Tafenoquine for travelers' malaria: evidence, rationale and recommendations

J Kevin Baird, J Kevin Baird

Abstract

Background: Endemic malaria occurring across much of the globe threatens millions of exposed travelers. While unknown numbers of them suffer acute attacks while traveling, each year thousands return from travel and become stricken in the weeks and months following exposure. This represents perhaps the most serious, prevalent and complex problem faced by providers of travel medicine services. Since before World War II, travel medicine practice has relied on synthetic suppressive blood schizontocidal drugs to prevent malaria during exposure, and has applied primaquine for presumptive anti-relapse therapy (post-travel or post-diagnosis of Plasmodium vivax) since 1952. In 2018, the US Food and Drug Administration approved the uses of a new hepatic schizontocidal and hypnozoitocidal 8-aminoquinoline called tafenoquine for the respective prevention of all malarias and for the treatment of those that relapse (P. vivax and Plasmodium ovale).

Methods: The evidence and rationale for tafenoquine for the prevention and treatment of malaria was gathered by means of a standard search of the medical literature along with the package inserts for the tafenoquine products Arakoda™ and Krintafel™ for the prevention of all malarias and the treatment of relapsing malarias, respectively.

Results: The development of tafenoquine-an endeavor of 40 years-at last brings two powerful advantages to travel medicine practice against the malaria threat: (i) a weekly regimen of causal prophylaxis; and (ii) a single-dose radical cure for patients infected by vivax or ovale malarias.

Conclusions: Although broad clinical experience remains to be gathered, tafenoquine appears to promise more practical and effective prevention and treatment of malaria. Tafenoquine thus applied includes important biological and clinical complexities explained in this review, with particular regard to the problem of hemolytic toxicity in G6PD-deficient patients.

Figures

Figure 1.
Figure 1.
Evolution of the 8-aminoquinoline hypnozoitocides, including the winnowing out of irreversible severe neurotoxicity of plasmocid and related compounds distinguished by fewer than four methylene groups separating the amino groups of the alkyl chain at the defining 8-amino position. Plasmochin and others (including primaquine) having at least four methylene groups exhibited no such neurotoxicity but instead reversible toxicity at sub-lethal doses involving principally hepatic, hematological and gastrointestinal systems
Figure 2.
Figure 2.
Antimalarial classes as guided by life cycle of the plasmodia
Figure 3.
Figure 3.
Schematic illustrating pitfalls and protections of suppressive (yellow dose indicators) or causal (orange dose indicators) chemoprevention of non-relapsing malaria like P. falciparum (top panel; red triangles and squares for inoculation and attack, respectively) or relapsing species like P. vivax (bottom panel; green triangles and squares)
Figure 4.
Figure 4.
Geographic distribution and prevalence of P. vivax (A) and P. falciparum (B) in 2010, reproduced here under Creative Commons license
Figure 5.
Figure 5.
Hypothesized relative attack rates in the months following radical cure illustrate possible impacts of variable risks of relapse or reinfection on the estimation hypnozoitocidal efficacy of tafenoquine (TQ) fixed at a presumed ‘actual’ 95% rate compared to a chloroquine (CQ) arm without hypnozoitocidal therapy (relapse and reinfection attacks)

References

    1. Newton CR, Taylor TE, Whitten RO. Pathophysiology of fatal falciparum malaria in African children. Am J Trop Med Hyg 1998; 58:673–83.
    1. Park SW, Kim DW, Park JW et al. . A case of fatal Plasmodium vivax malaria with multiple-organ failure. Infect Chemother 2005; 37:111–5.
    1. Groger M, Fischer HS, Veletzky L, Lalremruata A, Ramharter M. A systematic review of the clinical presentation, treatment, and relapse characteristics of human Plasmodium ovale malaria. Malar J 2017; 16:112.
