Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2022 Jun 1.
Published in final edited form as: Br J Clin Pharmacol. 2022 Jan 27;88(6):2657–2664. doi: 10.1111/bcp.15219

Methaemoglobinaemia and the radical curative efficacy of 8-aminoquinoline antimalarials

Nicholas J White 1,2,, James A Watson 1,2, J Kevin Baird 2,3
PMCID: PMC7612727  EMSID: EMS140787  PMID: 34997616

Abstract

Methaemoglobin results from the oxidation of ferrous to ferric iron in the centre of the haem moeity of haemoglobin. The production of dose-dependent methaemoglobinaemia by 8-aminoquinoline antimalarial drugs appears to be associated with, but is not directly linked to, therapeutic efficacy against latent Plasmodium vivax and P. ovale malarias (radical cure). Iatrogenic methaemoglobinaemia may be a useful pharmacodynamic measure in 8-aminoquinoline drug and dose optimization.

Keywords: methaemoglobinaemia, P. vivax, 8-aminoquinolines, primaquine, tafenoquine

Introduction

Hepatic latency in vivax and ovale malarias (dormant liver stage parasites called hypnozoites) requires treatment with 8-aminoquinoline drugs in order to prevent recurrent attacks called relapses. This is called radical cure). The first 8-aminoquinoline (variously named plasmoquine, plasmochin, or pamaquine) was discovered nearly one hundred years ago. Within four years of the reported discovery and introduction, Sinton and colleagues working in Northern India observed that the combination of quinine and plasmoquine was effective in preventing late recurrences (presumed relapses) of Plasmodium vivax malaria (1,2). This efficacy against late attacks of vivax malaria was very slow to be accepted by international authorities (notably the malaria commission of the League of Nations) and malaria experts. Plasmoquine was not well tolerated at the doses required for radical cure: abdominal pain and vomiting were dose limiting, and “cyanosis” was noted with higher doses (35). It was also observed that in patients of African or Asian origin, about 10% of developed significant haemolytic anaemia (5). The cyanosis following pamaquine therapy was caused by methaemoglobinaemia, and the sporadic severe haemolytic anaemia was later identified as resulting from the oxidant drug susceptibility of erythrocytes with reduced glucose-6-phosphate dehydrogenase (G6PD) activity. During the Second World War recurrent vivax malaria in the Indo-Burman and Pacific theatres of war was a major threat to soldiers on both sides. An intensive research effort, based in the United States, set out to discover more effective and better tolerated 8-aminoquinolines. Pharmacometric studies in volunteers infected with P. vivax (notably the frequent relapse Chesson strain originating in New Guinea) and in rhesus monkeys infected with P. cynomolgi continued after the end of the Second World War, and ultimately led to the replacement of pamaquine by primaquine in 1951 during the Korean war (6). These large and detailed pioneering volunteer studies recorded a wealth of valuable information.

Relapse in experimental and naturally acquired vivax malaria

In the experimental malaria studies, non-immune volunteers were infected either by blood taken from a malaria patient or by infected mosquito bites. The volunteers were then observed daily, usually for at least one year (many were prison volunteers). Building on the extensive experiences of malariatherapy of neurosyphilis during the early to mid-20th century, which had refined and standardized the methods of malaria infection and clinical management, at least five and usually ten infected (i.e. sporozoite positive) laboratory reared anopheline mosquitoes were used to initiate the infection. This ensured a substantial sporozoite inoculum was delivered to the recipient. With the Chesson strain of P. vivax the consequent early relapse rate was 100% (7).

