loading page

Population Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics Modeling of Oxypurinol in Hmong Adults with Gout and/or Hyperuricemia
  • +2
  • Ya-Feng Wen,
  • Richard Brundage,
  • Youssef Roman,
  • Kathleen Culhane-Pera,
  • Robert Straka
Ya-Feng Wen
University of Minnesota Twin Cities

Corresponding Author:[email protected]

Author Profile
Richard Brundage
University of Minnesota Twin Cities
Author Profile
Youssef Roman
Virginia Commonwealth University
Author Profile
Kathleen Culhane-Pera
Minnesota Community Care
Author Profile
Robert Straka
University of Minnesota Twin Cities
Author Profile

Abstract

Aim: Quantify identifiable sources of variability, including key pharmacogenetic variants in oxypurinol pharmacokinetics and their pharmacodynamic effect on serum urate. Methods: Hmong participants (n=34) received 100 mg allopurinol twice daily for 7 days followed by 150 mg allopurinol twice daily for 7 days. Serum urates (SU) were obtained pre- and post-allopurinol. Serum oxypurinol concentrations were obtained at times 0, 2, 4, and 6 hours after the last dose. A sequential population pharmacokinetic pharmacodynamics (PKPD) analysis with non-linear mixed-effects modeling evaluated the impact of anthropometrics, concomitant medications, and genetic variants on oxypurinol PKPD parameters. Allopurinol maintenance dose to achieve target SU was simulated based on the final PKPD model. Results: A one-compartment model with first order absorption and elimination best described the oxypurinol concentration-time data. Inhibitory of SU by oxypurinol was described with a direct inhibitory Emax model using steady-state oxypurinol concentrations. Fat-free body mass, estimated creatinine clearance and SLC22A12 rs505802 genotype were found to predict differences in oxypurinol clearance. Oxypurinol concentration required to inhibit 50% of xanthine dehydrogenase activity was affected by PDZK1 rs12129861 genotype. Most individuals with both PDZK1 rs12129861 AA and SLC22A12 rs505802 CC genotypes achieve target SU with allopurinol below the maximum dose, regardless of renal function and body mass. In contrast, individuals with both PDZK1 rs12129861 GG and SLC22A12 rs505802 TT genotypes would require more than the maximum dose, thus alternative medications. Conclusion: The proposed allopurinol dosing guide uses individuals’ fat-free mass, renal function, and SLC22A12 rs505802 and PDZK1 rs12129861 genotypes to achieve target SU.
29 Sep 2022Submitted to British Journal of Clinical Pharmacology
30 Sep 2022Submission Checks Completed
30 Sep 2022Assigned to Editor
05 Oct 2022Reviewer(s) Assigned
26 Oct 2022Review(s) Completed, Editorial Evaluation Pending
26 Oct 2022Editorial Decision: Revise Major
24 Jan 20231st Revision Received
24 Jan 2023Assigned to Editor
24 Jan 2023Submission Checks Completed
24 Jan 2023Review(s) Completed, Editorial Evaluation Pending
03 Feb 2023Reviewer(s) Assigned
03 Mar 2023Editorial Decision: Revise Major
19 Apr 20232nd Revision Received
20 Apr 2023Submission Checks Completed
20 Apr 2023Assigned to Editor
20 Apr 2023Review(s) Completed, Editorial Evaluation Pending
21 Apr 2023Editorial Decision: Revise Minor
04 May 20233rd Revision Received
05 May 2023Submission Checks Completed
05 May 2023Assigned to Editor
05 May 2023Review(s) Completed, Editorial Evaluation Pending
10 May 2023Editorial Decision: Accept