Abstract
Therapeutic anticoagulation is essential to prevent and treat venous and arterial thromboembolism. The available agents target coagulation factors involved in thrombus formation but are associated with an increased risk of bleeding. Factor XI plays a minor role in haemostasis but contributes substantially to thrombus expansion, making it an attractive target to mitigate bleeding while maintaining antithrombotic efficacy. Various novel inhibitors, including antisense oligonucleotides, monoclonal antibodies and small molecules, have been developed. Phase II trials in orthopaedic surgery showed dose-dependent reductions in venous thromboembolism without significantly increasing bleeding compared with enoxaparin. In the first phase III trial of a small-molecule inhibitor of activated factor XI in patients with atrial fibrillation, asundexian was associated with a reduction in bleeding but also a higher risk of stroke, compared with apixaban. Factor XI inhibitors appear safe and hold promise for secondary prevention in myocardial infarction and ischaemic stroke, with ongoing phase III trials assessing their broader efficacy and safety. This Review discusses the rationale, pharmacology, evidence and future directions of factor XI inhibitors across various clinical settings.
Key points
Factor XI (FXI) inhibitors aim to uncouple haemostasis from thrombosis, presenting a novel approach to anticoagulation, with a reduced risk of bleeding compared with that associated with traditional anticoagulant agents.
Several phase II studies indicate that FXI inhibitors decrease thrombotic complications dose-dependently, without corresponding increases in bleeding, suggesting a favourable safety profile.
FXI inhibitors showed non-inferiority and potential superiority to low-molecular-weight heparin for the prevention of venous thromboembolism in patients undergoing orthopaedic surgery, with minimal bleeding risks observed at therapeutic doses.
Milvexian and abelacimab are theoretically promising for reducing the risk of bleeding associated with thromboprophylaxis in patients with atrial fibrillation; phase III clinical trials of safety and efficacy are ongoing.
In phase II trials involving patients with non-cardioembolic ischaemic stroke, no significant benefit of FXI inhibitors was found in terms of the primary ischaemic end points, but future phase III trials might find benefits in selected patients with large-vessel atherosclerosis.
The ongoing LIBREXIA-ACS trial will assess the use of milvexian in 16,000 patients with recent acute coronary syndrome.
Access through your institution
Buy or subscribe
This is a preview of subscription content, access via your institution
Access options
Access through your institution
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Learn more
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Learn more
Buy this article
Purchase on SpringerLink
Instant access to full article PDF
Buy now
Prices may be subject to local taxes which are calculated during checkout
Additional access options:
Log in
Learn about institutional subscriptions
Read our FAQs
Contact customer support
Fig. 1: Contribution of factor XI to haemostasis and thrombosis.
Fig. 2: Mechanisms of action of anticoagulant drugs.
Fig. 3: Classification and mechanisms of action of factor XI inhibitors.
References
Lozano, R. et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380, 2095–2128 (2012).
PubMedPubMed CentralGoogle Scholar
Mueller, R. L. & Scheidt, S. History of drugs for thrombotic disease. Discovery, development, and directions for the future. Circulation 89, 432–449 (1994).
CASPubMedGoogle Scholar
Ruff, C. T. et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet 383, 955–962 (2014).
CASPubMedGoogle Scholar
Chan, N., Sobieraj-Teague, M. & Eikelboom, J. W. Direct oral anticoagulants: evidence and unresolved issues. Lancet 396, 1767–1776 (2020).
CASPubMedGoogle Scholar
Han, T. S. et al. Anticoagulation therapy in patients with stroke and atrial fibrillation: a registry-based study of acute stroke care in Surrey, UK. BMJ Open. 8, e022558 (2018).
PubMedPubMed CentralGoogle Scholar
Greco, A. et al. Pharmacology and clinical development of factor XI inhibitors. Circulation 147, 897–913 (2023).
CASPubMedGoogle Scholar
Gigante, B. & Ten Cate, H. Factor XI inhibitors in patients with cardiovascular disease and a high risk of bleeding: a cautionary tale. Nat. Rev. Cardiol. 20, 511–512 (2023).
PubMedGoogle Scholar
Asselta, R. et al. Exploring the global landscape of genetic variation in coagulation factor XI deficiency. Blood 130, e1–e6 (2017).
