Meta-Analysis of Noncompartmental Pharmacokinetic Parameters of Ertugliflozin to Evaluate Dose Proportionality and UGT1A9 Polymorphism Effect on Exposure

Jean-Claude Marshall, Yali Liang, Vaishali Sahasrabudhe, Thomas Tensfeldt, Daryl J Fediuk, Susan Zhou, Rajesh Krishna, Vikas Kumar Dawra, Linda S Wood, Kevin Sweeney, Jean-Claude Marshall, Yali Liang, Vaishali Sahasrabudhe, Thomas Tensfeldt, Daryl J Fediuk, Susan Zhou, Rajesh Krishna, Vikas Kumar Dawra, Linda S Wood, Kevin Sweeney

Abstract

Ertugliflozin, a sodium-glucose cotransporter 2 inhibitor, is primarily metabolized via glucuronidation by the uridine 5'-diphospho-glucuronosyltransferase (UGT) isoform UGT1A9. This noncompartmental meta-analysis of ertugliflozin pharmacokinetics evaluated the relationship between ertugliflozin exposure and dose, and the effect of UGT1A9 genotype on ertugliflozin exposure. Pharmacokinetic data from 25 phase 1 studies were pooled. Structural models for dose proportionality described the relationship between ertugliflozin area under the plasma concentration-time curve (AUC) or maximum observed plasma concentration (Cmax ) and dose. A structural model for the UGT1A9 genotype described the relationship between ertugliflozin AUC and dose, with genotype information on 3 UGT1A9 polymorphisms (UGT1A9-2152, UGT1A9*3, UGT1A9*1b) evaluated as covariates from the full model. Ertugliflozin AUC and Cmax increased in a dose-proportional manner over the dose range of 0.5-300 mg, and population-predicted AUC and Cmax values for the 5- and 15-mg ertugliflozin tablets administered in the fasted state demonstrated good agreement with the observed data. The largest change in ertugliflozin AUC was in subjects carrying the UGT1A9*3 heterozygous variant, with population-predicted AUC (90% confidence interval) values of 485 ng·h/mL (458 to 510 ng·h/mL) and 1560 ng·h/mL (1480 to 1630 ng·h/mL) for ertugliflozin 5 and 15 mg, respectively, compared with 436 ng·h/mL (418 to 455 ng·h/mL) and 1410 ng·h/mL (1350 to 1480 ng·h/mL), respectively, in wild-type subjects. Overall, the mean effects of the selected UGT1A9 variants on ertugliflozin AUC were within ±10% of the wild type. UGT1A9 genotype did not have any clinically meaningful effects on ertugliflozin exposure in healthy subjects. No ertugliflozin dose adjustment would be required in patients with the UGT1A9 variants assessed in this study.

Trial registration: ClinicalTrials.gov NCT00989079 NCT01018823 NCT01127308 NCT01114568 NCT01223339 NCT01948986 NCT02115347 NCT02411929.

Keywords: UDP-glucuronosyltransferase 1A9; ertugliflozin; genotype; pharmacogenetics; pharmacokinetics.

Conflict of interest statement

J.‐C.M., V.S., T.T., D.J.F., V.K.D., L.S.W., and K.S. are employees of Pfizer Inc., New York, New York, and may own shares/stock options in Pfizer Inc., New York, New York. Y.L. was an employee of Pfizer Inc., New York, New York, at the time the study was conducted. S.Z. is an employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, New Jersey, and may own stock in Merck & Co., Inc., Kenilworth, New Jersey. R.K. was an employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, New Jersey, at the time the study was conducted and may own stock in Merck & Co., Inc., Kenilworth, New Jersey.

