThe AE leading to discontinuation was premature rupture of membranes/stillbirth/umbilical cord abnormality. eThe causes of deaths were meningitis; postpartum hemorrhage and uterine rupture; and eclampsia. fThe death was due to peritonitis and intestinal perforation. gThe ‘other’ reasons for discontinuation were: relocation, n = 9; family, social, or personal issues, n = 5; non-compliance with study procedures, n = 2. hThe ‘other’ reasons for discontinuation were: relocation, n = 5; family, social, or personal issues, n = 5; no specific reason provided, n = 1.
Fig 2. Incidence of secondary outcomes in…
Fig 2. Incidence of secondary outcomes in the ITT population.
aDenominators are the number of subjects with a premature or full-term live birth and a non-missing birth weight. bDenominators are the number of pregnancy outcomes, excluding those that were unknown/missing. cIncluding neonatal death and congenital malformations. dDenominators are the number of subjects with a premature or full-term live birth. eDenominators are the number of subjects with available measurements. fSTI = sexually transmitted infection between first dose and Week 36 to 38 of gestation including T. pallidum, N. gonorrhoeae, C. trachomatis (diagnosed based on clinical presentation prior to week 36–38 and/or positive lab test results between week 36–38). gDiagnosed based on positive result at week 36 to 38 of gestation. hBetween first dose and delivery

