A Population Pharmacokinetic and Pharmacodynamic Analysis of Abemaciclib in a Phase I Clinical Trial in Cancer Patients

Sonya C Tate, Amanda K Sykes, Palaniappan Kulanthaivel, Edward M Chan, P Kellie Turner, Damien M Cronier, Sonya C Tate, Amanda K Sykes, Palaniappan Kulanthaivel, Edward M Chan, P Kellie Turner, Damien M Cronier

Abstract

Background and objectives: Abemaciclib, a dual inhibitor of cyclin-dependent kinases 4 and 6, has demonstrated clinical activity in a number of different cancer types. The objectives of this study were to characterize the pharmacokinetics of abemaciclib in cancer patients using population pharmacokinetic (popPK) modeling, and to evaluate target engagement at clinically relevant dose levels.

Methods: A phase I study was conducted in cancer patients which incorporated intensive pharmacokinetic sampling after single and multiple oral doses of abemaciclib. Data were analyzed by popPK modeling, and patient demographics contributing to pharmacokinetic variability were explored. Target engagement was evaluated by combining the clinical popPK model with a previously developed pre-clinical pharmacokinetic/pharmacodynamic model.

Results: The pharmacokinetic analysis incorporated 4012 plasma concentrations from 224 patients treated with abemaciclib at doses ranging from 50 to 225 mg every 24 h and 75 to 275 mg every 12 h. A linear one-compartment model with time- and dose-dependent relative bioavailability (F rel) adequately described the pharmacokinetics of abemaciclib. Serum albumin and alkaline phosphatase were the only significant covariates identified in the model, the inclusion of which reduced inter-individual variability in F rel by 10.3 percentage points. By combining the clinical popPK model with the previously developed pre-clinical pharmacokinetic/pharmacodynamic model, the extent of target engagement in skin in cancer patients was successfully predicted.

Conclusion: The proportion of abemaciclib pharmacokinetic variability that can be attributed to patient demographics is negligible, and as such there are currently no dose adjustments recommended for adult patients of different sex, age, or body weight.

Trial registration: NCT01394016 (ClinicalTrials.gov).

Conflict of interest statement

Funding

This study was sponsored by Eli Lilly and Company.

Conflict of interest

SCT, AKS, PK, EMC, PKT, and DMC are employees of Eli Lilly and Company. AKS, PK, EMC, PKT, and DMC own stock in Eli Lilly and Company.

Ethical approval

The clinical trial was conducted in accordance with the Declaration of Helsinki and the International Conference on Harmonisation. The study protocol was approved by all institutional review boards.

Informed consent

Written informed consent was collected from all patients before conducting study procedures.

Figures

Fig. 1
Fig. 1
Visual predictive checks of the clinical abemaciclib population pharmacokinetic model after every 12 h dosing at 150 mg (a) or 200 mg (b). The circles denote observed abemaciclib plasma concentration data, and the solid and dotted lines represent the median and the 5th and 95th percentiles of 1000 individual model simulations, respectively
Fig. 2
Fig. 2
The covariate relationships retained in the final population pharmacokinetic model for serum albumin (a) and alkaline phosphatase (b) versus adjusted post hoc estimates of CL/F (i.e., CL/F × 1/Frel). The circles denote observed patient covariates and model-predicted parameter estimates, and the solid line denotes the estimated covariate–parameter relationship. CL/F apparent clearance, Frel relative bioavailability
Fig. 3
Fig. 3
Simulated change in p-Rb expression from baseline using the abemaciclib clinical population pharmacokinetic model in combination with the semi-mechanistic pre-clinical pharmacodynamic model. Curtailed axes are used in the main plot to aid interpretation of the dose-response curve; the complete plot with all observed data is provided in the inset pane. The boxplots represent the observed change in p-Rb expression in epidermal keratinocytes at day 15 (pre-dose) compared to baseline for the most populated daily doses (150 mg [combined as 75 mg q12h and 150 mg q24h], 300 mg [150 mg q12h], and 400 mg [200 mg q12h]), where the box is constructed using the median, 25th, and 75th percentiles, and the whiskers extend to the most extreme datapoints not considered outliers; boxplot outliers are represented by red crosses. For the less populated doses (50, 100, and 225 mg q24h, and 100 and 275 mg q12h), the observed change in p-Rb expression at day 15 (pre-dose) for individual patients is represented by red circles. The solid and dotted lines represent the median, and the 5th and 95th percentiles of 1000 individual model simulations, respectively. CI confidence interval, p-Rb phosphorylated retinoblastoma protein, q12h every 12 h, q24h every 24 h