This high relapse rate in experimental malaria should be contrasted with natural infections in which a single mosquito, with an infection of variable age (and thus transmission potential), infects an individual who may already have significant natural immunity which attenuates illness (8). This distinction is important because it has a substantial bearing on the binary outcome of the therapeutic response to 8-aminoquinolines i.e. relapse or no relapse. Very high relapse rates usually result from multiple inoculations (e.g. in soldiers fighting in the Pacific theatre of World War 2) (9). Overall, the relapse prevention (i.e. radical curative) efficacy of 8-aminoquinolines was substantially better in clinical practice compared with the stern test applied in the volunteer infections. For example, the combination of quinine and plasmoquine at doses ≥ 30mg base/day was considered highly effective in preventing relapse in clinical studies in endemic areas, but doses below 90mg base/day were relatively ineffective in the experimental challenge model (2, 3, 10, 11). Thus, to compare radical curative efficacies it is necessary to consider both drug dosage, and thus drug exposure, and also the likely hypnozoite burden (12).

Predictors of relapse

Several different factors determine whether a P. vivax or P. ovale malaria infection will relapse. The hypnozoite burden, geographic origin (i.e. “strain”), and degree of immunity are all important determinants. Liver stage immunity may affect hepatic schizont development, and blood stage immunity suppresses the relapse parasitaemia to an extent that it can be asymptomatic, subpatent, or both (13, 14). Although there is no evidence of acquired 8-aminoquinoline resistance in hypnozoites, parasites do differ in intrinsic susceptibilities. For example, the long latency P. vivax “strains” prevalent in temperate regions are more susceptible to 8-aminoquinolines than tropical frequent relapse strains, and P. vivax parasites in Southeast Asia and Oceania appear to be less susceptible (i.e. require a larger primaquine dosage) than other tropical “strains” (15).

From a therapeutic perspective exposure to the biologically active oxidant metabolites of the 8-aminoquinoline is the critical factor determining relapse prevention (radical cure) efficacy (16, 17). The same metabolites are thought to cause haemolysis in G6PD deficiency. This exposure results from the total dose absorbed, and the degree of biotransformation to the active metabolites. Patients with cytochrome P450 2D6 polymorphisms which confer reduced function have reduced production of primaquine’s bioactive metabolite(s) and correspondingly reduced radical curative efficacy (18,19). The oxidative activity of the 8-aminoquinoline treatment is also reflected in the intraerythrocytic concentrations of methaemoglobin. The association of methaemoglobinaemia with radical curative efficacy of 8-aminoquinolines is discussed here.

Methaemoglobinaemia

Methaemoglobin results when the haem iron in haemoglobin is converted from the ferrous state (Fe2+), the reduced form, to the (Fe3+) ferric ion, the oxidized form (20). This process is coupled to redox cycles in the red cell. In the main cycle, driven by the NAD-cytochrome b5 reductase (the main methaemoglobin reductase), haemoglobin and methaemoglobin are cycled (Figure 1). In the second, a cell redox cycle system is driven by the oxidation of haemoglobin, with methaemoglobin as the product. Although the main enzyme responsible for intraerythrocytic methaemoglobin reduction is the NADH-cytochrome b5-reductase, there are alternative pathways. These include an NADPH-dependent methaemoglobin reductase (which has substantially reduced activity in G6PD deficiency), and direct reduction by intracellular ascorbate and glutathione (Figure 1). Methaemoglobin reduction is a first order process. Under normal steady state conditions approximately 3% of the body’s haemoglobin is oxidized each day to methaemoglobin but, because of back-conversion, the average proportion of methaemoglobin is less than 2% of the corresponding haemoglobin concentration. Numerous factors influence this balance (including foods, drugs, exercise, smoking, hypoxia).

Figure 1.

Figure 1

Intraerythrocytic haemoglobin - methaemoglobin interconversion. Each day approximately 3% of the red cells’ haemoglobin is oxidized to methaemoglobin. The major factor contributing to reduction of methaemoglobin back to haemoglobin is the activity of the red cell NADH cytochrome B5 MetHb reductase. MetHb: methaemoglobin.