References

    1. Dellicour S, Tatem AJ, Guerra CA, Snow RW, ter Kuile FO. Quantifying the number of pregnancies at risk of malaria in 2007: a demographic study. PLoS Med. 2010;7: e1000221 10.1371/journal.pmed.1000221
    1. Desai M, ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, et al. Epidemiology and burden of malaria in pregnancy. Lancet Infect Dis. 2007;7: 93–104.
    1. ter Kuile FO, Parise ME, Verhoeff FH, Udhayakumar V, Newman RD, van Eijk AM, et al. The burden of co-infection with human immunodeficiency virus type 1 and malaria in pregnant women in sub-saharan Africa. Am J Trop Med Hyg. 2004;71: 41–54.
    1. World Health Organization. A Strategic Framework for Malaria Prevention and Control During Pregnancy in the African Region. 2004. Available: . Accessed 14 Sept 2014.
    1. 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. Am J Trop Med Hyg. 1996;55: 33–41.
    1. McIntire DD, Bloom SL, Casey BM, Leveno KJ. Birth weight in relation to morbidity and mortality among newborn infants. N Engl J Med. 1999;340: 1234–1238.
    1. Marchant T, Willey B, Katz J, Clarke S, Kariuki S, ter Kuile F, et al. Neonatal mortality risk associated with preterm birth in East Africa, adjusted by weight for gestational age: individual participant level meta-analysis. PLoS Med. 2012;9: e1001292 10.1371/journal.pmed.1001292
    1. Zhong D, Afrane Y, Githeko A, Cui L, Menge DM, Yan G. Molecular epidemiology of drug-resistant malaria in western Kenya highlands. BMC Infect Dis. 2008;8: 105 10.1186/1471-2334-8-105
    1. World Health Organization. World Malaria Report 2014. 2014. Available: . Accessed 22 Jan 2015.
    1. World Health Organization. WHO policy brief for the implementation of intermittent preventive treatment of malaria in pregnancy using sulfadoxine-pyrimethamine (IPTp-SP). 2013 (Revised January 2014). Available: . Accessed 14 Sept 2014.
    1. Kayentao K, Kodio M, Newman RD, Maiga H, Doumtabe D, Ongoiba A, et al. Comparison of intermittent preventive treatment with chemoprophylaxis for the prevention of malaria during pregnancy in Mali. J Infect Dis. 2005;191: 109–116.
    1. Kayentao K, Garner P, van Eijk AM, Naidoo I, Roper C, Mulokozi A, et al. Intermittent preventive therapy for malaria during pregnancy using 2 vs 3 or more doses of sulfadoxine-pyrimethamine and risk of low birth weight in Africa: systematic review and meta-analysis. JAMA. 2013;309: 594–604. 10.1001/jama.2012.216231
    1. Malisa AL, Pearce RJ, Abdulla S, Mshinda H, Kachur PS, Bloland P, et al. Drug coverage in treatment of malaria and the consequences for resistance evolution—evidence from the use of sulphadoxine/pyrimethamine. Malar J. 2010;9: 190 10.1186/1475-2875-9-190
    1. Sridaran S, McClintock SK, Syphard LM, Herman KM, Barnwell JW, Udhayakumar V. Anti-folate drug resistance in Africa: meta-analysis of reported dihydrofolate reductase (dhfr) and dihydropteroate synthase (dhps) mutant genotype frequencies in African Plasmodium falciparum parasite populations. Malar J. 2010;9: 247 10.1186/1475-2875-9-247
    1. World Health Organization. Global report on antimalarial efficacy and drug resistance: 2000–2010. 2010. Available: . Accessed 13 Feb 2015.
    1. Chico RM, Pittrof R, Greenwood B, Chandramohan D. Azithromycin-chloroquine and the intermittent preventive treatment of malaria in pregnancy. Malar J. 2008;7: 255 10.1186/1475-2875-7-255
    1. Chico RM, Chandramohan D. Azithromycin plus chloroquine: combination therapy for protection against malaria and sexually transmitted infections in pregnancy. Expert Opin Drug Metab Toxicol. 2011;7: 1153–1167. 10.1517/17425255.2011.598506
    1. Ohrt C, Willingmyre GD, Lee P, Knirsch C, Milhous W. Assessment of azithromycin in combination with other antimalarial drugs against Plasmodium falciparum in vitro. Antimicrob Agents Chemother. 2002;46: 2518–2524.
    1. Pereira MR, Henrich PP, Sidhu AB, Johnson D, Hardink J, Van Deusen J, et al. In vivo and in vitro antimalarial properties of azithromycin-chloroquine combinations that include the resistance reversal agent amlodipine. Antimicrob Agents Chemother. 2011;55: 3115–3124. 10.1128/AAC.01566-10
    1. Dunne MW, Singh N, Shukla M, Valecha N, Bhattacharyya PC, Dev V, et al. A multicenter study of azithromycin, alone and in combination with chloroquine, for the treatment of acute uncomplicated Plasmodium falciparum malaria in India. J Infect Dis. 2005;191: 1582–1588.
    1. Sagara I, Oduro AR, Mulenga M, Dieng Y, Ogutu B, Tiono AB, et al. Efficacy and safety of a combination of azithromycin and chloroquine for the treatment of uncomplicated Plasmodium falciparum malaria in two multi-country randomised clinical trials in African adults. Malar J. 2014;13: 458 10.1186/1475-2875-13-458
    1. ter Kuile FO, van Eijk AM, Filler SJ. Effect of sulfadoxine-pyrimethamine resistance on the efficacy of intermittent preventive therapy for malaria control during pregnancy: a systematic review. JAMA. 2007;297: 2603–2616.
    1. Iriemenam NC, Shah M, Gatei W, van Eijk AM, Ayisi J, Kariuki S, et al. Temporal trends of sulphadoxine-pyrimethamine (SP) drug-resistance molecular markers in Plasmodium falciparum parasites from pregnant women in western Kenya. Malar J. 2012;11: 134 10.1186/1475-2875-11-134
    1. London School of Hygiene and Tropical Medicine. Drug resistance maps: Mapping the distribution of resistance genes of malaria in Africa. 2010 Available: . Accessed 10 Feb 2015.
    1. Chandra RS, Orazem J, Ubben D, Duparc S, Robbins J, Vandenbroucke P. Creative solutions to extraordinary challenges in clinical trials: methodology of a phase III trial of azithromycin and chloroquine fixed-dose combination in pregnant women in Africa. Malar J. 2013;12: 122 10.1186/1475-2875-12-122
    1. Zhao Q, Tensfeldt TG, Chandra R, Mould DR. Population pharmacokinetics of azithromycin and chloroquine in healthy adults and paediatric malaria subjects following oral administration of fixed-dose azithromycin and chloroquine combination tablets. Malar J. 2014;13: 36 10.1186/1475-2875-13-36
    1. Agresti A. Categorical Data Analysis. New York: John Wiley & Sons, Inc; 2003.
    1. Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. J Natl Cancer Inst. 1959;22: 719–748.
    1. Robins J, Breslow N, Greenland S. Estimators of the Mantel-Haenszel variance consistent in both sparse data and large-strata limiting models. Biometrics. 1986;42: 311–323.
    1. Hwang IK, Shih WJ, De Cani JS. Group sequential designs using a family of type I error probability spending functions. Stat Med. 1990;9: 1439–1445.
    1. Gonzalez 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 Med. 2014;11: e1001733 10.1371/journal.pmed.1001733
    1. ter Kuile FO, Terlouw DJ, Phillips-Howard PA, Hawley WA, Friedman JF, Kariuki SK, et al. Reduction of malaria during pregnancy by permethrin-treated bed nets in an area of intense perennial malaria transmission in western Kenya. Am J Trop Med Hyg. 2003;68: 50–60.
    1. Gamble C, Ekwaru PJ, Garner P, ter Kuile FO. Insecticide-treated nets for the prevention of malaria in pregnancy: a systematic review of randomised controlled trials. PLoS Med. 2007;4: e107
    1. Menendez C, D'Alessandro U, ter Kuile FO. Reducing the burden of malaria in pregnancy by preventive strategies. Lancet Infect Dis. 2007;7: 126–135.
    1. World Health Organization Evidence Review Group. Intermittent Preventive Treatment (IPT) of malaria in pregnancy. Malaria Policy Advisory Committee Meeting. 2013. Available: . Accessed 14 Sept 2014.
    1. 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. J Infect Dis. 2009;200: 991–1001. 10.1086/605474

Source: PubMed

3
Subscribe