© 2021 The Authors. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Observed ertugliflozin dose‐normalized (A) AUC and (B) Cmax values by dose from the dose‐proportionality analysis. Red circles represent dose‐normalized AUCinf or dose‐normalized Cmax following a single dose; black triangles represent dose‐normalized AUCtau at steady state; geometric mean values for each dose group are represented by blue asterisks. AUC, area under the plasma concentration‐time curve; AUCinf, AUC from time zero extrapolated to infinite time; AUCtau, AUC from time zero to time tau, the dosing interval, for which tau is 24 hours; Cmax, maximum observed plasma concentration; SD, single dose; SS, steady state.
Figure 2
Figure 2
Population‐predicted, individual‐predicted, and observed ertugliflozin (A, B) AUC and (C, D) Cmax values by dose from the full model in the dose‐proportionality analysis. Results are shown across the full dose range (A, C) and across the 0.5‐ to 15‐mg dose range (B, D). AUC, area under the plasma concentration‐time curve; Cmax, maximum observed plasma concentration.
Figure 3
Figure 3
Observed ertugliflozin dose‐normalized AUC values by (A) dose and (B) UGT1A9 genotype from the UGT1A9 genotype analysis. (A) Red circles represent dose‐normalized AUCinf following a single dose; black triangles represent dose‐normalized AUCtau at steady state; geometric means for each dose group are represented by blue asterisks. (B) Open circles represent individual subject dose‐normalized AUC values. Box plot provides median and 25%/75% quartiles with whiskers to the last point within 1.5× interquartile range. AUC, area under the plasma concentration‐time curve; AUCinf, AUC from time zero extrapolated to infinite time; AUCtau, AUC from time zero to time tau, the dosing interval, for which tau is 24 hours; het, heterozygous; hom, homozygous; SD, single dose; SS, steady state; WT, wild type.
Figure 4
Figure 4
UGT1A9 polymorphic effects on ertugliflozin AUC from the UGT1A9 genotype analysis. The 90th percentiles of the bootstrap confidence intervals for AUC are provided. Effects are reported relative to the wild‐type subjects in the analysis. A value of 1 represents no change relative to the wild type. AUC, area under the plasma concentration‐time curve.