Methaemoglobin is a dark blue-brown (“chocolate”) colour compared with the bright red of oxygenated haemoglobin, so the skin colouration in methaemoglobinaemia resembles that in cyanosis caused by increased concentrations of deoxygenated haemoglobin. Methaemoglobin has a distinct spectrum from haemoglobin. Methaemoglobinaemia can therefore be measured by spectrophotometry of fresh blood samples in the laboratory or, utilizing the same principle, by continuous transcutaneous oximetry devices. During exposure to the oxidizing agent methaemoglobinaemia increases, although there is substantial interindividual variation in iatrogenic methaemoglobinaemia with a skew distribution of steady state values. Interquartile ranges typically extend from approximately 60 to 140% of the median values and, for a given drug exposure, individual values can range tenfold in large series. With daily primaquine dosing methaemoglobinaemia has an estimated elimination half-life of approximately 1.5 days (Figure 2) i.e. 90% of the new steady state is reached in approximately 6 days.

Figure 2.

Figure 2

The figures show increasing levels of methaemoglobinaemia (as a proportion of the corresponding haemoglobin concentration) during radical curative treatment of vivax malaria with primaquine. On the left (Figure 2A) primaquine 30mg base/day was given to adults for 14 days either concurrently with quinine, or 28 days after a treatment dose of dihydroartemisinin-piperaquine (from Sutanto et al (21)). On the right (Figure 2B) primaquine was given to adults and children either at a dose of 0.5mg base/kg/day for 14 days or 1mg/kg/day for 7 days and patients were randomized to receive concurrent chloroquine or dihydroartemisinin-piperaquine (from Chu et al (22))

Several different oxidant chemicals and drugs can cause methaemoglobinaemia. Although methaemoglobin production (i.e. haem oxidation) by 8-aminoquinolines is generally considered not to lie in the causal pathway to haemolysis or antimalarial activity (26), it does provide an approximate correlate of these activities within the 8-aminoquinoline class. Under the rigorous test of the experimental P. vivax challenge studies conducted in the USA in the late 1940s and early 1950s, drugs or drug concentrations which produced less than 6% steady state methaemoglobinaemia were associated with sub-optimal radical cure rates (2325) (Figure 3). As for iatrogenic haemolysis, there is a slight delay before intraerythrocytic methaemoglobin levels begin to rise following 8-aminoquinoline administration. This presumably reflects depletion of the intrerythrocytic oxidant defences (notably reduced glutathione) (27). In a large recent study of vivax malaria, conducted on the Thailand-Myanmar border, the relationships between primaquine and carboxyprimaquine plasma concentrations, relapses, CYP2D6 polymorphisms and methaemoglobinaemia were investigated. After adjusting for age and partner drug, the day 7 concentrations of primaquine and carboxyprimaquine were not associated with the risk of recurrence, but methaemoglobinaemia was. Higher levels of methaemoglobinaemia were associated with reduced recurrences; a 1% absolute increase in day 7 methemoglobin was associated with a hazard ratio for recurrence of 0.9 (95% CI: 0.85-0.99, p=0.02) (28).

Figure 3.

Figure 3

During the clinical investigations conducted over 70 years ago in the USA to develop new 8-aminoquinolines 18 different compounds were evaluated in addition to pamaquine, and later primaquine (2325). Each was administered for 14 days concurrently with quinine. The volunteers were infected by multiple bites of A. quadrimaculatus infected with the Chesson strain of P. vivax. The figure shows the relationship between the radical cure proportion and the average blood methaemoglobin concentration (%) measured in the last 4 days of treatment. The size of the circles is proportional to the number of subjects recruited as shown in the inset box.

Tafenoquine is a slowly eliminated 8-aminoquinoline which has been introduced recently. At the currently recommended dose (adult dose 300mg) tafenoquine provides radical cure rates which are significantly inferior to the lower dose of primaquine (total dose; 3.5mg/kg) in South-East Asia (29). It is notable that this tafenoquine dose is associated with methaemoglobin concentrations which approximately half those associated with the lower dose primaquine regimen (30,31) and one third of those associated with the currently recommended primaquine radical cure regimen (total dose; 7mg/kg) in the region (Figure 4).

Figure 4.

