Phase 2 Study of the Factor XI Antisense Inhibitor IONIS-FXIRx in Patients With ESRD

Michael Walsh, Claudette Bethune, Andrew Smyth, Jessica Tyrwhitt, Shiangtung W Jung, Rosie Z Yu, Yanfeng Wang, Richard S Geary, Jeffrey Weitz, Sanjay Bhanot, CS4 Investigators, Michael Walsh, Claudette Bethune, Andrew Smyth, Jessica Tyrwhitt, Shiangtung W Jung, Rosie Z Yu, Yanfeng Wang, Richard S Geary, Jeffrey Weitz, Sanjay Bhanot, CS4 Investigators

Abstract

Introduction: Patients with end-stage renal disease (ESRD) requiring hemodialysis (HD) have an increased risk of thrombotic events and bleeding. Antisense reduction of factor XI (FXI) with IONIS-FXIRx is a novel strategy that may safely reduce the risk of thrombotic events.

Methods: This multicenter study enrolled 49 patients receiving HD in 2 parts. First, 6 participants (pharmacokinetics [PK] cohort) received 1 open-label 300 mg dose of IONIS-FXIRx both before and after HD. Subsequently, 43 participants were treated in a double-blind, randomized design with 200 mg or 300 mg IONIS-FXIRx or placebo for 12 weeks. The PK, pharmacodynamics (PD), and adverse events of IONIS-FXIRx were evaluated (ClinicalTrials.gov: NCT02553889).

Results: The PK of IONIS-FXIRx was consistent with previous studies and similar whether injected before or after HD. No accumulation of IONIS-FXIRx was observed after repeat administration. By day 85, mean levels of FXI activity fell 56.0% in the 200 mg group, 70.7% in the 300 mg group, and 3.9% in the placebo group compared with baseline. FXI antigen levels paralleled FXI activity. Dose-dependent prolongation of activated partial thromboplastin time (aPTT) was observed, with no changes in international normalized ratio (INR). IONIS-FXIRx was not associated with drug-related serious adverse events. In the randomized phase of the study, major bleeding events occurred in 0 (0.0%; 200 mg), 1 (6.7%; 300 mg), and 1 (7.7%; placebo) patients and were not considered related to treatment.

Conclusion: IONIS-FXIRx reduced FXI activity in patients with ESRD receiving HD. Further studies are needed to determine the benefit-risk profile of FXI as a therapeutic target for patients who require HD.

Keywords: FXI; anticoagulation; antisense oligonucleotide; hemodialysis; pharmacokinetics; randomized controlled trial.

© 2021 International Society of Nephrology. Published by Elsevier Inc.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Disposition of study participants. Three patients in the placebo group experienced protocol deviations and were excluded from the per-protocol population owing to disallowed concomitant treatment with warfarin (n = 1) and clopidogrel (n = 2). The 200 mg and 300 mg IONIS-FXIRx groups had 3 patients in each group who were excluded from the per-protocol population owing to incomplete dosing to week 8. Adverse events that led to discontinue treatment were as follows: In the PK cohort, 1 participant discontinued owing to sepsis with hemoptysis deemed unlikely related to IONIS-FXIRx. In the 200 mg group, 1 participant discontinued treatment owing to hepatic enzyme increased during hospitalization with pneumonia deemed unlikely related to IONIS-FXIRx. In the 300 mg group, 1 participant discontinued treatment owing to arteriovenous fistula site hemorrhage judged possibly related to IONIS-FXIRx. ITT, intent-to-treat; PK, pharmacokinetics.
Figure 2
Figure 2
Mean (±SD) IONIS-FXIRx concentrations after a single 300 mg dose administered to 6 participants after HD (day 1) and 5 participants before HD (day 29). Note: 1 of the 6 participants did not receive the day 29 dose owing to an intercurrent illness unrelated to IONIS-FXIRx. HD, hemodialysis.
Figure 3
Figure 3
Effect of IONIS-FXIRx 200 mg and 300 mg on FXI activity compared with placebo during the 78-day treatment period and subsequent 85-day washout period. Points represent mean; whiskers represent SEM (intent-to-treat population). Red arrowheads indicate dosing day. FXI, factor XI; LLN, lower limit of normal; ULN, upper limit of normal.

