An open label study of the safety and efficacy of a single dose of weekly chloroquine and azithromycin administered for malaria prophylaxis in healthy adults challenged with 7G8 chloroquine-resistant Plasmodium falciparum in a controlled human malaria infection model

Jeffrey Livezey, Patrick Twomey, Meshell Morrison, Susan Cicatelli, Elizabeth H Duncan, Melinda Hamer, Christine Lee, Jack Hutter, Kristin Mills, Jesse DeLuca, Lucas Poon, Daniel Selig, Chau Vuong, Jason Sousa, Thomas Oliver, Jason Bennett, James E Moon, April Sikaffy, Martha Sedegah, Donna Tosh, Mara Kreishman-Deitrick, Paige Waterman, Jeffrey Livezey, Patrick Twomey, Meshell Morrison, Susan Cicatelli, Elizabeth H Duncan, Melinda Hamer, Christine Lee, Jack Hutter, Kristin Mills, Jesse DeLuca, Lucas Poon, Daniel Selig, Chau Vuong, Jason Sousa, Thomas Oliver, Jason Bennett, James E Moon, April Sikaffy, Martha Sedegah, Donna Tosh, Mara Kreishman-Deitrick, Paige Waterman

Abstract

Background: Malaria remains the top infectious disease threat facing the U.S. military in many forward operating environments. Compliance with malaria chemoprophylaxis remains a critical component in preventing malaria in the deployed Service Member. Studies of previous military operations show that compliance is consistently higher with weekly versus daily dosing regimens. Current FDA approved weekly chemoprophylaxis options have contraindications that can limit prescribing. The combination of chloroquine (CQ) with azithromycin (AZ) has previously been shown to be an efficacious treatment option for malaria, has pharmacokinetics compatible with weekly dosing, and has shown synergy when combined in vitro.

Methods: In this open label study, 18 healthy volunteers, aged 18-50 years (inclusive), were randomly assigned to receive either 300 mg CQ or 300 mg CQ and 2 gm azithromycin (CQAZ) of directly observed therapy, weekly for 3 weeks prior to undergoing mosquito bite challenge with chloroquine-resistant Plasmodium falciparum. Volunteers that remained asymptomatic and had no evidence of parasitaemia continued to receive weekly post-exposure chemoprophylaxis for 3 weeks following malaria challenge. The primary endpoint was the number of volunteers that remained asymptomatic and had no evidence of parasitaemia 28 days after the malaria challenge.

Results: All 6 (100%) volunteers randomized to the CQ control group became symptomatic with parasitaemia during the 28-day post-challenge period. Only 1/12 (8.3%) of volunteers in the CQAZ group developed symptoms and parasitaemia during the 28-day post-challenge period. However, after chemoprophylaxis was discontinued an additional 6 volunteers developed parasitaemia between days 28-41 after challenge, with 4 of 6 experiencing symptoms. 80% of subjects in the CQAZ group experienced treatment related gastrointestinal adverse events (including 13% that experienced severe nausea) compared to 38% in the CQ group. A comparison of the pharmacokinetics in the CQAZ group demonstrated higher azithromycin Cmax (p = 0.03) and AUC (p = 0.044) levels in those volunteers who never became parasitaemic compared to those who did.

Conclusion: Given the high rate of side effects and poor efficacy when administered for 3 weeks before and after challenge, the combination of weekly chloroquine and azithromycin is a suboptimal regimen combination for weekly malaria chemoprophylaxis. Trial registration ClinicalTrials.gov NCT03278808.

Keywords: Azithromycin; Chloroquine; Controlled human malaria infection; Malaria chemoprophylaxis.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
CQAZ study design schema
Fig. 2
Fig. 2
Clinical trial participant flow diagram
Fig. 3
Fig. 3
Exposure-time curves for AZ, CQ and CQm in protected and non-protected subgroups of CQAZ group
Fig. 4
Fig. 4
Quantile plots relating exposure of azithromycin, chloroquine and desethylchloroquine to proportion of subjects protected from malaria infection. Solid circles and error bars represent mean AUC of each quantile and the corresponding 80% confidence intervals, respectively

