Pharmacokinetics and Safety of Glasdegib in Participants With Moderate/Severe Hepatic Impairment: A Phase I, Single-Dose, Matched Case-Control Study

Joanna C Masters, Robert R LaBadie, Joanne Salageanu, Jerry Li, Naveed Shaik, Joanna C Masters, Robert R LaBadie, Joanne Salageanu, Jerry Li, Naveed Shaik

Abstract

This phase I open-label trial (NCT03627754) assessed glasdegib pharmacokinetics and safety in otherwise healthy participants with moderate (Child-Pugh B) or severe (Child-Pugh C) hepatic impairment. Participants with hepatic impairment and age/weight-matched controls with normal hepatic function received a single oral 100-mg glasdegib dose under fasted conditions. The primary end points were area under the plasma concentration-time curve from time zero to infinity (AUCinf ) and maximum plasma concentration (Cmax ). Twenty-four participants (8/cohort) were enrolled. Glasdegib plasma exposures in moderate hepatic impairment were similar to controls, with adjusted geometric mean ratios (GMRs) of 110.8% (90% confidence interval [CI], 78.0-157.3) for AUCinf and 94.8% (69.9-128.4) for Cmax versus controls. In severe hepatic impairment, glasdegib plasma exposures were lower than controls (AUCinf GMR, 75.7%; 90%CI, 51.5-111.0; Cmax GMR, 58.0%; 90%CI, 37.8-89.0). Unbound glasdegib exposures were similar to controls for moderate (AUCinf,u GMR, 118.1%; 90%CI, 88.7-157.2; Cmax,u GMR, 101.1%; 90%CI, 78.4-130.3) and severe hepatic impairment (AUCinf,u GMR, 116.3%; 90%CI 81.8-165.5; Cmax,u GMR, 89.2%, 90%CI, 60.2-132.3). No treatment-related adverse events or clinically significant changes in laboratory values, vital signs, or electrocardiograms were observed. Together with previous findings, this suggests glasdegib dose modifications are not required based on hepatic impairment.

Keywords: acute myeloid leukemia; glasdegib; hepatic; oncology; pharmacokinetics.

Conflict of interest statement

All authors are employees of Pfizer. Joanna C. Masters, Robert R. LaBadie, Joanne Salageanu, and Naveed Shaik hold Pfizer stock.

© 2020 Pfizer Inc. Clinical Pharmacology in Drug Development published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Plasma concentration–time profiles for total glasdegib. Concentration values below the lower limit of quantification (3 ng/mL) were set to zero.
Figure 2
Figure 2
Individual and summary of glasdegib total and unbound AUCinf and Cmax. Circles represent individual participant values and stars represent geometric mean. Box plot provides median and 25%/75% quartiles with whiskers to the last data point within 1.5 × the interquartile range. Mean (SD) displayed numerically under box plot. AUCinf, AUC from time zero to infinity; Cmax, maximum plasma concentration; mean, arithmetic mean; SD, standard deviation; u, unbound.
Figure 3
Figure 3
Plasma concentration–time profiles for unbound glasdegib. Concentration values below the lower limit of quantification (3 ng/mL) were set to zero.

