An Open-Label, Phase 1 Study to Assess the Effects of Hepatic Impairment on Pomalidomide Pharmacokinetics

Yan Li, Xiaomin Wang, Liangang Liu, Chengyue Zhang, Diana Gomez, Josephine Reyes, Maria Palmisano, Simon Zhou, Yan Li, Xiaomin Wang, Liangang Liu, Chengyue Zhang, Diana Gomez, Josephine Reyes, Maria Palmisano, Simon Zhou

Abstract

Pomalidomide is an immunomodulatory drug and the dosage of 4 mg per day taken orally on days 1-21 of repeated 28-day cycles has been approved in the European Union and United States to treat patients with relapsed/refractory multiple myeloma. Because pomalidomide is extensively metabolized prior to excretion, a total of 32 subjects (8 healthy subjects in group 1; 8 subjects with severe hepatic impairment in group 2; 8 subjects with moderate hepatic impairment in group 3; and 8 subjects with mild hepatic impairment in group 4) were enrolled in a multicenter, open-label, single-dose study to assess the impact of hepatic impairment on pomalidomide exposure. Following administration of a single oral dose of 4-mg pomalidomide, the geometric mean ratios of pomalidomide total plasma exposures (AUC) were 171.5%, 157.5%, and 151.2% and the geometric mean ratios of pomalidomide plasma peak exposures (Cmax ) were 75.8%, 94.8%, and 94.2% for subjects with severe, moderate, or mild hepatic impairment, respectively, versus healthy subjects. Pomalidomide administered as a single oral 4-mg dose was safe and well tolerated by healthy subjects and subjects with severe, moderate, or mild hepatic impairment. Based on the pharmacokinetic results from this study, the pomalidomide prescribing information approved by the US Food and Drug Administration recommends for patients with mild or moderate hepatic impairment (Child-Pugh classes A or B), a 3-mg starting daily dose (25% dose reduction) and for patients with severe hepatic impairment (Child-Pugh class C), a 2-mg starting daily dose (50% dose reduction).

Keywords: dose recommendation; hepatic impairment; pharmacokinetics; pomalidomide.

© 2018 The Authors. Clinical Pharmacology in Drug Development Published by Wiley Periodicals, Inc. on behalf of The American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Arithmetic mean (± standard deviation) pomalidomide plasma concentrations‐time profiles, by hepatic impairment group (black lines and symbols: subjects with normal hepatic function [n = 8]; blue lines and symbols: subjects with mildly impaired hepatic function [n = 8]; red lines and symbols: subjects with moderately impaired hepatic function [n = 8]; green lines and symbols: subjects with severely impaired hepatic function [n = 8]).

