Population pharmacokinetics of pevonedistat alone or in combination with standard of care in patients with solid tumours or haematological malignancies

Hélène M Faessel, Diane R Mould, Xiaofei Zhou, Douglas V Faller, Farhad Sedarati, Karthik Venkatakrishnan, Hélène M Faessel, Diane R Mould, Xiaofei Zhou, Douglas V Faller, Farhad Sedarati, Karthik Venkatakrishnan

Abstract

Aims: A population pharmacokinetic (PK) analysis was conducted to quantify the impact of patient-specific and concurrent medication factors on pevonedistat PK.

Methods: Data were pooled from 6 clinical studies consisting of 335 patients with solid tumours or haematological malignancies administered pevonedistat alone or in combination with azacitidine, docetaxel, carboplatin + paclitaxel, or gemcitabine. Model development and covariate analysis followed standard methods. Parameters and bootstrap 95% confidence intervals were estimated using nonlinear mixed-effects modelling. The final model was evaluated using visual predictive checks and other goodness-of-fit criteria.

Results: A linear 2-compartment model best described pevonedistat PK. The final model included the effect of body surface area (BSA) on clearance (CL and Q) and volume of distribution of pevonedistat, effect of concomitantly administered carboplatin + paclitaxel on CL, and effect of albumin on Q. Race, sex, age, tumour type (haematological vs solid), mild or moderate renal impairment (creatinine clearance ≥30 mL/min), or mild hepatic impairment, had no impact on pevonedistat PK.

Conclusions: The clinical PK profile of pevonedistat is comparable in patients with solid tumours or haematological malignancies. All PK parameters exhibited ≥20% change over the observed BSA range (1.38-3 m2 ) with CL ranging from 75.5 to 208% of the reference value, with simulations supporting BSA-based dosing to minimize interindividual variability in drug exposures. Concurrent administration of carboplatin + paclitaxel decreased pevonedistat CL by approximately 44%, while coadministration with azacitidine, gemcitabine or docetaxel did not alter pevonedistat CL. No other factors were identified as influencing pevonedistat PK.

Trial registration: ClinicalTrials.gov NCT00677170 NCT00722488 NCT00911066 NCT01011530 NCT01814826 NCT01862328.

Keywords: anticancer drugs; pharmacokinetics; population analysis.

Conflict of interest statement

H.M.F., X.Z., D.V.F., F.S. and K.V. are employees of Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. D.R.M is a consultant for Millennium Pharmaceuticals, Inc., Cambridge, MA, USA, a wholly owned subsidiary of Takeda Pharmaceutical Company Limited.

© 2019 The Authors. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.

Figures

Figure 1
Figure 1
Visual predictive checks results for final model—observed and simulated pevonedistat dose‐normalized concentrations vs time after dose—all data. Blue open circles are the observed data; solid red line is the median of the observed data; red dashed lines are the 5th and 95th percentiles of the observed data; black solid line is the median of the simulated data; black dashed lines are the 5th and 95th percentiles of the simulated data; grey shaded areas are the 95% confidence intervals associated with the simulated lower and upper percentiles. The confidence intervals (CIs) are not computed beyond 60 hours as the number of observations is insufficient to determine confidence intervals
Figure 2
Figure 2
Visual predictive checks of dose‐normalized concentration–time data of pevonedistat coadministered with azacitidine (A) or carboplatin plus paclitaxel (B). Blue open circles are the observed data; solid red line is the median of the observed data; red dashed lines are the 5th and 95th percentiles of the observed data; black solid line is the median of the simulated data; black dashed lines are the 5th and 95th percentiles of the simulated data; grey shaded areas are the 95% confidence intervals associated with the simulated lower and upper percentiles
Figure 3
Figure 3
Pevonedistat clearance by patient covariates. Horizontal lines comprising the box are the 25th, 50th (median) and 75th percentiles. The whisker ends denote 1.5 times the difference between the 25th and 75th percentiles and the symbols beyond the whiskers are the outliers
Figure 4
Figure 4
Pevonedistat concentration vs time with and without concomitant administration of carboplatin + paclitaxel
Figure 5
Figure 5
Simulated pevonedistat exposures following fixed (mg) vs BSA based dosing (mg/m2). AUC, area under the concentration–time curve; BSA, body surface area
Figure 6
Figure 6
Simulated pevonedistat concentration–time profiles at the recommended clinical dose of 20 mg/m2 administered on days 1, 3 and 5. Note: Black solid line is the median simulated concentration; black dashed lines are the lower 5th and upper 95th percentiles of the simulated data; grey shaded areas are the 95% confidence intervals for each percentile

