Single-Dose Pharmacokinetics of Milvexian in Participants with Mild or Moderate Hepatic Impairment Compared with Healthy Participants

Vidya Perera, Grigor Abelian, Danshi Li, Zhaoqing Wang, Liping Zhang, Susan Lubin, Wei Chen, Akintunde Bello, Bindu Murthy, Vidya Perera, Grigor Abelian, Danshi Li, Zhaoqing Wang, Liping Zhang, Susan Lubin, Wei Chen, Akintunde Bello, Bindu Murthy

Abstract

Background: Patients with hepatic impairment receiving antithrombotic agents metabolized primarily through the liver can be at risk for bleeding. Milvexian (BMS-986177/JNJ-70033093) is a small-molecule, active-site inhibitor of activated Factor XI (FXIa). Modulation of FXI may provide systemic anticoagulation without increased risk of clinically significant bleeding.

Objective: This open-label study evaluated the effects of mild or moderate hepatic impairment on the pharmacokinetics of milvexian to assess their impact on safety and dosing.

Methods: Single doses of milvexian 60 mg were administered to participants with mild hepatic impairment (n = 9), moderate hepatic impairment (n = 8), and normal hepatic function (n = 9). Healthy participants were matched to participants with hepatic impairment by body weight, age, and sex. Analysis of variance was performed on natural log-transformed milvexian exposure parameters, with hepatic function group as a fixed effect.

Results: Single doses of milvexian 60 mg were generally well tolerated, with no serious adverse events (AEs), bleeding AEs, or discontinuations due to AEs. Geometric mean ratios (90% confidence interval) for total milvexian maximum observed plasma concentration and area under the plasma concentration-time curve from time zero extrapolated to infinite time were 1.180 (0.735-1.895) and 1.168 (0.725-1.882), respectively, for mild hepatic impairment versus normal hepatic function and 1.140 (0.699-1.857) and 0.996 (0.609-1.628), respectively, for moderate hepatic impairment versus normal hepatic function. Across groups, milvexian exposure-related increases were observed for activated partial thromboplastin time.

Conclusion: Milvexian was well tolerated in participants with normal, mildly impaired, and moderately impaired hepatic function. Observed pharmacokinetic changes suggest it is unlikely that dose adjustments will be necessary in patients with mild or moderate hepatic impairment. Clinical Trial RegistrationClinicaltrials.gov identifier: NCT02982707.

Conflict of interest statement

VP, GA, DL, ZW, SL, WC, AB, and BM are full-time employees of Bristol Myers Squibb. LZ is a full-time employee of Janssen Research & Development, LLC.

© 2022. The Author(s).

Figures

Fig 1
Fig 1
Study design
Fig 2
Fig 2
Mean (± standard deviation) plasma (a) total and (b) unbound milvexian concentration versus time profiles
Fig 3
Fig 3
Effects of hepatic impairment on (a) total and (b) unbound milvexian pharmacokinetic properties. AUC(0–t) area under the plasma concentration–time curve from time zero to the time of last quantifiable concentration, AUC(INF) area under the plasma concentration–time curve from time zero extrapolated to infinite time, CI confidence interval, Cmax maximum observed plasma concentration, fu fraction unbound
Fig 4
Fig 4
Mean (± standard deviation) (a) aPTT and (b) FXIc profile over time for milvexian. aPTT activated partial thromboplastin time, FXIc Factor XI clotting activity
Fig 5
Fig 5
Percentage change from baseline in (a) aPTT and (b) FXIc versus plasma concentrations of milvexian. aPTT activated partial thromboplastin time, FXIc Factor XI clotting activity