Figure 4

Dose-response relationships for primaquine and tafenoquine in generating methaemoglobinaemia (21,22, 2426, 2832). Peak values are shown. The dose shown is the daily adult dose of primaquine, whereas for tafenoquine the dose shown is the total dose administered. Primaquine was administered with chloroquine or quinine, tafenoquine was administered with chloroquine or alone. The radical cure rates in Southeast Asia in the two large randomized trials (29,42) are shown, in which the adult primaquine dose was 15mg base/day for two weeks versus a single 300mg dose of tafenoquine. The corresponding average peak methaemoglobin levels from these two trials are indicated by arrows. The size of the circles is proportional to the number of patients or volunteers recruited as shown in the inset box.

Drug interactions

The 8-aminoquinolines have important pharmacokinetic and pharmacodynamic interactions with other structurally related antimalarial drugs. Coadministration of mepacrine (atebrin, quinacrine) substantially increases plasma concentrations of pamaquine and also reduces tolerability. It also substantially increases the methaemoglobinaemia associated with pamaquine (33). Chloroquine, piperaquine and pyronaridine all increase plasma concentrations of primaquine by approximately 20%, but they do not affect tolerability (34). Pamaquine and primaquine radical curative activity is increased by concomitant (but not sequential) administration with quinine (35, 36). Chloroquine has also been shown to potentiate the radical curative efficacy of primaquine. During the clinical development of primaquine it was observed that coadministration of primaquine with quinine or chloroquine attenuated the consequent methaemoglobinaemia. This was not observed in comparable studies with pamaquine (plasmoquine) (Figure 5). However larger, more recent, studies have not confirmed this early finding (Figure 6). They do not suggest any substantial differences between methaemoglobin levels with or without concomitant medicines (quinine, chloroquine, artesunate, artesunate-pyronaridine, and dihydroartemisinin-piperaquine have all been evaluated), nor do they suggest differences between methemoglobin levels in the treatment of malaria (in which steady state methaemoglobin levels are reached well after resolution of symptoms), or in healthy subjects. Thus the impact of blood schizontocides on 8-aminoquinoline induced methaemoglobinaemia is specific to the drugs and is not a general class effect.

Figure 5.

Figure 5

Studies conducted during the development of primaquine which assessed the average methaemoglobin concentrations (%) as a proportion of the haemoglobin concentrations in the last 4 days of treatment in relation to the dosing of plasmoquine (pamaquine) (left) and primaquine (right) (11,33,37). These initial observations suggested that methaemoglobinaemia following primaquine, but not plasmoquine, was attenuated by concomitant administration of chloroquine or quinine. However, later studies (summarized in Figure 6) did not confirm this. The size of the circles is proportional to the number of patients or volunteers recruited as shown in the inset box.

Figure 6.

Figure 6

More recent studies in which methaemoglobinaemia has been assessed after different dose regimens of primaquine whether given alone (26,3941), or in combination with antimalarial drugs (18, 34, 37, 38), either to healthy volunteers, in antimalarial prophylaxis, or in the treatment of vivax malaria (15,19,21,22,2426,2832,37,42,43). These do not suggest attenuation of primaquine induced methaemoglobinaemia by concomitant antimalarial drugs (as reported by Clayman et al (37)), nor are there differences between healthy subjects and patients with malaria. The size of the circles is proportional to the number of patients or volunteers recruited, as shown in the inset box.

