Differential kinetic profiles and metabolism of primaquine enantiomers by human hepatocytes

Pius S Fasinu, Bharathi Avula, Babu L Tekwani, N P Dhammika Nanayakkara, Yan-Hong Wang, H M T Bandara Herath, James D McChesney, Gregory A Reichard, Sean R Marcsisin, Mahmoud A Elsohly, Shabana I Khan, Ikhlas A Khan, Larry A Walker, Pius S Fasinu, Bharathi Avula, Babu L Tekwani, N P Dhammika Nanayakkara, Yan-Hong Wang, H M T Bandara Herath, James D McChesney, Gregory A Reichard, Sean R Marcsisin, Mahmoud A Elsohly, Shabana I Khan, Ikhlas A Khan, Larry A Walker

Abstract

Background: The clinical utility of primaquine (PQ), used as a racemic mixture of two enantiomers, is limited due to metabolism-linked hemolytic toxicity in individuals with genetic deficiency in glucose-6-phosphate dehydrogenase. The current study investigated differential metabolism of PQ enantiomers in light of the suggestions that toxicity and efficacy might be largely enantioselective.

Methods: Stable isotope (13)C-labelled primaquine and its two enantiomers (+)-PQ, (-)-PQ were separately incubated with cryopreserved human hepatocytes. Time-tracked substrate depletion and metabolite production were monitored via UHPLC-MS/MS.

Results: The initial half-life of 217 and 65 min; elimination rate constants (λ) of 0.19 and 0.64 h(-1); intrinsic clearance (Clint) of 2.55 and 8.49 (µL/min)/million cells, which when up-scaled yielded Clint of 6.49 and 21.6 (mL/min)/kg body mass was obtained respectively for (+)- and (-)-PQ. The extrapolation of in vitro intrinsic clearance to in vivo human hepatic blood clearance, performed using the well-stirred liver model, showed that the rate of hepatic clearance of (+)-PQ was only 45 % that of (-)-PQ. Two major primary routes of metabolism were observed-oxidative deamination of the terminal amine and hydroxylations on the quinoline moiety of PQ. The major deaminated metabolite, carboxyprimaquine (CPQ) was preferentially generated from the (-)-PQ. Other deaminated metabolites including PQ terminal alcohol (m/z 261), a cyclized side chain derivative from the aldehyde (m/z 241), cyclized carboxylic acid derivative (m/z 257), a quinone-imine product of hydroxylated CPQ (m/z 289), CPQ glucuronide (m/z 451) and the glucuronide of PQ alcohol (m/z 437) were all preferentially generated from the (-)-PQ. The major quinoline oxidation product (m/z 274) was preferentially generated from (+)-PQ. In addition to the products of the two metabolic pathways, two other major metabolites were observed: a prominent glycosylated conjugate of PQ on the terminal amine (m/z 422), peaking by 30 min and preferentially generated by (+)-PQ; and the carbamoyl glucuronide of PQ (m/z 480) exclusively generated from (+)-PQ.

Conclusion: Metabolism of PQ showed enantioselectivity. These findings may provide important information in establishing clinical differences in PQ enantiomers.

Keywords: Enantioselectivity; Hepatocytes; Malaria; Metabolism; Primaquine enantiomers; Primaquine metabolites.

