Phase 1 study to evaluate the effects of rifampin on pharmacokinetics of pevonedistat, a NEDD8-activating enzyme inhibitor in patients with advanced solid tumors

Xiaofei Zhou, Ulka Vaishampayan, Devalingam Mahalingam, R Donald Harvey, Ki Young Chung, Farhad Sedarati, Cassie Dong, Douglas V Faller, Karthik Venkatakrishnan, Neeraj Gupta, Xiaofei Zhou, Ulka Vaishampayan, Devalingam Mahalingam, R Donald Harvey, Ki Young Chung, Farhad Sedarati, Cassie Dong, Douglas V Faller, Karthik Venkatakrishnan, Neeraj Gupta

Abstract

Pevonedistat (TAK-924/MLN4924) is an investigational small molecule inhibitor of the NEDD8-activating enzyme that has demonstrated clinical activity across solid tumors and hematological malignancies. Here we report the results of a phase 1 study evaluating the effect of rifampin, a strong CYP3A inducer, on the pharmacokinetics (PK) of pevonedistat in patients with advanced solid tumors (NCT03486314). Patients received a single 50 mg/m2 pevonedistat dose via a 1-h infusion on Days 1 (in the absence of rifampin) and 10 (in the presence of rifampin), and daily oral dosing of rifampin 600 mg on Days 3-11. Twenty patients were enrolled and were evaluable for PK and safety. Following a single dose of pevonedistat at 50 mg/m2, the mean terminal half-life of pevonedistat was 5.7 and 7.4 h in the presence and in the absence of rifampin, respectively. The geometric mean AUC0-inf of pevonedistat in the presence of rifampin was 79% of that without rifampin (90% CI: 69.2%-90.2%). The geometric mean Cmax of pevonedistat in the presence of rifampin was similar to that in the absence of rifampin (96.2%; 90% CI: 79.2%-117%). Coadministration of pevonedistat with rifampin, a strong metabolic enzyme inducer, did not result in clinically meaningful decreases in systemic exposures of pevonedistat. The study results support the recommendation that no pevonedistat dose adjustment is needed for patients receiving concomitant CYP3A inducers. CLINICALTRIALS.GOV IDENTIFIER: NCT03486314.

Keywords: Advanced malignancies; Pevonedistat; Pharmacokinetics [4 to 6 max]; Rifampin.

Conflict of interest statement

XZ is employed by Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA. UV has received honoraria from Sanofi, Exelixis, Pfizer, Bayer; consulting fees from BMS, Merck, Exelixis, Bayer, Aveo; and grants/funds from Merck. DM has received honoraria from Amgen, BMS, Exelixis, Eisai; consulting fees from Qurient, OncoOne; and grants/funds from Amgen, Merck, Oncolytics. RDH has received research funding to their institution that supports their salary from: Abbisko, AbbVie, Actuate, Amgen, AstraZeneca, Bayer, Bristol-Myers Squibb, Boston Biomedical, Genmab, GlaxoSmithKline, Infinity, InhibRx, Janssen, Merck, Mersana, Meryx, Nektar, Novartis, Pfizer, Regeneron, Sanofi, Sutro, Takeda, Turning Point Therapeutics, Xencor; and reports consultancy for: Amgen, GlaxoSmithKline. KYC has nothing to disclose. FS is employed by Takeda. CD is employed by Takeda; and has ownership of stock/shares in Takeda and Settle Genetics. DVF is employed by Takeda; has ownership of stock/shares in Viracta Therapeutics, Briacell Therapeutics, Phoenicia Biosciences, R2ccouncil, LLC; was on the advisory council/committee for Viracta Therapeutics, Briacell Therapeutics; and owns multiple patents unrelated to this work. KV is a previous employee of Takeda; and a current employee of EMD Serono Research & Development Institute, Inc., Billerica, MA. NG is employed by Takeda; and has ownership of stock/shares in Takeda.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Mean (+ SD) pevonedistat plasma concentration–time profiles in the absence (Day 1) or presence (Day 10) of rifampin (PK population) linear scale (Panel A), semi-log scale (Panel B)
Fig. 2
Fig. 2
Individual comparisons of AUC∞ and Cmax for pevonedistat in the absence (Day 1) or presence (Day 10) of rifampin (PK population)