Methaemoglobinaemia and G6PD deficiency

The discrepancy between 8-aminoquinoline haemolytic toxicity and methaemoglobinemia has been noted widely (26). In the research which led to registration of primaquine, Edgcomb et al (24) noted no correlation between haemolysis and methaemoglobinaemia following pamaquine or primaquine. The erythrocytes in G6PD deficiency have a reduced ability to reduce methaemoglobin in the presence of “electron-donors”, such as the bioactive metabolites of primaquine or methylene blue (this is the basis of the methylene blue methaemoglobin reduction test developed to diagnose G6PD deficiency). This is because of the reduced intraerythrocytic activity of the NADPH dependent methemoglobin reductase. Brewer et al showed that whereas G6PD deficient individuals had increased levels of methaemoglobinaemia following oral sodium nitrite (an oxidizing agent which does not cause haemolysis), they had decreased levels of methaemoglobinaemia following primaquine (26) (Figure 7). This apparent paradox was explained by the iatrogenic haemolysis of the older erythrocytes which contained the highest concentrations of methaemoglobin. Other mechanisms are also possible, such as the sequestration of the primaquine oxidant metabolites in the oxidized haemoglobin Heinz bodies. The mechanism of 8-aminoquinoline haemolytic toxicity has not been explained satisfactorily and, although increased oxidant stress is implicated, there are clearly other processes also involved.

Figure 7. Mean values of haemoglobin (g/dL) and methaemoglobin (%) in 12 normal and 12 African A-G6PD deficient adult volunteers given primaquine (from Brewer et al (26)).

Figure 7

Conclusions

Methaemoglobinaemia is associated with exposure to the biologically active metabolites produced by therapeutic doses of 8-aminoquinoline drugs. The proportion of methaemoglobin to haemoglobin in blood correlates with the efficacy of anti-relapse therapy, but methaemoglobin appears to be uninvolved directly either in that activity or in haemolytic toxicity in G6PD-deficient patients. This suggests that the oxidative processes which result in methaemoglobinaemia are necessary, but they are not sufficient, for the radical curative activity of the 8-aminoquinoline antimalarial drugs. Although there is substantial inter-individual variation in iatrogenic methaemoglobinaemia, the overall clinical data suggest that methaemoglobinaemia is a pharmacodynamic correlate of radical curative activity. Measurement of methaemoglobinaemia might be useful in drug screening and dose evaluation as a prelude to definitive phase 3 studies characterizing safety and efficacy

• What is Already Known About This Subject

The 8-aminoquinoline antimalarials are the only drugs which can prevent relapses of P. vivax and P. ovale malaria (radical cure), but no pharmacodynamic correlates of this activity have been identified. The 8-aminoquinolines cause dose dependent methaemoglobinaemia, and they cause haemolysis in patients with G6PD deficiency.

• What This Study Adds

Review of data accrued over the past 70 years indicates that methaemoglobinaemia is correlated with radical curative activity. Methaemoglobinaemia could be used to optimize 8-aminoquinoline antimalarial dose regimens.

Acknowledgements

We are very grateful to our many colleagues who have contributed extensively to the clinical studies.

Funding

Wellcome Trust

Footnotes

Declaration of Interests. We have no conflicts of interest.

Ethics approval and consent to participate. Not relevant.

Availability of data and material

All data used are available from the original publications.

