The metabolism of primaquine to its active metabolite is dependent on CYP 2D6

Brandon S Pybus, Sean R Marcsisin, Xiannu Jin, Gregory Deye, Jason C Sousa, Qigui Li, Diana Caridha, Qiang Zeng, Gregory A Reichard, Christian Ockenhouse, Jason Bennett, Larry A Walker, Colin Ohrt, Victor Melendez, Brandon S Pybus, Sean R Marcsisin, Xiannu Jin, Gregory Deye, Jason C Sousa, Qigui Li, Diana Caridha, Qiang Zeng, Gregory A Reichard, Christian Ockenhouse, Jason Bennett, Larry A Walker, Colin Ohrt, Victor Melendez

Abstract

Background: The efficacy of the 8-aminoquinoline (8AQ) drug primaquine (PQ) has been historically linked to CYP-mediated metabolism. Although to date no clear evidence exists in the literature that unambiguously assigns the metabolic pathway or specific metabolites necessary for activity, recent literature suggests a role for CYP 2D6 in the generation of redox active metabolites.

Methods: In the present study, the specific CYP 2D6 inhibitor paroxetine was used to assess its effects on the production of specific phenolic metabolites thought to be involved in PQ efficacy. Further, PQ causal prophylactic (developing liver stage) efficacy against Plasmodium berghei in CYP 2D knockout mice was assessed in comparison with a normal C57 background and with humanized CYP 2D6 mice to determine the direct effects of CYP 2D6 metabolism on PQ activity.

Results: PQ exhibited no activity at 20 or 40 mg/kg in CYP 2D knockout mice, compared to 5/5 cures in normal mice at 20 mg/kg. The activity against developing liver stages was partially restored in humanized CYP 2D6 mice.

Conclusions: These results unambiguously demonstrate that metabolism of PQ by CYP 2D6 is essential for anti-malarial causal prophylaxis efficacy.

Figures

Figure 1
Figure 1
CYP2D6-mediated hydroxylation of primaquine. A. Shown is the MSE fragmentation spectrum for primaquine. The (M + 1) charge state of primaquine is highlighted in blue. The major fragment ions of primaquine produced upon MSE fragmentation are also shown along with their corresponding m/z values and structures. B. MSE fragmentation of hydroxylated primaquine. Fragment ions utilized for localization of the hydroxylation to the quinoline core are indicated in red. It should be noted that MSE fragmentation localizes this transformation to the quinoline core, and that the assignment shown is tentative.
Figure 2
Figure 2
Primaquine metabolism with paroxetine inhibition of CYP2D6. A. Shown is the relative % primaquine remaining after 60-min incubations with CYP2D6 in the absence or presence of varying concentrations of paroxetine. B. Relative % hydroxylated metabolite present remaining after 60-min incubations with CYP2D6 in the absence or presence of varying concentrations of paroxetine. The structures of primaquine and the tentative hydroxylated metabolite are shown in each corresponding panel. Error bars were calculated from average values obtained from duplicate analyses.
Figure 3
Figure 3
Dissemination of malaria parasites by IVIS imaging of C57BL/6 Wild-type (WT), CYP 2D knockout, and humanized CYP 2D knockout/CYP 2D6 knock-in mice at 24-hr and 72-hr post-inoculation with luciferase expressing Plasmodium berghei. WT animals treated with 20 mg/kg x 3 days of oral PQ exhibited no parasitaemia at 24 or 72 hr. All five CYP 2D knockout mice exhibited liver stage parasitaemia at 24 hr which progressed to systemic infection by 72 hr. PQ efficacy was largely restored in mice in which the deleted CYP 2D cluster was replaced with human CYP 2D6.
Figure 4
Figure 4
Parasite dissemination by IVIS imaging of C57BL/6 WT, CYP 2D knockout, and humanized CYP 2D knockout/CYP 2D6 knock-in mice at 24-hr and 72-hr post-inoculation with luciferase expressing Plasmodium berghei.