References

    1. De Vriese A.S., Caluwé R., Raggi P. The atrial fibrillation conundrum in dialysis patients. Am Heart J. 2016;174:111–119. doi: 10.1016/j.ahj.2016.01.010.
    1. Preis M., Hirsch J., Kotler A., et al. Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events. Blood. 2017;129:1210–1215. doi: 10.1182/blood-2016-09-742262.
    1. Cushman M., O’Meara E.S., Folsom A.R., Heckbert S.R. Coagulation factors IX through XIII and the risk of future venous thrombosis: the Longitudinal Investigation of thromboembolism Etiology. Blood. 2009;114:2878–2883. doi: 10.1182/blood-2009-05-219915.
    1. Meijers J.C., Tekelenburg W.L., Bouma B.N., Bertina R.M., Rosendaal F.R. High levels of coagulation factor XI as a risk factor for venous thrombosis. N Engl J Med. 2000;342:696–701. doi: 10.1056/NEJM200003093421004.
    1. Salomon O., Steinberg D.M., Koren-Morag N., Tanne D., Seligsohn U. Reduced incidence of ischemic stroke in patients with severe factor XI deficiency. Blood. 2008;111:4113–4117. doi: 10.1182/blood-2007-10-120139.
    1. Salomon O., Steinberg D.M., 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;105:269–273. doi: 10.1160/TH10-05-0307.
    1. Siegerink B., Rosendaal F.R., Algra A. Antigen levels of coagulation factor XII, coagulation factor XI and prekallikrein, and the risk of myocardial infarction and ischemic stroke in young women. J Thromb Haemost. 2014;12:606–613. doi: 10.1111/jth.12531.
    1. Suri M.F., Yamagishi K., Aleksic N., Hannan P.J., Folsom A.R. Novel hemostatic factor levels and risk of ischemic stroke: the Atherosclerosis Risk in Communities (ARIC) study. Cerebrovasc Dis. 2010;29:497–502. doi: 10.1159/000297966.
    1. Büller H.R., Bethune C., Bhanot S., et al. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N Engl J Med. 2015;372:232–240. doi: 10.1056/NEJMoa1405760.
    1. Bethune C., Walsh M., Jung S., et al. Pharmacokinetics and pharmacodynamics of Ionis-FXIRx, an antisense inhibitor of factor XI, in patients with end-stage renal disease on hemodialysis. Blood. 2017;130(suppl 1):1116.
    1. Sagedal S., Hartmann A., Sundstrøm K., Bjørnsen S., Brosstad F. Anticoagulation intensity sufficient for haemodialysis does not prevent activation of coagulation and platelets. Nephrol Dial Transplant. 2001;16:987–993. doi: 10.1093/ndt/16.5.987.
    1. Yu R.Z., Baker B., Chappel A., Geary R.S., Chueng E., Levin A.A. Development of an ultrasensitive noncompetitive hybridization-ligation enzyme-linked immunosorbent assay for the determination of phosphorothioate oligodeoxynucleotide in plasma. Anal Biochem. 2002;304:19–25. doi: 10.1006/abio.2002.5576.
    1. Yu R.Z., Geary R.S., Flaim D.J., et al. Lack of pharmacokinetic interaction of mipomersen sodium (ISIS 301012), a 2’-O-methoxyethyl modified antisense oligonucleotide targeting apolipoprotein B-100 messenger RNA, with simvastatin and ezetimibe. Clin Pharmacokinet. 2009;48:39–50. doi: 10.2165/0003088-200948010-00003.
    1. von dem Borne P.A., Cox L.M., Bouma B.N. Factor XI enhances fibrin generation and inhibits fibrinolysis in a coagulation model initiated by surface-coated tissue factor. Blood Coagul Fibrinolysis. 2006;17:251–257. doi: 10.1097/01.mbc.0000224843.33216.5f.
    1. Bouma B.N., Mosnier L.O., Meijers J.C., Griffin J.H. Factor XI dependent and independent activation of thrombin activatable fibrinolysis inhibitor (TAFI) in plasma associated with clot formation. J Thromb Haemost. 1999;82:1703–1708.
    1. Thomas D., Thelen K., Kraff S., et al. BAY 1213790, a fully human IgG1 antibody targeting coagulation factor XIa: first evaluation of safety, pharmacodynamics, and pharmacokinetics. Res Pract Thromb Haemost. 2019;3:242–253. doi: 10.1002/rth2.12186.
    1. Weitz J.I., Bauersachs R., Becker B., et al. Effect of osocimab in preventing venous thromboembolism among patients undergoing knee arthroplasty: the FOXTROT randomized clinical trial. JAMA. 2020;323:130–139. doi: 10.1001/jama.2019.20687.
    1. Hu A., Niu J., Winkelmayer W.C. Oral anticoagulation in patients with end-stage kidney disease on dialysis and atrial fibrillation. Semin Nephrol. 2018;38:618–628. doi: 10.1016/j.semnephrol.2018.08.006.
    1. Shaw K., Amstutz U., Kim R.B., CPNDS Clinical Recommendation Group Clinical practice recommendations on genetic testing of CYP2C9 and VKORC1 variants in warfarin therapy. Ther Drug Monit. 2015;37:428–436. doi: 10.1097/FTD.0000000000000192.
    1. Hanni C., Petrovitch E., Ali M., et al. Outcomes associated with apixaban vs warfarin in patients with renal dysfunction. Blood Adv. 2020;4:2366–2371. doi: 10.1182/bloodadvances.2019000972.
    1. Geary R.S., Bradley D.J., Watanabe T., et al. Lack of pharmacokinetic interaction for ISIS 113715, a 2’-0-methoxyethyl modified antisense oligonucleotide targeting protein tyrosine phosphatase 1B messenger RNA, with oral antidiabetic compounds metformin, glipizide or rosiglitazone. Clin Pharmacokinet. 2006;45:789–801. doi: 10.2165/00003088-200645080-00003.
    1. Burnier M., Pruijm M., Wuerzner G., Santschi V. Drug adherence in chronic kidney diseases and dialysis. Nephrol Dial Transplant. 2015;30:39–44. doi: 10.1093/ndt/gfu015.
    1. Geary R.S., Norris D., Yu R., Bennett C.F. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev. 2015;87:46–51. doi: 10.1016/j.addr.2015.01.008.
    1. Watanabe T.A., Geary R.S., Levin A.A. Plasma protein binding of an antisense oligonucleotide targeting human ICAM-1 (ISIS 2302) Oligonucleotides. 2006;16:169–180. doi: 10.1089/oli.2006.16.169.
    1. Crooke S.T., Baker B.F., Witztum J.L., et al. The effects of 2’-O-methoxyethyl containing antisense oligonucleotides on platelets in human clinical trials. Nucleic Acid Ther. 2017;27:121–129. doi: 10.1089/nat.2016.0650.

Source: PubMed

3
Abonnere