References

  • 1.Sinton JA. Bird W: Studies in malaria with special reference to treatment; plasmoquine in treatment of malaria. Indian J Med Research. 1928;16:159. [Google Scholar]
  • 2.Sinton JA, Smith S, Potinger D. Studies in malaria, with special reference to treatment XII Further researches into treatment of chronic benign tertian malaria with plasmoquine and quinine. Indian J Med Res. 1930;17:793. [Google Scholar]
  • 3.Orachowatz D. Toxicity, cyanosis induced by plasmochin treatment of malaria. Arch f Schiffs u Tropen Hyg. 1928;32:119–21. [Google Scholar]
  • 4.Hardgrove M, Applebaum IL. Plasmochin toxicity; analysis of 258 cases. Ann Intern Med. 1946;25:103–12. doi: 10.7326/0003-4819-25-1-103. [DOI] [PubMed] [Google Scholar]
  • 5.Recht J, Ashley EA, White NJ. Safety of 8-aminoquinoline antimalarial medicines. Geneva: World Health Organization; 2014. [Google Scholar]
  • 6.Baird JK. 8-Aminoquinoline therapy for latent malaria. Clin Microbiol Rev. 2019;32:e00011–19. doi: 10.1128/CMR.00011-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Alving AS, Craige B, Pullman TN, Whorton CM, Jones R, Eichelberger L. Procedures used at Stateville Penitentiary for the testing of potential antimalarial agents. J Clin Invest. 1948;27(Suppl 2) doi: 10.1172/JCI101956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Boyd MF. The influence of sporozoite dosage in vivax malaria. Am J Trop Med. 1940;20:279–86. [Google Scholar]
  • 9.Hill E, Amatuzio DS. Southwest Pacific vivax malaria; clinical features and observations concerning duration of clinical activity. Am J Trop Med Hyg. 1949;29:203–14. [PubMed] [Google Scholar]
  • 10.Most H, Kane CA, Lavietes PH, London IM, Schroeder EF, Hayman DM. Combined quinine-plasmochin treatment of vivax malaria; effect of relapse rate. Am J Med Sci. 1946;212:550–60. doi: 10.1097/00000441-194611000-00007. [DOI] [PubMed] [Google Scholar]
  • 11.Craige B, Jones R, Whorton M, Pullman TN, Alving AS, Eichelberger L. Clinical standardization of Pamaquin (Plasmochin) in mosquito-induced vivax malaria, Chesson strain, a preliminary report. J Clin Invest. 1948;27:309–315. [Google Scholar]
  • 12.White NJ. Anti-malarial drug effects on parasite dynamics in vivax malaria. Malar J. 2021;20:161. doi: 10.1186/s12936-021-03700-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Christianson HB, Gordon HH, Daniels WB, Lippincott SW. Afebrile parasitemia in imported vivax malaria. Am J Publ Hlth. 1946;36:759–761. [PMC free article] [PubMed] [Google Scholar]
  • 14.Chaumeau V, Kajeechiwa L, Fustec B, Landier J, Nyo SN, Hsel SN, Phatharakokordbun P, Kittiphanakun P, Nosten S, Thwin MM, Tun SW, et al. The contribution of asymptomatic Plasmodium infections to the transmission of malaria in Kayin state, Myanmar. J Infect Dis. 2019;219:1499–1509. doi: 10.1093/infdis/jiy686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Alving AS, Hankey DD, Coatney GR, Jones R, Jr, Coker WG, Garrison PL, Donovan WN. Korean vivax malaria. II. Curative treatment with pamaquine and primaquine. Am J Trop Med Hyg. 1953;2:970–6. [PubMed] [Google Scholar]
  • 16.Pybus BS, Sousa JC, Jin X, Ferguson JA, Christian RE, Barnhart R, Vuong C, Sciotti RJ, Reichard GA, Kozar MP, Walker LA, et al. CYP450 phenotyping and accurate mass identification of metabolites of the 8-aminoquinoline, anti-malarial drug primaquine. Malar J. 2012;11:259. doi: 10.1186/1475-2875-11-259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Camarda G, Jirawatcharadech P, Priestley RS, Saif A, March S, Wong MHL, Leung S, Miller AB, Baker DA, Alano P, Paine MJI, et al. Antimalarial activity of primaquine operates via a two-step biochemical relay. Nat Commun. 2019;10:e3226. doi: 10.1038/s41467-019-11239-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bennett JW, Pybus BS, Yadava A, Tosh D, Sousa JC, McCarthy WF, Deye G, Melendez V, Ockenhouse CF. Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med. 2013;369:13812. doi: 10.1056/NEJMc1301936. [DOI] [PubMed] [Google Scholar]