References

    1. Gay F, Mina R, Troia R, Bringhen S. Pharmacokinetic evaluation of pomalidomide for the treatment of myeloma. Expert Opin Drug Metab Toxicol. 2013;9(11):1517‐1527.
    1. Li Y, Zhou S, Hoffmann M, Kumar G, Palmisano M. Modeling and simulation to probe the pharmacokinetic disposition of pomalidomide R‐ and S‐enantiomers. J Pharmacol Exp Ther. 2014;350(2):265–272.
    1. Mitsiades N, Mitsiades CS, Poulaki V, et al. Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: therapeutic implications. Blood. 2002;99(12):4525–4530.
    1. Zhu D, Corral LG, Fleming YW, Stein B. Immunomodulatory drugs Revlimid (lenalidomide) and CC‐4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation. Cancer Immunol Immunother. 2008;57(12):1849–1859.
    1. Gupta D, Treon SP, Shima Y, et al. Adherence of multiple myeloma cells to bone marrow stromal cells upregulates vascular endothelial growth factor secretion: therapeutic applications. Leukemia. 2001;15(12):1950–1961.
    1. Hideshima T, Chauhan D, Shima Y, et al. Thalidomide and its analogs overcome drug resistance of human multiple myeloma cells to conventional therapy. Blood. 2000;96(9):2943–2950.
    1. Lu L, Payvandi F, Wu L, et al. The anti‐cancer drug lenalidomide inhibits angiogenesis and metastasis via multiple inhibitory effects on endothelial cell function in normoxic and hypoxic conditions. Microvasc Res. 2009;77(2):78–86.
    1. Reddy N, Hernandez‐Ilizaliturri FJ, Deeb G, et al. Immunomodulatory drugs stimulate natural killer‐cell function, alter cytokine production by dendritic cells, and inhibit angiogenesis enhancing the anti‐tumour activity of rituximab in vivo. Br J Haematol. 2008;140(1):36–45.
    1. Verhelle D, Corral LG, Wong K, et al. Lenalidomide and CC‐4047 inhibit the proliferation of malignant B cells while expanding normal CD34+ progenitor cells. Cancer Res. 2007;67(2):746–755.
    1. Corral LG, Haslett PA, Muller GW, et al. Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF‐alpha. J Immunol. 1999;163(1):380–386.
    1. Hayashi T, Hideshima T, Akiyama M, et al. Molecular mechanisms whereby immunomodulatory drugs activate natural killer cells: clinical application. Br J Haematol. 2005;128(2):192–203.
    1. Teo SK, Chen Y, Muller GW, et al. Chiral inversion of the second generation IMiD CC‐4047 (ACTIMID) in human plasma and phosphate‐buffered saline. Chirality. 2003;15(4):348–351.
    1. San Miguel J, Weisel K, Moreau P, et al. Pomalidomide plus low‐dose dexamethasone versus high‐dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM‐003): a randomised, open‐label, phase 3 trial. Lancet Oncol. 2013;14(11):1055–1066.
    1. Daver N, Shastri A, Kadia T, et al. Phase II study of pomalidomide in combination with prednisone in patients with myelofibrosis and significant anemia. Leuk Res. 2014;38(9):1126–1129.
    1. Richardson PG, Siegel DS, Vij R, et al. Pomalidomide alone or in combination with low‐dose dexamethasone in relapsed and refractory multiple myeloma: a randomized phase 2 study. Blood. 2014;123(12):1826–1832.
    1. Hoffmann M, Kasserra C, Reyes J, et al. Absorption, metabolism and excretion of [14C]pomalidomide in humans following oral administration. Cancer Chemother Pharmacol. 2013;71(2):489–501.
    1. Dao K, Chtioui H, Lu Y, et al. Pharmacokinetics of pomalidomide in a patient receiving hemodialysis using a high‐cutoff filter. Am J Kidney Dis. 2017;69(4):553–554.
    1. Li Y, Wang X, O'Mara E, et al. Population pharmacokinetics of pomalidomide in patients with relapsed or refractory multiple myeloma with various degrees of impaired renal function. Clin Pharmacol. 2017;9:133–145.
    1. Kasserra C, Assaf M, Hoffmann M, et al. Pomalidomide: evaluation of cytochrome P450 and transporter‐mediated drug‐drug interaction potential in vitro and in healthy subjects. J Clin Pharmacol. 2014;55(2):168–178.
    1. EMA . Guideline on the evaluation of the pharmacokinetics of medicinal products in patients with impaired hepatic function. 2005. EMA/CHMP/83874/2014. . Published December 2015. Accessed April 11, 2018.
    1. FDA . Pharmacokinetics in patients with impaired hepatic function: study design, data analysis, and impact on dosing and labeling. FDA Guidance 2003. . Published May 2003. Accessed April 11, 2018.
    1. Kasserra C, Assaf M, Hoffmann M, et al. Pomalidomide: evaluation of cytochrome P450 and transporter‐mediated drug‐drug interaction potential in vitro and in healthy subjects. J Clin Pharmacol. 2015;55(2):168–178.
    1. Li Y, Xu Y, Liu L, Wang X, Palmisano M, Zhou S. Population pharmacokinetics of pomalidomide. J Clin Pharmacol. 2015;55(5):563–572.

Source: PubMed

3
Iratkozz fel