    1. Collins WE, Jeffery GM. Plasmodium malariae: parasite and disease. Clin Microbiol Rev 2007; 20:579–92.
    1. Rajahram G, Barber BE, William T, Menon J, Anstey NM, Yeo TW. Deaths due to Plasmodium knowlesi malaria in Sabah, Malaysia: association with reporting as P. malariae and delayed parenteral artesunate. Malar J 2012; 11:284.
    1. Kain KC, Harrington MA, Tennyson S, Keystone JS. Imported malaria: prospective analysis of problems in diagnosis and management. Clin Infect Dis 1998; 27:142–9.
    1. Baird JK, Nelwan J, Taylor WR. Approach to the patient with malaria In: Keystone JS, Kozarsky PE, Connor BA, Nothdurft HD, Leder K, Mendelson M (eds). Travel Medicine, 4th edn Elsevier, 2019. Chapter 17.
    1. Tediosi F, Lengeler C, Castro M et al. Chapter 13: Malaria control In: Holmes KK, Bertozzi S, Bloom BR, and Jha P (eds). Major Infectious Diseases. Disease Control Priorities, 3rd edn, Vol. 6. Washington: DC: World Bank, 2017, pp. 347–364.
    1. Bhatt S, Weiss DJ, Cameron E et al. . The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature 2015; 526:207–11.
    1. Cibulskis RE, Alonso PE, Aponte J et al. . Malaria: global progress 2000–2015 and future challenges. Infect Dis Poverty 2016; 9:61.
    1. Mendis K, Sina BJ, Marchesini P, Carter R. The neglected burden of Plasmodium vivax malaria. Am J Trop Med Hyg 2001; 64:97–106.
    1. Baird JK. Neglect of Plasmodium vivax malaria. Trends Parasitol 2007; 23(11). PMID: 17933585.
    1. Price RN, Tjitra E, Guerra CA, Yeung S, White NJ, Anstey NM. Vivax malaria: neglected and not benign. Am J Trop Med Hyg 2007; 77:79–87.
    1. Bassat Q, Velarde M, Mueller I et al. . Key knowledge gaps for Plasmodium vivax control and elimination. Am J Trop Med Hyg 2016; 95:62–71.
    1. Carlton JM, Sina BJ, Adams JH. Why is Plasmodium vivax a neglected tropical disease? PLoS Negl Trop Dis 2011; 5:e1160.
    1. World Health Organization World Malaria Report 2017. Geneva; 2018. Geneva: World Health Organization; ISBN: 978 92 4 156552 3.
    1. Freedman DO, Chen LH, Kozarsky PE. Medical considerations before international travel. N Engl J Med 2016; 375:247–60.
    1. Boggild A, Brophy J, Charlebois P et al. . Summary of recommendations for the prevention of malaria by the Committee to Advise on Tropical Medicine and Travel (CATMAT). Can Commun Dis Rep 2014; 40:118–32.
    1. PHE Advisory Committee for Malaria Prevention for UK Travelers Guidelines for malaria prevention in travelers from the UK: 2017; 2017. London: Public Health England; 146pp.
    1. Baird JK. Management of Plasmodium vivax risk and illness in travelers. Trop Dis Travel Med Vaccines 2017; 3:7.
    1. Mühlberger N, Jelinek T, Gascon J et al. . Epidemiology and clinical features of vivax malaria imported to Europe: sentinel surveillance data from TropNetEurop. Malar J 2004; 3:5.
    1. Steinhardt LC, Magill AJ, Arguin PM. Review: malaria chemoprophylaxis for travellers to Latin America. Am J Trop Med Hyg 2011; 85:1015–24.
    1. Schwartz E, Parise M, Kozarsky P, Cetron M. Delayed onset of malaria – implications for chemoprophylaxis in travellers. N Engl J Med 2003; 349:1510–16.
    1. Tan KR, Hwang J. Tafenoquine receives regulatory approval in U.S. for prophylaxis of malaria and radical cure of Plasmodium vivax. J Travel Med 2018. .