References

    1. Wolenski FS, Fisher CD, Sano T, et al. The NAE inhibitor pevonedistat (MLN4924) synergizes with TNF-α to activate apoptosis. Cell death discovery. 2015;1:15034. doi: 10.1038/cddiscovery.2015.34.
    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. doi: 10.1182/blood-2017-09-805895.
    1. Zhou L, Zhang W, Sun Y, Jia L. Protein neddylation and its alterations in human cancers for targeted therapy. Cell Signal. 2018;44:92–102. doi: 10.1016/j.cellsig.2018.01.009.
    1. Zhou L, Jiang Y, Luo Q, Li L, Jia L. Neddylation: a novel modulator of the tumor microenvironment. Mol Cancer. 2019;18(1):77. doi: 10.1186/s12943-019-0979-1.
    1. Abidi N, Xirodimas DP. Regulation of cancer-related pathways by protein NEDDylation and strategies for the use of NEDD8 inhibitors in the clinic. Endocr Relat Cancer. 2015;22(1):T55–70. doi: 10.1530/erc-14-0315.
    1. Jiang Y, Jia L. Neddylation pathway as a novel anti-cancer target: mechanistic investigation and therapeutic implication. Anticancer Agents Med Chem. 2015;15(9):1127–1133. doi: 10.2174/1871520615666150305111257.
    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. doi: 10.1158/1078-0432.CCR-09-0343.
    1. Milhollen MA, Traore T, Adams-Duffy J, et al. MLN4924, a NEDD8-activating enzyme inhibitor, is active in diffuse large B-cell lymphoma models: rationale for treatment of NF-(kappa)B-dependent lymphoma. Blood. 2010;116(9):1515–1523. doi: 10.1182/blood-2010-03-272567.
    1. Swords RT, Kelly KR, Smith PG, et al. Inhibition of NEDD8-activating enzyme: a novel approach for the treatment of acute myeloid leukemia. Blood. 2010;115(18):3796–3800. doi: 10.1182/blood-2009-11-254862.
    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. doi: 10.1007/s10637-016-0348-5.
    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. doi: 10.1007/s10637-018-0610-0.
    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. doi: 10.1158/1078-0432.CCR-15-1338.
    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. doi: 10.1158/1078-0432.CCR-15-1237.
    1. Zhou X, Sedarati F, Faller DV, et al. Phase I study assessing the mass balance, pharmacokinetics, and excretion of [(14)C]-pevonedistat, a NEDD8-activating enzyme inhibitor in patients with advanced solid tumors. Invest New Drugs. 2021;39(2):488–498. doi: 10.1007/s10637-020-01017-x.
    1. Faessel HM, Mould DR, Zhou X, et al. Population pharmacokinetics of pevonedistat alone or in combination with standard of care in patients with solid tumours or haematological malignancies. Br J Clin Pharmacol. 2019;85(11):2568–2579. doi: 10.1111/bcp.14078.
    1. Faessel H, Nemunaitis J, Bauer TM, et al. Effect of CYP3A inhibitors on the pharmacokinetics of pevonedistat in patients with advanced solid tumours. Br J Clin Pharmacol. 2019;85(7):1464–1473. doi: 10.1111/bcp.13915.
    1. Cotreau MM, Siebers NM, Miller J, Strahs AL, Slichenmyer W. Effects of ketoconazole or rifampin on the pharmacokinetics of tivozanib hydrochloride, a vascular endothelial growth factor receptor tyrosine kinase inhibitor. Clin Pharmacol Drug Dev. 2015;4(2):137–142. doi: 10.1002/cpdd.145.
    1. Gupta N, Hanley MJ, Venkatakrishnan K, et al. Effects of strong CYP3A inhibition and induction on the pharmacokinetics of ixazomib, an oral proteasome inhibitor: results of drug-drug interaction studies in patients with advanced solid tumors or lymphoma and a physiologically based pharmacokinetic analysis. J Clin Pharmacol. 2018;58(2):180–192. doi: 10.1002/jcph.988.
    1. Faucette S, Wagh S, Trivedi A, Venkatakrishnan K, Gupta N. Reverse translation of US Food and Drug Administration reviews of oncology new molecular entities approved in 2011–2017: lessons learned for anticancer drug development. Clin Transl Sci. 2018;11(2):123–146. doi: 10.1111/cts.12527.
    1. Bullock JM, Lin T, Bilic S. Clinical pharmacology tools and evaluations to facilitate comprehensive dose finding in oncology: a continuous risk-benefit approach. J Clin Pharmacol. 2017;57(Suppl 10):S105–s115. doi: 10.1002/jcph.908.
    1. Varma MV, Lai Y, Feng B, et al. Physiologically based modeling of pravastatin transporter-mediated hepatobiliary disposition and drug-drug interactions. Pharm Res. 2012;29(10):2860–2873. doi: 10.1007/s11095-012-0792-7.
    1. ACCP Abstract Booklet (2021) Clinical Pharmacology in Drug Development. 10(S1):1–104. 10.1002/cpdd.1004

Source: PubMed

3
Abonnieren