Figures

Fig. 1
Fig. 1
a Hepatocyte viability time course determined through cell counts in the presence and absence of (±)-primaquine and its (+)- and (−)-enantiomers; b differential depletion of the 20 µM racemic (±)-primaquine and its (+)-, and (−)-enantiomers in primary human hepatocytes (1 million cells/mL) after 2 h incubation. Each point represents values mean ±SD (n = 4)
Fig. 2
Fig. 2
Putative identities and predicted structures of primaquine metabolites generated in human hepatocytes, as determined through MS/MS fragmentation, twin peak detection on the UHPLC chromatogram and prediction by Waters’ Metabolynx® software package
Fig. 3
Fig. 3
The relative concentrations of the major metabolites generated by primaquine and its enantiomers at 1 h incubation time point in primary human hepatocytes
Fig. 4
Fig. 4
A time-course analysis of the in vitro generation of a carboxyprimaquine (1), b primaquine alcohol (2) and c, d their glucuronide conjugates (5, 6), from (+)-, (−)- and (±)-primaquine in human hepatocytes. Each point represents value mean ±SD of four observations
Fig. 5
Fig. 5
A time-course analysis of a cyclized PQ aldehyde (m/z 241) (3), b cyclized carboxyprimaquine (m/z 257) (4), c hydroxylated CPQ quinone-imine (m/z 289) (7) and d hydroxyprimaquine quinone-imine (m/z 274) (8) differentially generated from (+)-, (−)- and (±)-primaquine in vitro in human hepatocytes. Each point represents value mean ±SD of four observations
Fig. 6
Fig. 6
A time-course analysis of a glycosylated primaquine (9) generated through the activity of human hepatocytes; b non-enzymatic generation of primaquine-glucose conjugates observed following the incubation of primaquine and its metabolites in cell-free hepatocyte media and; c primaquine carbamoyl-glucuronide (10) differentially generated from (+)-, (−)-and (±)-primaquine in vitro in human hepatocytes. Each point represents value mean ±SD of four observations

