We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Published Online:https://doi.org/10.4155/bio-2020-0214

Background: This paper describes for the first-time analytical procedures established to resolve the challenges associated with simultaneous and direct quantification of ataluren and ataluren-O-1β-acyl glucuronide (AAG) by LC–MS/MS in human plasma and urine matrices. Methodology/results: The plasma quantification method was validated for calibration range of 12.5–12500 ng/ml for ataluren and 6.25–2500 ng/ml for AAG. The urine quantification method was validated for calibration range of 0.01–10 and 1–1000 μg/ml for ataluren and AAG, respectively. Plasma and urine samples were stabilized upon collection and through storage to prevent hydrolysis and acyl migration of AAG. Conclusion: Methods described in this paper enabled successful completion of ataluren clinical pharmacology studies for simultaneous pharmacokinetic assessment of ataluren and AAG.

References

  • 1. Peltz SW, Morsy M, Welch EM, Jacobson A. Ataluren as an agent for therapeutic nonsense suppression. Annu. Rev. Med. 64, 407–425 (2013).
  • 2. Welch EM, Barton ER, Zhuo J et al. PTC124 targets genetic disorders caused by nonsense mutations. Nature 447(7140), 87–91 (2007).
  • 3. Peltz SW, Welch EM, Jacobson A et al. Nonsense suppression activity of PTC124 (ataluren). Proc. Natl Acad. Sci. USA 106(25), E64 (2009).
  • 4. Kayali R, Ku JM, Khitrov G, Jung ME, Prikhodko O, Bertoni C. Read-through compound 13 restores dystrophin expression and improves muscle function in the mdx mouse model for Duchenne muscular dystrophy. Hum. Mol. Genet. 21(18), 4007–4020 (2012).
  • 5. Finkel RS, Flanigan KM, Wong B et al. Phase IIa study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS ONE 8(12), e81302 (2013).
  • 6. Kong R, Ma J, Hwang S et al. Metabolism and disposition of ataluren after oral administration to mice, rats, dogs and humans. Drug Metab. Dispos. 48(4), 317–325 (2020).
  • 7. Kong R, Ma J, Hwang S et al. In vitro metabolism, reaction phenotyping, enzyme kinetics, CYP inhibition and induction potential of ataluren. Pharmacol. Res. Perspect 8(2), e00576 (2020).
  • 8. US FDA. Bioanalytical Method Validation Guidance for Industry (2018).
  • 9. European Medicines Agency. Guideline on Bioanalytical Method Validation (2012).
  • 10. Wenkui Li, Jie Zhang, Francis L, Tse S. Handbook of LC–MS Bioanalysis: Best Practices, Experimental Protocols and Regulations. John Wiley & Sons, Inc, NJ, USA (2013).
  • 11. Stachulski AV, Harding JR, Lindon JC, Maggs JL, Park BK, Wilson ID. Acyl glucuronides: biological activity, chemical reactivity and chemical synthesis. J. Med. Chem. 49(24), 6931–6945 (2006).
  • 12. Vree TB, van-Ewijk-Beneken Kolmer EW, Verwey-van Wissen CP, Hekster YA. Direct gradient reversed phase high-performance liquid chromatographic determination of salicylic acid, with the corresponding glycine and glucuronide conjugates in human plasma and urine. J. Chromatogr. 652(2), 161–170 (1994).
  • 13. Shipkova M, Armstrong VW, Oellerich M, Wieland E. Acyl glucuronide drug metabolites: toxicological and analytical implications. Ther. Drug Monit. 25(1), 1–16 (2003).
  • 14. Liu DQ, Pereira T. Interference of a carbamoyl glucuronide metabolite in quantitative liquid chromatography/tandem mass spectrometry. Rapid Commun. Mass Spectrom. 16(2), 142–146 (2002).
  • 15. Xue YJ, Simmons NJ, Liu J, Unger SE anderson DF, Jenkins RG. Separation of a BMS drug candidate and acyl glucuronide from seven glucuronide positional isomers in rat plasma via high performance liquid chromatography with tandem mass spectrometric detection. Rapid Commun. Mass Spectrom. 20(11), 1776–1786 (2006).