References

    1. Maude RJ, Socheat D, Nguon C, Saroth P, Dara P, Li G, Song J, Yeung S, Dondorp AM, Day NP, White NJ, White LJ. Optimising strategies for Plasmodium falciparum malaria elimination in Cambodia: primaquine, mass drug administration and artemisinin resistance. PLoS One. 2012;7:e37166. doi: 10.1371/journal.pone.0037166.
    1. Vale N, Moreira R, Gomes P. Primaquine revisited six decades after its discovery. Eur J Med Chem. 2009;44:937–953. doi: 10.1016/j.ejmech.2008.08.011.
    1. Tekwani BL, Walker LA. 8-Aminoquinolines: future role as antiprotozoal drugs. Curr Opin Infect Dis. 2006;19:623–631. doi: 10.1097/QCO.0b013e328010b848.
    1. Constantino L, Paixao P, Moreira R, Portela MJ, Do Rosario VE, Iley J. Metabolism of primaquine by liver homogenate fractions: evidence for monoamine oxidase and cytochrome P450 involvement in the oxidative deamination of primaquine to carboxyprimaquine. Exp Toxic Pathol. 1999;51:299–303. doi: 10.1016/S0940-2993(99)80010-4.
    1. Ganesan S, Tekwani BL, Sahu R, Tripathi LM, Walker LA. Cytochrome P450-dependent toxic effects of primaquine on human erythrocytes. Tox App Pharm. 2009;241:14–22. doi: 10.1016/j.taap.2009.07.012.
    1. Pybus BS, Sousa JC, Jin X, Ferguson JA, Christian RE, Barnhart R, Vuong C, Sciotti RJ, Reichard GA, Kozar MP, Walker LA, Ohrt C, Melendez V. 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.
    1. Vasquez-Vivar J, Augusto O. Hydroxylated metabolites of the antimalarial drug primaquine. J Biol Chem. 1992;267:6848–6854.
    1. Idowu OR, Peggins JO, Brewer TG, Kelley C. Metabolism of a candidate 8-aminoquinoline antimalarial agent, WR 238605, by rat liver microsomes. Drug Metab Dispos. 1995;23:1–17.
    1. De Gregori M, Allegri M, De Gregori S, Garbin G, Tinelli C, Regazzi M, Govoni S, Ranzani GN. How and why to screen for CYP2D6 interindividual variability in patients under pharmacological treatments. Current Drug Metabolism. 2010;11:276–282. doi: 10.2174/138920010791196274.
    1. Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3:229–243. doi: 10.1517/14622416.3.2.229.
    1. Scheer N, Kapelyukh Y, McEwan J, Beuger V, Stanley LA, Rode A, Wolf CR. Modeling human cytochrome P450 2D6 metabolism and drug-drug interaction by a novel panel of knockout and humanized mouse lines. Mol Pharmacol. 2012;81:63–72. doi: 10.1124/mol.111.075192.
    1. Organization WH, editor. WHO. Guidelines for the treatment of malaria. 2. 2010. .
    1. Eziefula AC, Gosling R, Hwang J, Hsiang MS, Bousema T, Von Seidlein L, Drakeley C. Primaquine in Africa Discussion G. Rationale for short course primaquine in Africa to interrupt malaria transmission. Malar J. 2012;11:360. doi: 10.1186/1475-2875-11-360.
    1. Graves PM, Gelband H, Garner P. Primaquine for reducing Plasmodium falciparum transmission. Cochrane Database Syst Rev. 2012;9:CD008152.
    1. Myint HY, Berman J, Walker L, Pybus B, Melendez V, Baird JK, Ohrt C. Review: Improving the therapeutic index of 8-aminoquinolines by the use of drug combinations: review of the literature and proposal for future investigations. Am J Trop Med Hyg. 2011;85:1010–1014. doi: 10.4269/ajtmh.2011.11-0498.
    1. Bolchoz LJ, Gelasco AK, Jollow DJ, McMillan DC. Primaquine-induced hemolytic anemia: formation of free radicals in rat erythrocytes exposed to 6-methoxy-8-hydroxylaminoquinoline. J Pharmacol Exp Ther. 2002;303:1121–1129. doi: 10.1124/jpet.102.041459.
    1. Bolchoz LJ, Morrow JD, Jollow DJ, McMillan DC. Primaquine-induced hemolytic anemia: effect of 6-methoxy-8-hydroxylaminoquinoline on rat erythrocyte sulfhydryl status, membrane lipids, cytoskeletal proteins, and morphology. J Pharmacol Exp Ther. 2002;303:141–148. doi: 10.1124/jpet.102.036921.
    1. Bolchoz LJ, Budinsky RA, McMillan DC, Jollow DJ. Primaquine-induced hemolytic anemia: formation and hemotoxicity of the arylhydroxylamine metabolite 6-methoxy-8-hydroxylaminoquinoline. J Pharmacol Exp Ther. 2001;297:509–515.
    1. Bowman ZS, Oatis JE Jr, Whelan JL, Jollow DJ, McMillan DC. Primaquine induced hemolytic anemia: susceptibility of normal versus glutathione-depleted rat erythrocytes to 5-hydroxyprimaquine. J Pharmacol Exp Ther. 2004;309:79–85. doi: 10.1124/jpet.103.062984.
    1. Bowman ZS, Jollow DJ, McMillan DC. Primaquine-induced hemolytic anemia: role of splenic macrophages in the fate of 5-hydroxyprimaquine treated rat erythrocytes. J Pharm Exp Ther. 2005;315:980–986. doi: 10.1124/jpet.105.090407.
    1. Link CM, Theoharides AD, Anders JC, Chung H, Canfield CJ. Structure-activity relationships of putative primaquine metabolites causing methemoglobin formation in canine hemolysates. Toxicol Appl Pharmacol. 1985;81:192–202. doi: 10.1016/0041-008X(85)90155-3.
    1. Yu AM, Idle JR, Gonzalez FJ. Polymorphic cytochrome P450 2D6: humanized mouse model and endogenous substrates. Drug Metab Rev. 2004;36:243–277. doi: 10.1081/DMR-120034000.
    1. Zanger UM, Raimundo S, Eichelbaum M. Cytochrome P450 2D6: overview and update on pharmacology, genetics, biochemistry. Naunyn Schmiedebergs Arch Pharmacol. 2004;369:23–37. doi: 10.1007/s00210-003-0832-2.

Source: PubMed

3
订阅