  • 19.Baird JK, Louisa M, Noviyanti R, Ekawati L, Elyazar I, Subekti D, Chand K, Gayatri A, Instiaty, Soebianto S, Crenna-Darusallam C, et al. Association of Impaired Cytochrome P450 2D6 Activity Genotype and Phenotype with Therapeutic Efficacy of Primaquine Treatment for Latent Plasmodium vivax Malaria. JAMA Netw Open. 2018;1:e181449. doi: 10.1001/jamanetworkopen.2018.1449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Umbreit J. Methemoglobin--it’s not just blue: a concise review. Am J Hematol. 2007;82:134–44. doi: 10.1002/ajh.20738. [DOI] [PubMed] [Google Scholar]
  • 21.Sutanto I, Tjahjono B, Basri H, Taylor WR, Putri FA, Meilia RA, Setiabudy R, Nurleila S, Ekawati LL, Elyazar I, Farrar J, et al. Randomized, open-label trial of primaquine against vivax malaria relapse in Indonesia. Antimicrob Agents Chemother. 2013;57:1128–35. doi: 10.1128/AAC.01879-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chu CS, Phyo AP, Turner C, Win HH, Poe NP, Yotyingaphiram W, Thinraow S, Wilairisak P, Raksapraidee R, Carrara VI, Paw MK, et al. Chloroquine versus dihydroartemisinin-piperaquine with standard high-dose primaquine given either for 7 days or 14 days in Plasmodium vivax malaria. Clin Infect Dis. 2019;68:1311–9. doi: 10.1093/cid/ciy735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Alving AS, Pullman TN, Craige B, Jones R, Whorton CM, Eichelberger L. The clinical trial of eighteen analogues of pamaquin (plasmochin) in vivax malaria (Chesson strain) J Clin Invest. 1948;27:34–45. doi: 10.1172/JCI101963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Edgcomb JH, Arnold J, Yount EH, Alving AS, Eichelberger L, Jeffery G, et al. Primaquine, SN 13272, a new curative agent in vivax malaria; a preliminary report. J Natl Malar Soc. 1950;9:285–92. [PubMed] [Google Scholar]
  • 25.Cooper WC, Myatt AV, Hernandez T, Jeffery GM, Coatney GR. Studies in human malaria. XXXI. Comparison of primaquine, isopentaquine, SN-3883, and pamaquine as curative agents against Chesson strain vivax malaria. Am J Trop Med Hyg. 1953;2:949–57. [PubMed] [Google Scholar]
  • 26.Brewer GJ, Tarlov AR, Kellermeyer RW, Alving AS. The hemolytic effect of primaquine XV Role of methemoglobin. J Lab Clin Med. 1962;59:905–17. [PubMed] [Google Scholar]
  • 27.Beutler E. G6PD deficiency. Blood. 1994;84:3613–36. [PubMed] [Google Scholar]
  • 28.Chu CS, Watson JA, Phyo AP, Win HH, Yotyingaphiram W, Thinraow S, Soe NL, Aung AA, Wilaisrisak P, Kraft K, Imwong M, et al. Determinants of primaquine and carboxyprimaquine exposures in children and adults with Plasmodium vivax malaria. Antimicrob Agents Chemother. 2021;65:e0130221. doi: 10.1128/AAC.01302-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Llanos-Cuentas A, Lacerda MVG, Hien TT, Vélez ID, Namaik-Larp C, Chu CS, et al. Tafenoquine versus primaquine to prevent relapse of Plasmodium vivax malaria. N Engl J Med. 2019;380:229–41. doi: 10.1056/NEJMoa1802537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Walsh DS, Wilairatana P, Tang DB, Heppner DG, Jr, Brewer TG, Krudsood S, Silachamroon U, Phumratanaprapin W, Siriyanonda D, Looareesuwan S. Randomized trial of 3-dose regimens of tafenoquine (WR238605) versus low-dose primaquine for preventing Plasmodium vivax malaria relapse. Clin Infect Dis. 2004;39:1095–103. doi: 10.1086/424508. [DOI] [PubMed] [Google Scholar]