References

    1. Harbour JW, Luo RX, Dei Santi A, Postigo AA, Dean DC. Cdk phosphorylation triggers sequential intramolecular interactions that progressively block Rb functions as cells move through G1. Cell. 1999;98(6):859–869. doi: 10.1016/S0092-8674(00)81519-6.
    1. Knudsen KE, Weber E, Arden KC, Cavenee WK, Feramisco JR, Knudsen ES. The retinoblastoma tumor suppressor inhibits cellular proliferation through two distinct mechanisms: inhibition of cell cycle progression and induction of cell death. Oncogene. 1999;18(37):5239–5245. doi: 10.1038/sj.onc.1202910.
    1. Paggi MG, Baldi A, Bonetto F, Giordano A. Retinoblastoma protein family in cell cycle and cancer: a review. J Cell Biochem. 1996;62(3):418–430. doi: 10.1002/(SICI)1097-4644(199609)62:3<418::AID-JCB12>;2-E.
    1. Nevins JR. The Rb/E2F pathway and cancer. Hum Mol Genet. 2001;10(7):699–703. doi: 10.1093/hmg/10.7.699.
    1. Gelbert LM, Cai S, Lin X, Sanchez-Martinez C, Del Prado M, Lallena MJ, et al. Preclinical characterization of the CDK4/6 inhibitor LY2835219: in-vivo cell cycle-dependent/independent anti-tumor activities alone/in combination with gemcitabine. Investig New Drugs. 2014;32(5):825–837. doi: 10.1007/s10637-014-0120-7.
    1. Yadav V, Burke TF, Huber L, Van Horn RD, Zhang Y, Buchanan SG, et al. The CDK4/6 inhibitor LY2835219 overcomes vemurafenib resistance resulting from MAPK reactivation and cyclin D1 upregulation. Mol Cancer Ther. 2014;13(10):2253–2263. doi: 10.1158/1535-7163.MCT-14-0257.
    1. Raub TJ, Wishart GN, Kulanthaivel P, Staton BA, Ajamie RT, Sawada GA, et al. Brain exposure of two selective dual CDK4 and CDK6 inhibitors and the antitumor activity of CDK4 and CDK6 inhibition in combination with temozolomide in an intracranial glioblastoma xenograft. Drug Metab Dispos. 2015;43(9):1360–1371. doi: 10.1124/dmd.114.062745.
    1. Patnaik A, Rosen LS, Tolaney SM, Tolcher AW, Goldman JW, Gandhi L, et al. Efficacy and safety of abemaciclib, an inhibitor of CDK4 and CDK6, for patients with breast cancer, non-small cell lung cancer, and other solid tumors. Cancer Discov. 2016;6(7):740–753. doi: 10.1158/-16-0095.
    1. Tate SC, Cai S, Ajamie RT, Burke T, Beckmann RP, Chan EM, et al. Semi-mechanistic pharmacokinetic/pharmacodynamic modeling of the antitumor activity of LY2835219, a new cyclin-dependent kinase 4/6 inhibitor, in mice bearing human tumor xenografts. Clin Cancer Res. 2014;20(14):3763–3774. doi: 10.1158/1078-0432.CCR-13-2846.
    1. Duffull SB, Wright DF, Winter HR. Interpreting population pharmacokinetic–pharmacodynamic analyses—a clinical viewpoint. Br J Clin Pharmacol. 2011;71(6):807–814. doi: 10.1111/j.1365-2125.2010.03891.x.
    1. Mould DR, Upton RN. Basic concepts in population modeling, simulation, and model-based drug development-part 2: introduction to pharmacokinetic modeling methods. CPT Pharmacometr Syst Pharmacol. 2013;2:e38. doi: 10.1038/psp.2013.14.
    1. Kulanthaivel P, Mahadevan D, Turner PK, Royalty J, Ng WT, Yi P, et al. Pharmacokinetic drug interactions between abemaciclib and CYP3A inducers and inhibitors [abstract no. CT153]. In: AACR annual meeting, 16–20 April 2016, New Orleans.
    1. Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41. doi: 10.1159/000180580.
    1. Burke T, Torres R, McNulty A, Dempsey J, Kolis S, Kulanthaivel P, et al. The major human metabolites of abemaciclib are inhibitors of CDK4 and CDK6 [abstract no. 2830]. In: AACR annual meeting, 16–20 April 2016, New Orleans.
    1. Beal SL. Ways to fit a PK model with some data below the quantification limit. J Pharmacokinet Pharmacodyn. 2001;28(5):481–504. doi: 10.1023/A:1012299115260.
    1. Lavielle M, Aarons L. What do we mean by identifiability in mixed effects models? J Pharmacokinet Pharmacodyn. 2016;43(1):111–122. doi: 10.1007/s10928-015-9459-4.
    1. Turner PK, Chappell J, Kulanthaivel P, Ng WT, Royalty J. Food effect on the pharmacokinetics of 200-mg abemaciclib in healthy subject [abstract no. CT152]. In: AACR annual meeting, 16–20 April 2016, New Orleans.
    1. Mager DE, Woo S, Jusko WJ. Scaling pharmacodynamics from in vitro and preclinical animal studies to humans. Drug Metab Pharmacokinet. 2009;24(1):16–24. doi: 10.2133/dmpk.24.16.

Source: PubMed

3
Abonner