References

    1. WHO. World malaria report 2019. Geneva: World Health Organization; 2019. . Accessed 12 Mar 2020.
    1. Mace KE, Arguin PM, Lucchi NW, Tan KR. Malaria surveillance—United States, 2016. MMWR Surveill Summ. 2019;68:1–35. doi: 10.15585/mmwr.ss6805a1.
    1. Saunders DL, Garges E, Manning JE, Bennett K, Schaffer S, Kosmowski, et al. Safety, tolerability, and compliance with long-term antimalarial chemoprophylaxis in American soldiers in Afghanistan. Am J Trop Med Hyg. 2015;93:584–590. doi: 10.4269/ajtmh.15-0245.
    1. Degrees Pharmaceuticals Limited. ARAKODA™ (tafenoquine) Label. . Accessed 11 Feb 2020.
    1. Sidhu AB, Sun Q, Nkrumah LJ, Dunne MW, Sacchettini JC, Fidock DA. In vitro efficacy, resistance selection, and structural modeling studies implicate the malarial parasite apicoplast as the target of azithromycin. J Biol Chem. 2007;282:2494–2504. doi: 10.1074/jbc.M608615200.
    1. Rosenthal PJ. Azithromycin for malaria? Am J Trop Med Hyg. 2016;95:2–4. doi: 10.4269/ajtmh.16-0332.
    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. doi: 10.1128/AAC.46.8.2518-2524.2002.
    1. Pereira MR, Henrich PP, Sidhu AB, Johnson D, Hardink D, 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. doi: 10.1128/AAC.01566-10.
    1. Cook JA, Randinitis EJ, Bramsom CR, Wesche DL. Lack of a pharmacokinetic interaction between azithromycin and chloroquine. Am J Trop Med Hyg. 2006;74:407–412. doi: 10.4269/ajtmh.2006.74.407.
    1. Andersen SL, Oloo AJ, Gordon DM, Ragama OB, Aleman M, Berman JD, et al. Successful double-blinded, randomized, placebo controlled field trial of azithromycin and doxycycline as prophylaxis for malaria in western Kenya. Clin Infect Dis. 1998;26:146–150. doi: 10.1086/516281.
    1. Taylor WR, Richie TL, Fryauff DJ, Ohrt C, Picarima H, Tang D, et al. Tolerability of azithromycin as malaria prophylaxis in adults in northeast Papua, Indonesia. Antimicrob Agents Chemother. 2003;47:2199–2203. doi: 10.1128/AAC.47.7.2199-2203.2003.
    1. Taylor WR, Richie TL, Fryauff DJ, Picarima H, Ohrt C, Tang D, et al. Malaria prophylaxis using azithromycin: a double-blind, placebo-controlled trial in Irian Jaya, Indonesia. Clin Infect Dis. 1999;28:74–81. doi: 10.1086/515071.
    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. doi: 10.1086/429343.
    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. doi: 10.1186/1475-2875-13-458.
    1. Kshirsagar NA, Gogtay NJ, Moran D, Utz G, Sethia A, Sarkar S, et al. Treatment of adults with acute uncomplicated malaria with azithromycin and chloroquine in India, Colombia, and Suriname. Res Rep Trop Med. 2017;8:85–104.
    1. Update: Malaria, U.S. Armed Forces, 2018. MSMR. 2019;26(2):2–7.
    1. Brisson M, Brisson P. Compliance with antimalaria chemoprophylaxis in a combat zone. Am J Trop Med Hyg. 2012;86:587–590. doi: 10.4269/ajtmh.2012.11-0511.
    1. Newton JA, Jr, Schnepf GA, Wallace MR, Lobel HO, Kennedy CA, Oldfield E. Malaria in US Marines returning from Somalia. JAMA. 1994;272:397–399. doi: 10.1001/jama.1994.03520050077034.
    1. Kotwal RS, Wenzel RB, Sterling RA, Porter WD, Jordan NN, Petruccelli BP. An outbreak of malaria in US Army Rangers returning from Afghanistan. JAMA. 2005;292:212–216. doi: 10.1001/jama.293.2.212.
    1. Burkot TR, Williams JL, Schneider I. lnfectivity to mosquitoes of Plasmodium falciparum clones grown in vitro from the same isolate. Trans R Soc Trop Med Hyg. 1984;78:339–341. doi: 10.1016/0035-9203(84)90114-7.
    1. WHO . Methods for surveillance of antimalarial drug efficacy. Geneva: World Health Organization; 2009. pp. 1–90.
    1. Kobayashi T, Gamboa D, Ndiaye D, Cui L, Sutton PL, Vinetz JM. Malaria diagnosis across the International Centers of Excellence for Malaria Research: platforms, performance, and standardization. Am J Trop Med Hyg. 2015;93(3 Suppl):99–109. doi: 10.4269/ajtmh.15-0004.
    1. Teuscher F, Gatton ML, Chen N, Peters J, Kyle DE, Cheng Q. Artemisinin-induced dormancy in Plasmodium falciparum: duration, recovery rates, and implications in treatment failure. J Infect Dis. 2010;202:1362–1368. doi: 10.1086/656476.
    1. Duvalsaint M, Kyle DE. Phytohormones, isoprenoids, and role of the apicoplast in recovery from dihydroartemisinin-induced dormancy of Plasmodium falciparum. Antimicrob Agents Chemother. 2018;62:e01771-17. doi: 10.1128/AAC.01771-17.
    1. Handsfield HH, Dalu ZA, Martin DH, Douglas JM, Jr, McCarty JM, Schlossberg D. Multicenter trial of single-dose azithromycin vs. ceftriaxone in the treatment of uncomplicated gonorrhea. Azithromycin Gonorrhea Study Group. Sex Transm Dis. 1994;21:107–111. doi: 10.1097/00007435-199403000-00010.
    1. Takahashi S, Kiyota H, Ito S, Iwasawa A, Hiyama Y, Uehara T, et al. Clinical efficacy of a single two gram dose of azithromycin extended release for male patients with urethritis. Antibiotics (Basel). 2014;3:109–120. doi: 10.3390/antibiotics3020109.
    1. Zithromax, F. L. A. Azithromycin (Zithromax) Label 2009. . Accessed 2 Mar 2020.
    1. Stanisic DI, McCarthy JS, Good MF. Controlled human malaria infection: applications, advances, and challenges. Infect Immun. 2017;86:e00479-17. doi: 10.1128/IAI.00479-17.

Source: PubMed

3
Abonneren