Phase I study assessing the mass balance, pharmacokinetics, and excretion of [14C]-pevonedistat, a NEDD8-activating enzyme inhibitor in patients with advanced solid tumors

Xiaofei Zhou, Farhad Sedarati, Douglas V Faller, Dan Zhao, Hélène M Faessel, Swapan Chowdhury, Jayaprakasam Bolleddula, Yuexian Li, Karthik Venkatakrishnan, Zsuzsanna Papai, Xiaofei Zhou, Farhad Sedarati, Douglas V Faller, Dan Zhao, Hélène M Faessel, Swapan Chowdhury, Jayaprakasam Bolleddula, Yuexian Li, Karthik Venkatakrishnan, Zsuzsanna Papai

Abstract

Pevonedistat (TAK-924/MLN4924) is an investigational small-molecule inhibitor of the NEDD8-activating enzyme that has demonstrated preclinical and clinical activity across solid tumors and hematological malignancies. Here we report the results of a phase I trial characterizing the mass balance, pharmacokinetics, and clearance pathways of [14C]-pevonedistat in patients with advanced solid tumors (NCT03057366). In part A (n = 8), patients received a single 1-h intravenous infusion of [14C]-pevonedistat 25 mg/m2. In part B (n = 7), patients received pevonedistat 25 or 20 mg/m2 on days 1, 3, and 5 in combination with, respectively, docetaxel 75 mg/m2 or carboplatin AUC5 plus paclitaxel 175 mg/m2 on day 1 every 3 weeks. Following the single dose of [14C]-pevonedistat 25 mg/m2 in part A, there was a parallel log-linear decline in plasma and whole blood pevonedistat concentration, with systemic exposure of unchanged pevonedistat representing 41% of drug-related material (i.e., unchanged pevonedistat and its metabolites). The mean terminal half-life of pevonedistat and drug-related material in plasma was 8.4 and 15.6 h, respectively. Pevonedistat distributed preferentially in whole blood with a mean whole-blood-to-plasma ratio for pevonedistat AUC∞ of 40.8. By 1 week post dose, the mean recovery of administered radioactivity was 94% (41% in urine and 53% in feces). The pevonedistat safety profile during both study parts was consistent with previous clinical experience, with no new safety signals observed. In part B, pevonedistat in combination with docetaxel or carboplatin plus paclitaxel was generally well tolerated. ClinicalTrials.gov identifier: NCT03057366 .

Keywords: advanced solid tumors; elimination; mass balance; pevonedistat; pharmacokinetics; phase I.

Conflict of interest statement

Xiaofei Zhou declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Farhad Sedarati declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Douglas V. Faller declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Swapan Chowdhury declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Yuexian Li declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Hélène M. Faessel declares employment with, and ownership of stocks/shares in Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited. Jayaprakasam Bolleddula declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited at the time of the study. Karthik Venkatakrishnan declares employment with Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited at the time of the study. Zsuzsanna Papai declares no conflict of interest.

Figures

Fig. 1
Fig. 1
Mean concentration–time profiles of pevonedistat and total radioactivity (drug-related material) in plasma following a single infusion of [14C]-pevonedistat 25 mg/m2
Fig. 2
Fig. 2
Mean concentration–time profiles of pevonedistat in plasma and whole blood following a single infusion of [14C]-pevonedistat 25 mg/m2
Fig. 3
Fig. 3
Mean time course of cumulative excretion of total radioactivity (drug-related material) in urine and feces following a single infusion of [14C]-pevonedistat 25 mg/m2

References

    1. Wolenski FS, Fisher CD, Sano T, et al. The NAE inhibitor pevonedistat (MLN4924) synergizes with TNF-alpha to activate apoptosis. Cell Death Discov. 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–T70. doi: 10.1530/ERC-14-0315.
    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. Jiang Y, Jia L (2015) Neddylation pathway as a novel anti-cancer target: mechanistic investigation and therapeutic implication. Anticancer Agents Med Chem 15(9):1127–1133
    1. Milhollen MA, Traore T, Adams-Duffy J et al (2010) MLN4924, a NEDD8-activating enzyme inhibitor, is active in diffuse large B-cell lymphoma models: rationale for treatment of NF-κB-dependent lymphoma. Blood 116(9):1515–1523. 10.1182/blood-2010-03-272567
    1. Nawrocki ST, Griffin P, Kelly KR, Carew JS. MLN4924: a novel first-in-class inhibitor of NEDD8-activating enzyme for cancer therapy. Expert Opin Investig Drugs. 2012;21(10):1563–1573. doi: 10.1517/13543784.2012.707192.
    1. Swords RT, Kelly KR, Smith PG et al (2010) Inhibition of NEDD8-activating enzyme: a novel approach for the treatment of acute myeloid leukemia. Blood 115(18):3796–3800.
    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. 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. doi: 10.1111/bjh.13323.
    1. Swords RT, Watts J, Erba HP, et al. Expanded safety analysis of pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, in patients with acute myeloid leukemia and myelodysplastic syndromes. Blood Cancer J. 2017;7(2):e520. doi: 10.1038/bcj.2017.1.
    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. Bullock JM, Lin T, Bilic S (2017) Clinical pharmacology tools and evaluations to facilitate comprehensive dose finding in oncology: a continuous risk-benefit approach. J Clin Pharmacol 57(Suppl 10):S105–S115. 10.1002/jcph.908
    1. Faucette S, Wagh S, Trivedi A, Venkatakrishnan K, Gupta N (2018) 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 11(2):123–146. 10.1111/cts.12527
    1. Roffey SJ, Obach RS, Gedge JI, Smith DA. What is the objective of the mass balance study? A retrospective analysis of data in animal and human excretion studies employing radiolabeled drugs. Drug Metab Rev. 2007;39(1):17–43. doi: 10.1080/03602530600952172.
    1. Akbar SA, Brown PR. Human erythrocyte CAI and CAII isoenzymes in hypoxemic and anemic fetuses. Clin Biochem. 1996;29(1):57–62. doi: 10.1016/0009-9120(95)02020-9.
    1. Ali Akbar S, Brown PR (1996) Measurement of human erythrocyte CAI and CAII in adult, newborn, and fetal blood. Clin Biochem 29(2):157–164. 10.1016/0009-9120(95)02021-7
    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.

Source: PubMed

3
Se inscrever