An Adaptive Physiologically Based Pharmacokinetic-Driven Design to Investigate the Effect of Itraconazole and Rifampicin on the Pharmacokinetics of Molibresib (GSK525762) in Healthy Female Volunteers

Kylie Riddell, Aarti Patel, Gary Collins, Yanyan Zhou, Dan Schramek, Brandon E Kremer, Geraldine Ferron-Brady, Kylie Riddell, Aarti Patel, Gary Collins, Yanyan Zhou, Dan Schramek, Brandon E Kremer, Geraldine Ferron-Brady

Abstract

Molibresib (GSK525762), an orally bioavailable small molecule with 2 major equipotent active metabolites, is being developed for the treatment of cancers. Molibresib is a substrate of cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp). To enable administering safe doses of molibresib to healthy volunteers, this 2-part randomized, open-label, crossover drug-drug interaction trial was conducted as an adaptive design study using physiologically based pharmacokinetic (PBPK) modeling and simulation to predict the lowest doses of molibresib that could be safely administered alone (10 mg) or with itraconazole and rifampicin (strong inhibitors and inducers of CYP3A and P-gp, respectively). PBPK simulation guided the molibresib dose (5 mg) to be administered along with itraconazole in part 1. Itraconazole increased total exposure (AUC) of molibresib by 4.15-fold with a 66% increase in Cmax , whereas the total AUC and Cmax for the 2 major active metabolites of molibresib decreased by about 70% and 87%, respectively. A second PBPK simulation was conducted with part 1 data to also include the active metabolites to update the recommendation for the molibresib dose (20 mg) with rifampicin. With rifampicin, the AUC and Cmax of molibresib decreased by approximately 91% and 80%, respectively, whereas the AUC of the 2 active metabolites decreased to a lesser extent (8%), with a 2-fold increase in Cmax . The results of this study confirmed the in vitro data that molibresib is a substrate for CYP3A4. The adaptive design, including Simcyp simulations, allowed evaluation of 2 drug interactions of an oncology drug in a single trial, thus minimizing time and exposures administered to healthy subjects.

Keywords: GSK525762; Simcyp; drug-drug interaction; itraconazole; molibresib; pharmacokinetics; rifampicin.

Conflict of interest statement

All authors are employees of GSK, hold company stock, and meet the criteria for authorship set forth by the International Committee for Medical Journal Editors. Editorial support (development of the first draft, assembling tables and figures, collating author comments, and referencing) was provided by Guissou Dabiri, PhD, and was funded by GSK.

© 2020 Glaxo Group Limited. The Journal of Clinical Pharmacology published by Wiley Periodicals LLC on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
PBPK simulation workflow.
Figure 2
Figure 2
Adaptive study design.
Figure 3
Figure 3
Observed and Simcyp‐simulated molibresib concentration‐time profile after a single 60‐ or 100‐mg dose (BET115521).
Figure 4
Figure 4
Dose‐normalized mean molibresib, active metabolite, and total active moiety concentration‐versus‐time plots when molibresib is administered alone (open symbols) or in combination (closed symbols).
Figure 5
Figure 5
Observed and Simcyp‐simulated molibresib (A) and GSK3529246 (B) concentration‐time profiles after a single 10‐mg dose.

References

    1. Nicodeme E, Jeffrey KL, Schaefer U, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468(7327):1119‐1123.
    1. French CA, Ramirez CL, Kolmakova J, et al. BRD‐NUT oncoproteins: a family of closely related nuclear proteins that block epithelial differentiation and maintain the growth of carcinoma cells. Oncogene. 2008;27(15):2237‐2242.
    1. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeutic strategy to target c‐Myc. Cell. 2011;146(6):904‐197.
    1. Mertz JA, Conery AR, Bryant BM, et al. Targeting MYC dependence in cancer by inhibiting BET bromodomains. Proc Natl Acad Sci U S A. 2011;108(40):16669‐16674.
    1. Chung CW, Coste H, White JH, et al. Discovery and characterization of small molecule inhibitors of the BET family bromodomains. J Med Chem. 2011;54(11):3827‐3838.
    1. Piha‐Paul S, Hann C, French C et al. Phase I study of molibresib (GSK525762), a bromodomain and extra terminal protein (BET) inhibitor, in patients with advanced solid tumors including NUT carcinoma (NC). JNCI Cancer Spectrum. 2019;4(2):pkz093.
    1. Ke AB, Zamek‐Gliszczynski MJ, Higgins JW, Hall SD. Itraconazole and clarithromycin as ketoconazole alternatives for clinical CYP3A inhibition studies. Clin Pharm Ther. 2014;95(5):473‐C>.
    1. Chen J, Raymond K. Roles of rifampicin in drug‐drug interactions: underlying molecular mechanisms involving the nuclear pregnane X receptor. Ann Clin Microbiol Antimimicrob. 2006;5:3.
    1. Stoch SA, Friedman E, Maes A, et al. Effect of different durations of ketoconazole dosing on the single‐dose pharmacokinetics of midazolam: shortening the paradigm. J Clin Pharmacol. 2009;49(4):398‐406.
    1. Backmann JT, Olkkola KT, Neuvonen PJ. Rifampin drastically reduces plasma concentrations and effects of oral midazolam. Clin Pharmacol Ther. 1996;59(1):7‐13.
    1. Olkkola KT, Backman JT, Neuvonen PJ. Midazolam should be avoided in patients receiving the systemic antimycotics ketoconazole or itraconazole. Clin Pharmacol Ther. 1994;55(5):481‐485.
    1. Almond LM, Mukadam S, Gardner I, et al. Prediction of drug‐drug interactions arising from CYP3A induction using a physiologically based dynamic model [published correction appears in Drug Metab Dispos. 2016;44(6):877]. Drug Metab Dispos. 2016;44(6):821‐832.
    1. Greenblatt HK, Greenblatt DJ. Liver injury associated with ketoconazole: review of the published evidence. J Clin Pharmacol. 2014;54(12):1321‐1329.
    1. Liu L, Bello A, Dresser MJ, et al. Best practices for the use of itraconazole as a replacement for ketoconazole in drug‐drug interaction studies. J Clin Pharm. 2016;56(2):143‐151.

Source: PubMed

3
Abonnieren