    1. Gutteridge WE. Antimalarial drugs currently in development. J R Soc Med 1989; 17:63–6.
    1. Davidson DE Jr, Ager AL, Brown JL, Chapple FE, Whitmire RE, Rossan RN. New tissue schizontocidal antimalarial drugs. Bull World Health Organ 1981; 59:463–79.
    1. Ashley EA, Phyo AP. Drugs in development for malaria. Drugs 2018; 78:861–79.
    1. Peters W. The evolution of tafenoquine – antimalarial for a new millennium? J R Soc Med 1999; 92:345–52.
    1. Kitchener S, Nasveld P, Edstein MD. Tafenoquine for the treatment of current Plasmodium vivax malaria. Am J Trop Med Hyg 2007; 76:494–6.
    1. Crockett M, Kain KC. Tafenoquine: a promising new antimalarial agent. Expert Opin Investig Drugs 2007; 16:705–15.
    1. Jeffery GM. Infectivity of mosquitoes of Plasmodium vivax following treatment with chloroquine and other antimalarials. Am J Trop Med Hyg 1958; 7:207–11.
    1. Peters W, Robinson BL, Milhous WK. The chemotherapy of rodent malaria. LI. Studies on a new 8-aminoquinoline, WR 238,605. Ann Trop Med Parasitol 1993; 87:547–52.
    1. Coleman RE. Sporontocidal activity of the antimalarial WR-238605 against Plasmodium berghei ANKA in Anopheles stephensi. Am J Trop Med Hyg 1990; 42:196–2015.
    1. Shapiro TA, Ranasinha CD, Kumar N, Barditch-Crovo P. Prophylactic activity of atovaquone against Plasmodium falciparum in humans. Am J Trop Med Hyg 1999; 60:831–6.
    1. Berman JD, Nielsen R, Chulay JD et al. . Causal prophylactic activity of atovaquone-proguanil (Malarone) in a human challenge model. Trans R Soc Trop Med Hyg 2001; 95:429–32.
    1. Maguire JD, Llewellyn DM. Relapsing malaria after 6 months of daily atovaquone-proguanil in Afghanistan: the case for expanded use of primaquine as a causal prophylactic. J Travel Med 2007; 14:411–14.
    1. Metlzer E, Rahav G, Schwartz E. Vivax malaria chemoprophylaxis: the role of atovaquone-proguanil compared to other options. Clin Infect Dis 2018; 66:1751–5.
    1. DiTusa C, Kozar MP, Pybus B et al. . Causal prophylactic efficacy of primaquine, tafenoquine, and atovaquone-proguanil against Plasmodium cynomolgi in a rhesus monkey model. J Parasitol 2014; 100:671–3.
    1. Baird JK, Fryauff DJ, Hoffman SL. Primaquine for the prevention of malaria in travelers. Clin Infect Dis 2003; 37:1659–67.
    1. Nasveld P, Kitchener S. Treatment of acute vivax malaria with tafenoquine. Trans R Soc Trop Med Hyg 2005; 99:2–5.
    1. Shanks GD. Historical review: problematic malaria prophylaxis with quinine. Am J Trop Med Hyg 2016; 95:269–72.
    1. Arguin PM, Magill AJ For the record: a history of malaria chemoprophylaxis. . Visited 13 September 2018.
    1. Sinton JA, Smith S, Pottinger D. Studies on malaria with special reference to treatment. XII. Further researches into the treatment of chronic benign tertian malaria with plasmoquine and quinine. Indian J Med Res 1929; 20:793–814.
    1. Shannon JA. Chemotherapy in malaria. Bull N Y Acad Med 1946; 22:345–57.
    1. Centers for Disease Control Malaria among US military personnel returning from Somalia, 1993. MMWR Morb Mortal Wkly Rep 1993; 42:524–6.
    1. Krafts K, Hempelmann E, Skorska-Stania A. From methylene blue to chloroquine: a brief review of the development of antimalarial therapy. Parasitol Res 2012; 111:1–6.