  • 31.Llanos-Cuentas A, Lacerda MV, Rueangweerayut R, Krudsood S, Gupta SK, Kochar SK, Arthur P, Chuenchom N, Möhrle JJ, Duparc S, Ugwuegbulam C, et al. Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study. Lancet. 2014;383:1049–58. doi: 10.1016/S0140-6736(13)62568-4. [DOI] [PubMed] [Google Scholar]
  • 32.Nelwan EJ, Ekawati LL, Tjahjono B, Setiabudy R, Sutanto I, Chand K, Ekasari T, Djoko D, Basri H, Taylor WR, Duparc S, 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. doi: 10.1186/s12916-015-0535-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Earle DP, Jr, Bigelow FS, Zubrod CH, Kane C. Studies on the chemotherapy of the human malarias. IX. Effect of pamaquine on the blood cells of man. J Clin Invest. 1948;27:121–129. doi: 10.1172/JCI101950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chairat K, Jittamala P, Hanboonkunupakarn B, Pukrittayakamee S, Hanpithakpong W, Blessborn D, White NJ, Day NPJ, Tarning J. Enantiospecific pharmacokinetics and drug-drug interactions of primaquine and blood-stage antimalarial drugs. J Antimicrob Chemother. 2018;73:3102–3113. doi: 10.1093/jac/dky297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Berliner RW, Earle DP, Jr, Taggart JV, Welch WJ, Zubrod CG, Knowlton P, Atchley JA, Shannon JA. Studies on the chemotherapy of the human malarias VII The antimalarial activity of pamaquine. J Clin Invest. 1948;27:108–113. doi: 10.1172/JCI101947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Alving AS, Arnold J, Hockwald RS, Clayman CB, Dern RJ, Beutler E, Flanagan CL. Potentiation of the curative action of primaquine in vivax malaria by quinine and chloroquine. J Lab Clin Med. 1955;46:301–306. [PubMed] [Google Scholar]
  • 37.Clayman CB, Arnold J, Hockwald RS, Yount EH, Edgcomb JH, Alving AS. Status of primaquine III Toxicity of primaquine in Caucasians. JAMA. 1952;149:1563–1568. doi: 10.1001/jama.1952.72930340022010b. [DOI] [PubMed] [Google Scholar]
  • 38.Finch CA. Methemoglobinemia and sulfhemoglobinemia. N Engl J Med. 1948;239:470–8. doi: 10.1056/NEJM194809232391305. [DOI] [PubMed] [Google Scholar]
  • 39.Fletcher KA, Evans DA, Gilles HM, Greaves J, Bunnag D, Harinasuta T. Studies on the pharmacokinetics of primaquine. Bull World Health Organ. 1981;59:407–12. [PMC free article] [PubMed] [Google Scholar]
  • 40.Fryauff DJ, Baird JK, Basri H, Sumawinata I, Purnomo, Richie TL, Ohrt CK, Mouzin E, Church CJ, Richards AL, et al. Randomised placebo-controlled trial of primaquine for prophylaxis of falciparum and vivax malaria. Lancet. 1995;346:1190–3. doi: 10.1016/s0140-6736(95)92898-7. [DOI] [PubMed] [Google Scholar]
  • 41.Baird JK, Lacy MD, Basri H, Barcus MJ, Maguire JD, Bangs MJ, Gramzinski R, Sismadi P, Krisin Ling J, Wiady I, Kusumaningsih M, et al. Randomized, parallel placebo-controlled trial of primaquine for malaria prophylaxis in Papua Indonesia. Clin Infect Dis. 2001;33:1990–7. doi: 10.1086/324085. [DOI] [PubMed] [Google Scholar]
  • 42.Lacerda MVG, Llanos-Cuentas A, Krudsood S, Lon C, Saunders DL, Mohammed R, Yilma D, Batista Pereira D, Espino FEJ, Mia RZ, Chuquiyauri R, et al. Single-Dose Tafenoquine to Prevent Relapse of Plasmodium vivax Malaria. N Engl J Med. 2019;380:215–228. doi: 10.1056/NEJMoa1710775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nelwan EJ, Ekawati LL, Tjahjono B, Setiabudy R, Sutanto I, Chand K, Ekasari T, Djoko D, Basri H, Taylor WR, Duparc S, 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. doi: 10.1186/s12916-015-0535-9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

All data used are available from the original publications.

RESOURCES