References

    1. Döhner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia. N Engl J Med. 2015;373(12):1136‐1152.
    1. American Cancer Society . Cancer Facts & Figures 2020. Atlanta: American Cancer Society; 2020.
    1. Cortes JE, Gutzmer R, Kieran MW, Solomon JA. Hedgehog signaling inhibitors in solid and hematological cancers. Cancer Treat Rev. 2019;76:41‐50.
    1. Munchhof MJ, Li Q, Shavnya A, et al. Discovery of PF‐04449913, a potent and orally bioavailable inhibitor of smoothened. ACS Med Chem Lett. 2012;3(2):106‐111.
    1. Hoy SM. Glasdegib: first global approval. Drugs. 2019;79(2):207‐213.
    1. Cortes JE, Dombret H, Merchant A, et al. Glasdegib plus intensive/nonintensive chemotherapy in untreated acute myeloid leukemia: BRIGHT AML 1019 phase III trials. Future Oncol. 2019;15(31):3531‐3545.
    1. Cortes JE, Douglas Smith B, Wang ES, et al. Glasdegib in combination with cytarabine and daunorubicin in patients with AML or high‐risk MDS: phase 2 study results. Am J Hematol. 2018;93(11):1301‐1310.
    1. Sallman DA, Komrokji RS, Sweet KL, et al. A phase 2 trial of the oral smoothened inhibitor glasdegib in refractory myelodysplastic syndromes (MDS). Leuk Res. 2019;81:56‐61.
    1. Martinelli G, Oehler VG, Papayannidis C, et al. Treatment with PF‐04449913, an oral smoothened antagonist, in patients with myeloid malignancies: a phase 1 safety and pharmacokinetics study. Lancet Haematol. 2015;2(8):e339‐e346.
    1. Lam JL, Vaz A, Hee B, et al. Metabolism, excretion and pharmacokinetics of [(14)C]glasdegib (PF‐04449913) in healthy volunteers following oral administration. Xenobiotica. 2017;47(12):1064‐1076.
    1. Shaik MN, LaBadie RR, Rudin D, Levin WJ. Evaluation of the effect of food and ketoconazole on the pharmacokinetics of the smoothened inhibitor PF‐04449913 in healthy volunteers. Cancer Chemother Pharmacol. 2014;74(2):411‐418.
    1. Shaik MN, Hee B, Wei H, LaBadie RR. Evaluation of the effect of rifampin on the pharmacokinetics of the smoothened inhibitor glasdegib in healthy volunteers. Br J Clin Pharmacol. 2018;84(6):1346‐1353.
    1. Shaik N, Hee B, Wei H, LaBadie RR. Evaluation of the effects of formulation, food, or a proton‐pump inhibitor on the pharmacokinetics of glasdegib (PF‐04449913) in healthy volunteers: a randomized phase I study. Cancer Chemother Pharmacol. 2019;83(3):463‐472.
    1. Krens SD, Lassche G, Jansman FGA, et al. Dose recommendations for anticancer drugs in patients with renal or hepatic impairment. Lancet Oncol. 2019;20(4):e200‐e207.
    1. Lin S, Shaik N, Martinelli G, et al. Population pharmacokinetics of glasdegib in patients with advanced hematologic malignancies and solid tumors. J Clin Pharmacol. 2019;60(5):605‐616.
    1. U.S. Department of Health and Human Services Food and Drug Administration . Guidance for industry: pharmacokinetics in patients with impaired hepatic function: study design, data analysis, and impact on dosing and labeling; May 2003. Silver Spring, MD: US Department of Health and Human Services Food and Drug Administration; 2003.
    1. European Medicines Agency Committee for Medicinal Products for Human Use . Guideline on the evaluation of the pharmacokinetics of medicinal products in patients with impaired hepatic function. February 2005. London: European Medicines Agency; 2005.
    1. Lewis GP, Jusko WJ. Pharmacokinetics of ampicillin in cirrhosis. Clin Pharmacol Ther. 1975;18(4):475‐484.
    1. Piafsky KM. Disease‐induced changes in the plasma binding of basic drugs. Clin Pharmacokinet. 1980;5(3):246‐262.
    1. Rowland M. Protein binding and drug clearance. Clin Pharmacokinet. 1984;9(suppl 1):10‐17.
    1. Shaik N, Hee B, Liang Y, LaBadie RR. Absolute oral bioavailability of glasdegib (PF‐04449913), a smoothened inhibitor, in randomized healthy volunteers. Clin Pharmacol Drug Dev. 2019;8(7):895‐902.
    1. Greenblatt DJ, Sellers EM, Koch‐Weser J. Importance of protein binding for the interpretation of serum or plasma drug concentrations. J Clin Pharmacol. 1982;22(5‐6):259‐263.
    1. Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical relevance. Clin Pharmacol Ther. 2002;71(3):115‐121.
    1. Patel H, Egorin MJ, Remick SC, et al. Comparison of Child‐Pugh (CP) criteria and NCI Organ Dysfunction Working Group (NCI‐ODWG) criteria for hepatic dysfunction (HD): implications for chemotherapy dosing. J Clin Oncol. 2004;22(14_suppl):6051.

Source: PubMed

3
Abonneren