    1. Kolifarhood G, Raeisi A, Ranjbar M et al. . Prophylactic efficacy of primaquine for preventing Plasmodium falciparum and Plasmodium vivax parasitemaemia in travelers: a meta-analysis and systematic review. Travel Med Infect Dis 2017; 17:5–18.
    1. Arnold J, Alving AS, Hockwald RS et al. . The antimalarial action of primaquine against the blood and tissue stages of falciparum malaria (Panama P-F-6 strain). J Lab Clin Med 1955; 46:391–7.
    1. Flaherty GT, Walden LJ, Townend M. Travel medicine physician adherence to guidelines for the emergency self-treatment of malaria. J Travel Med 2016; 23(5). doi:.
    1. Behrens R. Standy emergency treatment of malaria for travelers to low transmission destinations: does it make sense or save lives? J Travel Med 2017; 24(5). doi:.
    1. Boubaker R, Harard Fossati A, Meige P et al. . Malaria prevention strategies and recommendations from chemoprophylaxis to stand-by emergency treatment: a 10-year prospective study in a Swiss travel clinic. J Travel Med 2017; 24(5). doi:.
    1. Hwang J, Cullen CA, Kachur SP, Arguin PM, Baird JK. Severe morbidity and mortality risk from malaria in the United States, 1985-2011. Open Forum Infect Dis 2014; 1:ofu034.
    1. Barber BE, Grigg MJ, Piera KA et al. . Intravascular haemolysis in severe Plasmodium knowlesi malaria: association with endothelial activation, microvascular dysfunction, and acute kidney injury. Emerg Microbes Infect 2018; 7:106.
    1. Anstey NM, Douglas NM, Poespoprodjo JR, Price RN. Plasmodium vivax: clinical spectrum, risk factors and pathogenesis. Adv Parasitol 2012; 80:151–202.
    1. Baird JK. Evidence and implications of mortality in acute Plasmodium vivax malaria. Clin Microbiol Rev 2013; 26:36–57.
    1. Naing C, Whittaker MA, Wai VN, Mak JW. Is Plasmodium vivax malaria a severe malaria? A systematic review and meta-analysis. PLoS Negl Trop Dis 2014; 8:e3071.
    1. Quispe AM, Pozo E, Guerrero E et al. . Plasmodium vivax hospitalizations in a monoendemic malaria region: severe vivax malaria? Am J Trop Med Hyg 2014; 91:11–7.
    1. Douglas NM, Pontororing GJ, Lampah DA et al. . Mortality attributable to Plasmodium vivax malaria: a clinical audit from Papua, Indonesia. BMC Med 2014; 12:217.
    1. Siqueira AM, Lacerda MVG, Magalhaes BML et al. . Characterization of Plasmodium vivax-associated admissions to reference hospitals in Brazil and India. BMC Med 2015; 13:57.
    1. Mace KE, Arguin PM, Tan KR. Malaria surveillance – United States, 2015. MMWR Surveill Summ 2018; 67:1–28.
    1. Tatem AJ, Jia P, Ordanovich D et al. . The geography of imported malaria to non-endemic countries: a meta-analysis of nationally reported statistics. Lancet Infect Dis 2017; 17:98–107.
    1. Ryan JR, Stoute JA, Amon J et al. . Evidence for transmission of Plasmodium vivax among a duffy antigen negative population in Western Kenya. Am J Trop Med Hyg 2006; 75:575–81.
    1. Howes RE, Reiner RC Jr, Battle KE et al. . Plasmodium vivax transmission in Africa. PLoS Negl Trop Dis 2015; 9:e0004222.
    1. Brazeau NF, Whitesell A, Doctor SM et al. . Plasmodium vivax infections in Duffy-negative individuals in the Democratic Republic of the Congo. Am J Trop Med Hyg 2018. .