References

    1. Virani SS, Alonso A, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, et al. Heart disease and stroke statistics —2020 update: a report from the American Heart Association. Circulation. 2020;141(9):e139–e596. doi: 10.1161/CIR.0000000000000757.
    1. GBD 2019 Diseases and Injuries Collaborators Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet. 2020;396(10258):1204–1222. doi: 10.1016/S0140-6736(20)30925-9.
    1. Bracey A, Shatila W, Wilson J. Bleeding in patients receiving non-vitamin K oral anticoagulants: clinical trial evidence. Ther Adv Cardiovasc Dis. 2018;12(12):361–380. doi: 10.1177/1753944718801554.
    1. Johnson EO, Konstandi M, Babis GC, Piagkou M, Soucacos PN. The plasma coagulation cascade: potential targets for novel anticoagulants in major lower limb surgery. Curr Vasc Pharmacol. 2011;9(1):3–10. doi: 10.2174/157016111793744779.
    1. Quan ML, Pinto DJP, Smallheer JM, Ewing WR, Rossi KA, Luettgen JM, et al. Factor XIa inhibitors as new anticoagulants. J Med Chem. 2018;61(17):7425–7447. doi: 10.1021/acs.jmedchem.8b00173.
    1. Pant A, Kopec AK, Luyendyk JP. Role of the blood coagulation cascade in hepatic fibrosis. Am J Physiol Gastrointest Liver Physiol. 2018;315(2):G171–G176. doi: 10.1152/ajpgi.00402.2017.
    1. Adams RL, Bird RJ. Coagulation cascade and therapeutics update: relevance to nephrology. Part 1: overview of coagulation, thrombophilias and history of anticoagulants. Nephrology (Carlton) 2009;14(5):462–470. doi: 10.1111/j.1440-1797.2009.01128.x.
    1. Salomon O, Steinberg DM, Koren-Morag N, Tanne D, Seligsohn U. Reduced incidence of ischemic stroke in patients with severe factor XI deficiency. Blood. 2008;111(8):4113–4117. doi: 10.1182/blood-2007-10-120139.
    1. Salomon O, Steinberg DM, Zucker M, Varon D, Zivelin A, Seligsohn U. Patients with severe factor XI deficiency have a reduced incidence of deep-vein thrombosis. Thromb Haemost. 2011;105(2):269–273. doi: 10.1160/TH10-05-0307.
    1. Rosenthal RL, Dreskin OH, Rosenthal N. Plasma thromboplastin antecedent (PTA) deficiency; clinical, coagulation, therapeutic and hereditary aspects of a new hemophilia-like disease. Blood. 1955;10(2):120–131. doi: 10.1182/blood.V10.2.120.120.
    1. Franchini M, Veneri D, Lippi G. Inherited factor XI deficiency: a concise review. Hematology. 2006;11(5):307–309. doi: 10.1080/10245330600921964.
    1. Lee SE, Choi YJ, Chi SI, Kim HJ, Seo KS. Factor XI deficiency and orthognathic surgery: a case report on anesthesia management. J Dent Anesth Pain Med. 2015;15(1):25–29. doi: 10.17245/jdapm.2015.15.1.25.
    1. Preis M, Hirsch J, Kotler A, Zoabi A, Stein N, Rennert G, et al. Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events. Blood. 2017;129(9):1210–1215. doi: 10.1182/blood-2016-09-742262.
    1. Meijers JC, Tekelenburg WL, Bouma BN, Bertina RM, Rosendaal FR. High levels of coagulation factor XI as a risk factor for venous thrombosis. N Engl J Med. 2000;342(10):696–701. doi: 10.1056/NEJM200003093421004.
    1. Yang DT, Flanders MM, Kim H, Rodgers GM. Elevated factor XI activity levels are associated with an increased odds ratio for cerebrovascular events. Am J Clin Pathol. 2006;126(3):411–415. doi: 10.1309/QC259F09UNMKVP0R.
    1. Merlo C, Wuillemin WA, Redondo M, Furlan M, Sulzer I, Kremer-Hovinga J, et al. Elevated levels of plasma prekallikrein, high molecular weight kininogen and factor XI in coronary heart disease. Atherosclerosis. 2002;161(2):261–267. doi: 10.1016/s0021-9150(01)00666-9.
    1. Berliner JI, Rybicki AC, Kaplan RC, Monrad ES, Freeman R, Billett HH. Elevated levels of factor XI are associated with cardiovascular disease in women. Thromb Res. 2002;107(1–2):55–60. doi: 10.1016/s0049-3848(02)00190-1.
    1. Wong PC, Crain EJ, Bozarth JM, Wu Y, Dilger AK, Wexler RR, et al. Milvexian, an orally bioavailable, small-molecule, reversible, direct inhibitor of factor XIa: in vitro studies and in vivo evaluation in experimental thrombosis in rabbits. J Thromb Haemost. 2021;20(2):399–408. doi: 10.1111/jth.15588.
    1. Graff J, Harder S. Anticoagulant therapy with the oral direct factor Xa inhibitors rivaroxaban, apixaban and edoxaban and the thrombin inhibitor dabigatran etexilate in patients with hepatic impairment. Clin Pharmacokinet. 2013;52(4):243–254. doi: 10.1007/s40262-013-0034-0.
    1. US Food and Drug Administration. Guidance for industry: pharmacokinetics in patients with impaired hepatic function: study design, data analysis, and impact on dosing and labeling. May 2003. . Accessed 27 Oct 2020.
    1. European Medicines Agency. Committee for Medicine Products for Human Use (CHMP). Note for guidance on the evaluation of the pharmacokinetics of medicinal products in patients with impaired renal function. 23 Jun 2004. . Accessed 24 Mar 2021.
    1. Government of Canada. Guidance document: pre-market evaluation of hepatotoxicity in health products. 18 Apr 2012. . Accessed 2 Apr 2021.
    1. Weitz JI, Strony J, Ageno W, Gailani D, Hylek EM, Lassen MR, et al. Milvexian for the prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161–2172. doi: 10.1056/NEJMoa2113194.
    1. . NCT03766581. A study on BMS-986177 for the prevention of stroke in patients receiving aspirin and clopidogrel (AXIOMATIC-SSP). . Accessed 1 Sept 2020.
    1. Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg. 1973;60(8):646–649. doi: 10.1002/bjs.1800600817.
    1. Perera V, Wang Z, Luettgen J, Li D, DeSouza M, Cerra M, et al. First-in-human study of milvexian, an oral, direct, small molecule factor XIa inhibitor. Clin Transl Sci. 2021;15(2):330–342. doi: 10.1111/cts.13148.
    1. Amitrano L, Guardascione MA, Brancaccio V, Balzano A. Coagulation disorders in liver disease. Semin Liver Dis. 2002;22(1):83–96. doi: 10.1055/s-2002-23205.

Source: PubMed

3
Předplatit