References

    1. US Food and Drug Administration. Merck Sharp & Dohme Corp., Whitehouse Station, NJ, USA. Steglatro (ertugliflozin): Prescribing information. Rockville, MD; 2017. . Accessed October 20, 2020.
    1. European Medicines Agency. Merck Sharp & Dohme Ltd, Hoddesdon, UK. Steglatro (ertugliflozin): Summary of product characteristics; 2018. . Accessed October 20, 2020.
    1. Fediuk DJ, Nucci G, Dawra VK, et al. Overview of the clinical pharmacology of ertugliflozin, a novel sodium‐glucose cotransporter 2 (SGLT2) inhibitor. Clin Pharmacokinet. 2020;59(8):949‐965.
    1. Nucci G, Le V, Sweeney K, Amin N. Single‐ and multiple‐dose pharmacokinetics and pharmacodynamics of ertugliflozin, an oral selective inhibitor of SGLT2, in healthy subjects. Clin Pharmacol Ther. 2018;103(S1):S83.
    1. Sahasrabudhe V, Fediuk DJ, Matschke K, et al. Effect of food on the pharmacokinetics of ertugliflozin and its fixed‐dose combinations ertugliflozin/sitagliptin and ertugliflozin/metformin. Clin Pharmacol Drug Dev. 2019;8(5):619‐627.
    1. Dawra VK, Cutler DL, Zhou S, et al. Assessment of the drug interaction potential of ertugliflozin with sitagliptin, metformin, glimepiride, or simvastatin in healthy subjects. Clin Pharmacol Drug Dev. 2018;8(3):314‐325.
    1. Sahasrabudhe V, Terra SG, Hickman A, et al. The effect of renal impairment on the pharmacokinetics and pharmacodynamics of ertugliflozin in subjects with type 2 diabetes mellitus. J Clin Pharmacol. 2017;57(11):1432‐1443.
    1. Sahasrabudhe V, Terra SG, Hickman A, et al. Pharmacokinetics of single‐dose ertugliflozin in patients with hepatic impairment. Clin Ther. 2018;40(10):1701‐1710.
    1. Miao Z, Nucci G, Amin N, et al. Pharmacokinetics, metabolism, and excretion of the antidiabetic agent ertugliflozin (PF‐04971729) in healthy male subjects. Drug Metab Dispos. 2013;41(2):445‐456.
    1. Kalgutkar AS, Tugnait M, Zhu T, et al. Preclinical species and human disposition of PF‐04971729, a selective inhibitor of the sodium‐dependent glucose cotransporter 2 and clinical candidate for the treatment of type 2 diabetes mellitus. Drug Metab Dispos. 2011;39(9):1609‐1619.
    1. Lapham K, Callegari E, Cianfrogna J, et al. In vitro characterization of ertugliflozin metabolism by UDP‐glucuronosyltransferase and cytochrome P450 enzymes. Drug Metab Dispos. 2020;48(12):1350‐1363.
    1. Stingl JC, Bartels H, Viviani R, Lehmann ML, Brockmoller J. Relevance of UDP‐glucuronosyltransferase polymorphisms for drug dosing: A quantitative systematic review. Pharmacol Ther. 2014;141(1):92‐116.
    1. European Medicines Agency. Committee for Medicinal Products for Human Use . Guideline on the use of pharmacogenetic methodologies in the pharmacokinetic evaluation of medicinal products; 2011. . Accessed October 20, 2020.
    1. US Food and Drug Administration . Guidance for industry: Clinical pharmacogenomics: Premarket evaluation in early‐phase clinical studies and recommendations for labeling. Rockville, MD; 2013. . Accessed October 20, 2020.
    1. Girard H, Court MH, Bernard O, et al. Identification of common polymorphisms in the promoter of the UGT1A9 gene: evidence that UGT1A9 protein and activity levels are strongly genetically controlled in the liver. Pharmacogenetics. 2004;14(8):501‐515.
    1. Villeneuve L, Girard H, Fortier LC, Gagne JF, Guillemette C. Novel functional polymorphisms in the UGT1A7 and UGT1A9 glucuronidating enzymes in Caucasian and African‐American subjects and their impact on the metabolism of 7‐ethyl‐10‐hydroxycamptothecin and flavopiridol anticancer drugs. J Pharmacol Exp Ther. 2003;307(1):117‐128.
    1. Yamanaka H, Nakajima M, Katoh M, et al. A novel polymorphism in the promoter region of human UGT1A9 gene (UGT1A9*22) and its effects on the transcriptional activity. Pharmacogenetics. 2004;14(5):329‐332.
    1. Guillemette C, Levesque E, Rouleau M. Pharmacogenomics of human uridine diphospho‐glucuronosyltransferases and clinical implications. Clin Pharmacol Ther. 2014;96(3):324‐339.
    1. Owen RP, Sangkuhl K, Klein TE, Altman RB. Cytochrome P450 2D6. Pharmacogenet Genomics. 2009;19(7):559‐562.
    1. Dawra VK, Liang Y, Shi H, et al. A PK/PD study comparing twice‐daily to once‐daily dosing regimens of ertugliflozin in healthy subjects. Int J Clin Pharmacol Ther. 2019;57(4):207‐216.
    1. Whirl‐Carrillo M, McDonagh EM, Hebert JM, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012;92(4):414‐417.
    1. Amin NB, Wang X, Jain SM, Lee DS, Nucci G, Rusnak JM. Dose‐ranging efficacy and safety study of ertugliflozin, a sodium‐glucose co‐transporter 2 inhibitor, in patients with type 2 diabetes on a background of metformin. Diabetes Obes Metab. 2015;17(6):591‐598.
    1. Amin NB, Wang X, Mitchell JR, Lee DS, Nucci G, Rusnak JM. Blood pressure‐lowering effect of the sodium glucose co‐transporter‐2 inhibitor ertugliflozin, assessed via ambulatory blood pressure monitoring in patients with type 2 diabetes and hypertension. Diabetes Obes Metab. 2015;17(8):805‐808.
    1. Patel S, Hickman A, Frederich R, et al. Safety of ertugliflozin in patients with type 2 diabetes mellitus: pooled analysis of seven phase 3 randomized controlled trials. Diabetes Ther. 2020;11(6):1347‐1367.
    1. Fediuk DJ, Nucci G, Dawra VK, et al. End‐to‐end application of model‐informed drug development for ertugliflozin, a novel sodium‐glucose cotransporter 2 inhibitor. CPT Pharmacometrics Syst Pharmacol. 2021;10(6):529‐542.
    1. Dawra VK, Sahasrabudhe V, Liang Y, et al. Effect of rifampin on the pharmacokinetics of ertugliflozin in healthy subjects. Clin Ther. 2018;40(9):1538‐1547.
    1. Callegari E, Lin J, Tse S, Goosen TC, Sahasrabudhe V. Physiologically‐based pharmacokinetic modeling of the drug‐drug interaction of the UGT substrate ertugliflozin following co‐administration with the UGT inhibitor mefenamic acid. CPT Pharmacometrics Syst Pharmacol. 2021;10(2):127‐136.

Source: PubMed

3
Abonneren