    1. Gething PW, Elyazar IR, Moyes CL et al. . A long neglected world map: Plasmodium vivax endemicity in 2010. PLoS Negl Trop Dis 2012; 6:e1814.
    1. Schlagenhauf P, Petersen E. Malaria chemoprophylaxis: strategies for risk group. Clin Microbiol Rev 2008; 21:466–72.
    1. Davlantes EA, Tan KR, Arguin PM. Quantifying malaria risk in travelers: a quixotic pursuit. J Travel Med 2017; 24(6). doi.
    1. Luzzatto L, Seneca E. G6PD deficiency: a classic example of pharmacogenetics with on-going clinical implications. Br J Haematol 2014; 164:469–80.
    1. Howes RE, Piel FB, Patil AP et al. . G6PD deficiency prevalence estimates of affected populations in malaria endemic countries: a geostatistical model-based map. PLoS Med 2012; 9:e1001339.
    1. World Health Organization Evidence Review Group Point-of-care testing to support safe use of primaquine for the treatment of vivax malaria. Malaria Policy Advisory Committee Meeting, 5-7 March, 2015, Geneva, Switzerland. WHO/HTM/GMP/MPAC/2015.6. accessed on 15 September 2018.
    1. Howes RE, Dewi M, PIel FB et al. . Spatial distribution of G6PD deficiency variants across malaria-endemic regions. Malar J 2013; 12:418.
    1. Rueangweerayut R, Bancone G, Harrell EJ et al. . Hemolytic potential of tafenoquine in female volunteers heterozygous for glucose-6-phosophate dehydrogenase (G6PD) deficiency (G6PD Mahidol variant) versus G6PD-normal volunteers. Am J Trop Med Hyg 2017; 97:702–11.
    1. Chu CS, Bancone G, Moore KA et al. . Haemolysis in G6PD heterozygous females treated with primaquine for Plasmodium vivax malaria: a nested cohort in a trial of radical curative regimens. PLoS Med 2017; 14:e1002224.
    1. Arakoda™ package insert 60 Degrees Pharmaceuticals LLC, Washington, DC; 2018.
    1. Baird JK, Dewi M, Subekti D, Elyazar I, Satyagraha AW. Noninferiority of glucose-6-phosphate dehydrogenase deficiency diagnosis by a point-of-care rapid test vs the laboratory fluorescent spot test demonstrated by copper inhibition in normal human red blood cells. Transl Res 2015; 165:677–88.
    1. Chu CS, Bancone G, Nosten F, White NJ, Luzzatto L. Primaquine-induced haemolysis in females heterozygous for G6PD deficiency. Malar J 2018; 17:101.
    1. Recht J, Ashley EA, White NJ. Use of primaquine and glucose-6-phosphate dehydrogenase deficiency testing: divergent policies and practices in endemic countries. PLoS Negl Trop Dis 2018; 12:e0006230.
    1. Roca-Feltrer A, Khim N, Kim S et al. . Field trial evaluation of the performance of point-of-care tests for screening G6PD deficiency in Cambodia. PLoS One 2014; 9:e116143.
    1. Bancone G, Chu CS, Chowwiwat N et al. . Suitability of capillary blood for quantitative assessment of G6PD activity and performances of G6PD point-of-care tests. Am J Trop Med Hyg 2015; 92:818–24.
    1. Oo NN, Bancone G, Maw LZ et al. . Validation of G6PD point-of-care tests among healthy volunteers in Yangon, Myanmar. PLoS One 2016; 11:e0152304.
    1. Ley B, Bancone G, von Seidlein L et al. . Methods for the field evaluation of quantitative G6PD diagnostics: a review. Malar J 2017; 16:361.
    1. Marcsisin SR, Reichard G, Pybus BS. Primaquine pharmacology in the context of CYP 2D6 pharmacogenomics: current state of the art. Pharmacol Ther 2016; 16:1–10.