References

    1. Ashley EA, Recht J, White NJ. Primaquine: the risks and the benefits. Malar J. 2014;13:418. doi: 10.1186/1475-2875-13-418.
    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. Hill DR, Baird JK, Parise ME, Lewis LS, Ryan ET, Magill AJ. Primaquine: report from CDC expert meeting on malaria chemoprophylaxis. Am J Trop Med Hyg. 2006;75:402–415.
    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. Fernando D, Rodrigo C, Rajapakse S. Primaquine in vivax malaria: an update and review on management issues. Malar J. 2011;10:351. doi: 10.1186/1475-2875-10-351.
    1. WHO. Global Plan for Artemisinin Resistance Containment (GPARC). Accessed 27 Nov 2015.
    1. Schmidt LH, Alexander S, Allen L, Rasco J. Comparison of the curative antimalarial activities and toxicities of primaquine and its d and l isomers. Antimicrob Agents Chemother. 1977;12:51–60. doi: 10.1128/AAC.12.1.51.
    1. Nanayakkara ND, Tekwani BL, Herath HB, Sahu R, Gettayacamin M, Tungtaeng A, et al. Scalable preparation and differential pharmacologic and toxicologic profiles of primaquine enantiomers. Antimicrob Agents Chemother. 2014;58:4737–4744. doi: 10.1128/AAC.02674-13.
    1. Saunders D, Vanachayangkul P, Imerbsin R, Khemawoot P, Siripokasupkul R, Tekwani BL, et al. Pharmacokinetics and pharmacodynamics of (+)-primaquine and (−)-primaquine enantiomers in rhesus macaques (Macaca mulatta) Antimicrob Agents Chemother. 2014;58:7283–7291. doi: 10.1128/AAC.02576-13.
    1. Bennett JW, Pybus BS, Yadava A, Tosh D, Sousa JC, McCarthy WF, et al. Primaquine failure and cytochrome P-450 2D6 in Plasmodium vivax malaria. N Engl J Med. 2013;369:1381–1382. doi: 10.1056/NEJMc1301936.
    1. Pybus BS, Marcsisin SR, Jin X, Deye G, Sousa JC, Li Q, et al. The metabolism of primaquine to its active metabolite is dependent on CYP 2D6. Malar J. 2013;12:212. doi: 10.1186/1475-2875-12-212.
    1. Fasinu PS, Tekwani BL, Nanayakkara NP, Avula B, Herath HM, Wang YH, et al. Enantioselective metabolism of primaquine by human CYP2D6. Malar J. 2014;13:507. doi: 10.1186/1475-2875-13-507.
    1. Allahyari R, Strother A, Fraser IM, Verbiscar AJ. Synthesis of certain hydroxy analogues of the antimalarial drug primaquine and their in vitro methemoglobin-producing and glutathione-depleting activity in human erythrocytes. J Med Chem. 1984;27:407–410. doi: 10.1021/jm00369a031.
    1. Mihaly GW, Ward SA, Edwards G, Orme ML, Breckenridge AM. Pharmacokinetics of primaquine in man: identification of the carboxylic acid derivative as a major plasma metabolite. Br J Clin Pharmacol. 1984;17:441–446. doi: 10.1111/j.1365-2125.1984.tb02369.x.
    1. Tekwani BL, Avula B, Sahu R, Chaurasiya ND, Khan SI, Jain S, et al. Enantioselective pharmacokinetics of primaquine in healthy human volunteers. Drug Metab Dispos. 2015;43:571–577. doi: 10.1124/dmd.114.061127.
    1. Avula B, Tekwani BL, Chaurasiya ND, Nanayakkara NP, Wang YH, Khan SI, et al. Profiling primaquine metabolites in primary human hepatocytes using UHPLC-QTOF-MS with 13C stable isotope labeling. J Mass Spectrom. 2013;48:276–285. doi: 10.1002/jms.3122.
    1. Herath HM, McChesney JD, Walker LA, Nanayakkara NP. Synthesis of [13C6] primaquine. J Labelled Comp Radiopharm. 2013;56:341–343. doi: 10.1002/jlcr.3039.
    1. McChesney JD, Sarangan S. Synthesis of 8-(3-carboxy-l-methyl-propylammo)-6-methoxyquinoline: a newly characterized primaquine metabolite. Pharm Res. 1984;1:96–98. doi: 10.1023/A:1016311616719.
    1. Hufford CD, Clark AM, Quinones IN, Baker JK, McChesney JD. Microbial metabolism studies on the major microbial and mammalian metabolite of primaquine. J Pharm Sci. 1983;72:92–94. doi: 10.1002/jps.2600720124.
    1. Obach RS. Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos. 1999;27:1350–1359.
    1. Davies B, Morris T. Physiological parameters in laboratory animals and humans. Pharm Res. 1993;10:1093–1095. doi: 10.1023/A:1018943613122.
    1. Potter BM, Xie LH, Vuong C, Zhang J, Zhang P, Duan D, et al. Differential CYP 2D6 metabolism alters primaquine pharmacokinetics. Antimicrob Agents Chemother. 2015;59:2380–2387. doi: 10.1128/AAC.00015-15.
    1. Avula B, Khan SI, Tekwani BL, Nanayakkara NPD, McChesney JD, Walker LA, et al. Analysis of primaquine and its metabolite carboxyprimaquine in biological samples: enantiomeric separation, method validation and quantification. Biomed Chromatogr. 2011;25:1010–1017. doi: 10.1002/bmc.1557.
    1. Ganesan S, Chaurasiya ND, Sahu R, Walker LA, Tekwani BL. Understanding the mechanisms for metabolism-linked hemolytic toxicity of primaquine against glucose 6-phosphate dehydrogenase deficient human erythrocytes: evaluation of eryptotic pathway. Toxicology. 2012;294:54–60. doi: 10.1016/j.tox.2012.01.015.
    1. Potter BM, Xie LH, Vuong C, Zhang J, Zhang P, Duan D, et al. Differential CYP 2D6 metabolism alters primaquine pharmacokinetics. Antimicrob Agents Chemother. 2015;59:2380–2387. doi: 10.1128/AAC.00015-15.
    1. Pybus BS, Sousa JC, Jin X, Ferguson JA, Christian RE, Barnhart R, 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.
    1. Obach RS, Reed-Hagen AE, Krueger SS, Obach BJ, O’Connell TN, Zandi KS, et al. Metabolism and disposition of varenicline, a selective α4β2 acetylcholine receptor partial agonist, in vivo and in vitro. Drug Metab Dispos. 2006;34:121–130. doi: 10.1124/dmd.105.006767.
    1. Sadeque AJ, Usmani KA, Palamar S, Cerny MA, Chen WG. Identification of human UDP-glucuronosyltransferases involved in N-carbamoyl glucuronidation of lorcaserin. Drug Metab Dispos. 2012;40:772–778. doi: 10.1124/dmd.111.043448.

Source: PubMed

3
Abonneren