References

    1. Reinstein E, Ciechanover A. Narrative review: protein degradation and human diseases: the ubiquitin connection. Ann Intern Med. 2006;145(9):676‐684.
    1. Soucy TA, Smith PG, Rolfe M. Targeting NEDD8‐activated cullin‐RING ligases for the treatment of cancer. Clin Cancer Res. 2009;15(12):3912‐3916.
    1. Soucy TA, Dick LR, Smith PG, Milhollen MA, Brownell JE. The NEDD8 conjugation pathway and its relevance in cancer biology and therapy. Genes Cancer. 2010;1(7):708‐716.
    1. Soucy TA, Smith PG, Milhollen MA, et al. An inhibitor of NEDD8‐activating enzyme as a new approach to treat cancer. Nature. 2009;458(7239):732‐736.
    1. Brownell JE, Sintchak MD, Gavin JM, et al. Substrate‐assisted inhibition of ubiquitin‐like protein‐activating enzymes: the NEDD8 E1 inhibitor MLN4924 forms a NEDD8‐AMP mimetic in situ. Mol Cell. 2010;37(1):102‐111.
    1. Swords RT, Erba HP, Deangelo DJ, et al. Pevonedistat (MLN4924), a first‐in‐class NEDD8‐activating enzyme inhibitor, in patients with acute myeloid leukaemia and myelodysplastic syndromes: a phase 1 study. Br J Haematol. 2015;169(4):534‐543.
    1. Sarantopoulos J, Shapiro GI, Cohen RB, et al. Phase I study of the investigational NEDD8‐activating enzyme inhibitor pevonedistat (TAK‐924/MLN4924) in patients with advanced solid tumors. Clin Cancer Res. 2016;22(4):847‐857.
    1. Shah JJ, Jakubowiak AJ, O'connor OA, et al. Phase I study of the novel investigational NEDD8‐activating enzyme inhibitor pevonedistat (MLN4924) in patients with relapsed/refractory multiple myeloma or lymphoma. Clin Cancer Res. 2016;22(1):34‐43.
    1. Bhatia S, Pavlick AC, Boasberg P, et al. A phase I study of the investigational NEDD8‐activating enzyme inhibitor pevonedistat (TAK‐924/MLN4924) in patients with metastatic melanoma. Invest New Drugs. 2016;34(4):439‐449.
    1. Lockhart AC, Bauer TM, Aggarwal C, et al. Phase Ib study of pevonedistat, a NEDD8‐activating enzyme inhibitor, in combination with docetaxel, carboplatin and paclitaxel, or gemcitabine, in patients with advanced solid tumors. Invest New Drugs. 2019;37(1):87‐97. 10.1007/s10637-018-0610-0
    1. Swords RT, Coutre S, Maris MB, et al. Pevonedistat, a first‐in‐class NEDD8‐activating enzyme inhibitor, combined with azacitidine in patients with AML. Blood. 2018;131(13):1415‐1424.
    1. Wu S‐J, Cheong J‐W, Onishi Y, Iida H, Kobayashi Y, Kim H‐J, et al. Phase 1/1b study of pevonedistat (PEV) as a single agent or combined with azacitidine (AZA) in east asian patients (pts) with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS) EHA Learning Center 2018; Abstract PS997.
    1. Bhatia S, Hamid O, Pavlick AC, et al. MLN4924, an investigational NEDD8‐activating enzyme (NAE) inhibitor, in patients (pts) with metastatic melanoma: results of a phase I study. J Clin Oncol. 2011;29(Suppl):Abstract 8529.
    1. Swords RT, Savona MR, Maris MB, et al. Pevonedistat (MLN4924), an investigational, first‐in‐class NAE inhibitor, in combination with azacitidine in elderly patients with acute myeloid leukemia (AML) considered unfit for conventional chemotherapy: updated results from the phase 1 C15009 trial. Blood. 2014;124(21):Abstract 2313.
    1. Boeckmann AJ, Bea LSL, Sheiner LB. In: NONMEM Users Guide ‐ Parts I to VIII , ed. NONMEM Installation Guide, NONMEM Project Group. San Francisco: University of California; March 1998.
    1. Ortmann O, Weiss JM, Diedrich K. Gonadotrophin‐releasing hormone (GnRH) and GnRH agonists: mechanisms of action. Reprod Biomed Online. 2002;5(Suppl 1):1‐7.
    1. Post TM, Freijer JI, Ploeger BA, Danhof M. Extensions to the visual predictive check to facilitate model performance evaluation. J Pharmacokinet Pharmacodyn. 2008;35(2):185‐202.
    1. Yafune A, Ishiguro M. Bootstrap approach for constructing confidence intervals for population pharmacokinetic parameters. I: a use of bootstrap standard error. Stat Med. 1999;18(5):581‐599.
    1. Harding SD, Sharman JL, Faccenda E, et al. The IUPHAR/BPS guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acids Res. 2018;46:D1091‐D1106.
    1. Alexander SPH, Fabbro D, Kelly E, et al. The Concise Guide to PHARMACOLOGY 2017/18: enzymes. Br J Pharmacol. 2017;174:S272‐S359.
    1. Faessel HM, Mould DR, Dezube BJ, Santillana S, Venkatakrishnan K. Population pharmacokinetics (PK) of the investigational NEDD8‐activating enzyme (NAE) inhibitor pevonedistat (TAK‐924/MLN4924) administered alone or in combination with azacitidine in patients (pts) with solid or hematologic malignancies. Mol Cancer Ther. 2015;14(12 Suppl 2):Abstract B148.
    1. Venkatakrishnan K, Friberg LE, Ouellet D, et al. Optimizing oncology therapeutics through quantitative translational and clinical pharmacology: challenges and opportunities. Clin Pharmacol Ther. 2015;97(1):37‐54.
    1. Zhang Y, Zhang L, Abraham S, et al. Assessment of the impact of renal impairment on systemic exposure of new molecular entities: evaluation of recent new drug applications. Clin Pharmacol Ther. 2009;85(3):305‐311.

Source: PubMed

3
Abonnieren