    1. Bennett JW, Pybus BS, Yadava A et al. . Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med 2013; 369:1381–2.
    1. Ingram RJ, Crenna-Darusallam C, Soebianto S, Noviyanti R, Baird JK. The clinical and public health problem of relapse despite primaquine therapy: case review of repeated relapses of Plasmodium vivax acquired in Papua New Guinea. Malar J 2014; 13:488.
    1. Nelwan EJ, Ekawati LL, Tjahjono B et al. . Randomized trial of primaquine hypnozoitocidal efficacy when administered with artemisinin-combined blood schizontocides for radical cure of Plasmodium vivax in Indonesia. BMC Med 2015; 13:294.
    1. Baird JK, Louisa M, Noviyanti R et al. . Association of impaired cytochrome P-450 2D6 activity genotype and phenotype with therapeutic efficacy of primaquine treatment for latent Plasmodium vivax malaria. JAMA Open Network 2018; 1:e181449.
    1. Marcisisin SR, Sousa JC, Reichard GA et al. . Tafenoquine and NPC-1161B require CYP2D metabolism for antimalarial activity: implications for the 8-aminoquinoline class of antimalarial compounds. Malar J 2014; 13:2.
    1. St Jean PL, Zue Z, Carter N et al. . Tafenoquine treatment of Plasmodium vivax malaria: suggestive evidence that CYP2D6 reduced metabolism is not associated with relapse in the Phase 2b DETECTIVE trial. Malar J 2016; 15:97.
    1. Hill DR, Baird JK, Parise ME, Lewis LS, Ryan T, Magill AJ. Primaquine: report from CDC expert meeting on malaria chemoprophylaxis I. Am J Trop Med Hyg 2006; 75:402–15.
    1. Nasveld PE, Edstein MD, Brennan RM et al. . Randomized, double-blind study of the safety, tolerability, and efficacy of tafenoquine versus mefloquine for malaria prophylaxis in nonimmune subjects. Antimicrob Agents Chemother 2010; 54:792–8.
    1. Shanks GD, Oloo AJ, Aleman GM et al. . A new primaquine analog, tafenoquine (WR 238605), for prophylaxis against Plasmodium falciparum malaria. Clin Infect Dis 2001; 33:1968–74.
    1. Hale BR, Owusu-Agyei S, Fryauff DJ et al. . A randomized, double-blind, placebo-controlled, dose-ranging trial of tafenoquine for weekly prophylaxis against Plasmodium falciparum. Clin Infect Dis 2003; 36:541–9.
    1. 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 antimalarials in non-immune individuals during deployment to a malaria-endemic area. Malar J 2014; 14:49.
    1. Dow GS, Liu J, Lin G et al. . Summary of antimalarial prophylactic efficacy of tafenoquine from three placebo-controlled studies of residents of malaria-endemic areas. Malar J 2015; 14:473.
    1. Shanks D. The conundrum of malaria chemoprophylaxis. J Travel Med 2016; 23(6). pii: taw065. PMID: 27694470.
    1. Brueckner RP, Coster T, Wesche DL, Shmuklarsky M, Schuster B. Prophylaxis of Plasmodium falciparum infection in a human challenge model with WR238605, a new 8-aminoquinoline compound. Antimicrob Agents Chemother 1998; 42:1293–4.
    1. Dow G, Smith B. The blood schizontocidal activity of tafenoquine makes an essential contribution to its prophylactic efficacy in nonimmune subjects at the intended dose (200mg). Malar J 2017; 16:209.
    1. Lell B, Faucher J-P, Missinou MA et al. . Malaria chemoprophylaxis with tafenoquine: a randomized study. Lancet 2000; 355:2041–5.
    1. Thakkar N, Green JA, Koh GC, Duparc S, Tenero D, Goyal N. Population pharmacokinetics of tafenoquine, a novel antimalarial. Antimicrob Agents Chemother 2018. .
    1. Novitt-Moreno A, Ransom J, Dow G, Smith B, Read LT, Toovey S. Tafenoquine for malaria prophylaxis in adults: an integrated safety analysis. Travel Med Infect Dis 2017; 17:19–27.
    1. Leary KJ, Riel MA, Roy MJ et al. . A randomized, double-blind, safety and tolerability study to assess the ophthalmic and renal effects of tafenoquine 200mg weekly versus placebo for 6 months in healthy volunteers. Am J Trop Med Hyg 2009; 81:356–62.
    1. Fukuda M, Krudsood S, Mohamed K et al. . A randomized, double-blind, active-control trial to evaluate the efficacy and safety of a three day course of tafenoquine monotherapy for the treatment of Plasmodium vivax malaria. PLoS One 2017; 12:e0187376.
    1. Baird JK, Hoffman SL. Primaquine therapy for malaria. Clin Infect Dis 2004; 39:1336–45.
    1. Chu CS, White NJ. Management of relapsing Plasmodium vivax malaria. Expert Rev Anti Infect Ther 2016; 14:885–900.
    1. Baird JK, Valecha N, Duparc S, White NJ, Price RN. Diagnosis and treatment of Plasmodium vivax malaria. Am J Trop Med Hyg 2016; 95:35–51.
    1. Ashley EA, Recht J, White NJ. Primaquine: the risks and benefits. Malar J 2014; 13:418.
    1. Baird JK. Resistance to therapies by Plasmodium vivax. Clin Microbiol Rev 2009; 22:508–34.
    1. White NJ, Imwong M. Relapse. Adv Parasitol 2012; 80:113–42.
    1. Sutanto I, Tjahjono B, Basri H et al. . Randomized, open-label trial of primaquine against vivax malaria relapse in Indonesia. Antimicrob Agents Chemother 2013; 57:1128–35.
    1. Llanos-Cuentas A, Lacerda MV, Rueangweerayut R et al. . Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): a multicenter, couble-blind, randomized, phase 2b dose-selection study. Lancet 2014; 383:1049–58.
    1. Krintafel™ package insert. Washington, DC: GlaxoSmithKline Pharmaceuticals; 2018. Reference ID: 4294835.
    1. Baird JK. Resistance to chloroquine unhinges vivax malaria therapeutics. Antimicrob Agents Chemother 2011; 55:1827–30.
    1. Dow GS, Gettayacamin M, Hansukjariya P et al. . Radical curative efficacy of tafenoquine combination regimens in Plasmodium cynomolgi-infected Rhesus monkeys (Macaca mulatta). Malar J 2011; 10:212.
    1. Alving AS, Arnold J, Hockwald RS et al. . Potentiation of the curative action of primaquine in vivax malaria by quinine and chloroquine. J Lab Clin Med 1955; 46:301–6.
    1. Miller AK, Harrell E, Ye L et al. . Pharmacokinetic interactions and safety evaluations of coadministered tafenoquine and chloroquine in healthy subjects. Br J Clin Pharmacol 2013; 76:858–67.
    1. Green JA, Mohamed K, Goyal N et al. . Pharmacokinetic interactions between tafenoquine and dihydroartemisinin-piperaquine or artemether-lumefantrine in healthy adult subjects. Antimicrob Agents Chemother 2016; 60:7321–32.
    1. Clyde DF. Clinical problems associated with use of primaquine as a tissue schizontocidal and gametocytocidal drug. Bull World Health Organ 1981; 59:391–5.
    1. Recht J, Ashley EA, White N. Safety of 8-aminoquinoline antimalarial medicines. Geneva: World Health Organization, 2014, 222 ISBN 978 92 4 150697 7.
    1. Carson PE, Hohl R, Nora MV et al. . Toxicology of the 8-aminoquinolines and genetic factors associated with their toxicity in man. Bull World Health Organ 1981; 59:427–37.
    1. Gething PW, Patil AP, Smith DL et al. . A new world malaria map: Plasmodium falciparum endemicity in 2010. Malar J 2011; 10:378.

Source: PubMed

3
Abonneren