CASPubMedGoogle Scholar
Bolton-Maggs, P. H. Factor XI deficiency and its management. Haemophilia 6, 100–109 (2000).
PubMedGoogle Scholar
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. 342, 696–701 (2000).
CASPubMedGoogle Scholar
Preis, M. et al. Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events. Blood 129, 1210–1215 (2017).
CASPubMedGoogle Scholar
Salomon, O. et al. Patients with severe factor XI deficiency have a reduced incidence of deep-vein thrombosis. Thromb. Haemost. 105, 269–273 (2011).
CASPubMedGoogle Scholar
Paszek, E., Polak, M. & Undas, A. Elevated factor XI is associated with increased risk of recurrent cerebral venous sinus thrombosis: a cohort study. J. Thromb. Thrombolysis 57, 402–407 (2024).
CASPubMedGoogle Scholar
Wang, X. et al. Effects of factor IX or factor XI deficiency on ferric chloride-induced carotid artery occlusion in mice. J. Thromb. Haemost. 3, 695–702 (2005).
CASPubMedGoogle Scholar
Tucker, E. I. et al. Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI. Blood 113, 936–944 (2009).
CASPubMedPubMed CentralGoogle Scholar
Crosby, J. R. et al. Antithrombotic effect of antisense factor XI oligonucleotide treatment in primates. Arterioscler. Thromb. Vasc. Biol. 33, 1670–1678 (2013).
CASPubMedPubMed CentralGoogle Scholar
Yau, J. W. et al. Selective depletion of factor XI or factor XII with antisense oligonucleotides attenuates catheter thrombosis in rabbits. Blood 123, 2102–2107 (2014).
CASPubMedGoogle Scholar
Gailani, D. & Gruber, A. Factor XI as a therapeutic target. Arterioscler. Thromb. Vasc. Biol. 36, 1316–1322 (2016).
CASPubMedPubMed CentralGoogle Scholar
Grover, S. P. & Mackman, N. Intrinsic pathway of coagulation and thrombosis. Arterioscler. Thromb. Vasc. Biol. 39, 331–338 (2019).
CASPubMedGoogle Scholar
Hoffman, M. & Monroe, D. M. 3rd A cell-based model of hemostasis. Thromb. Haemost. 85, 958–965 (2001).
CASPubMedGoogle Scholar
Becker, R. C. Cell-based models of coagulation: a paradigm in evolution. J. Thromb. Thrombolysis 20, 65–68 (2005).
PubMedGoogle Scholar
Ho, K. M. & Pavey, W. Applying the cell-based coagulation model in the management of critical bleeding. Anaesth. Intensive Care 45, 166–176 (2017).
CASPubMedGoogle Scholar
Aleman, M. M. et al. Factor XIII activity mediates red blood cell retention in venous thrombi. J. Clin. Invest. 124, 3590–3600 (2014).
CASPubMedPubMed CentralGoogle Scholar
Mann, K. G., Brummel-Ziedins, K., Orfeo, T. & Butenas, S. Models of blood coagulation. Blood Cell Mol. Dis. 36, 108–117 (2006).
CASGoogle Scholar
Ying, J. W., Lin, C. Y. & Anne, C. M. The cell-based model of coagulation. LearnHaemwww.learnhaem.com/courses/coagulation/lessons/normal-haemostasis/topic/the-cell-based-model-of-coagulation/ (2024).
Hsu, C., Hutt, E., Bloomfield, D. M., Gailani, D. & Weitz, J. I. Factor XI inhibition to uncouple thrombosis from hemostasis: JACC review topic of the week. J. Am. Coll. Cardiol. 78, 625–631 (2021).
CASPubMedPubMed CentralGoogle Scholar
Hoffman, M., Monroe, D. M., Oliver, J. A. & Roberts, H. R. Factors IXa and Xa play distinct roles in tissue factor-dependent initiation of coagulation. Blood 86, 1794–1801 (1995).
CASPubMedGoogle Scholar
Brummel-Ziedins, K. E., Branda, R. F., Butenas, S. & Mann, K. G. Discordant fibrin formation in hemophilia. J. Thromb. Haemost. 7, 825–832 (2009).
CASPubMedPubMed CentralGoogle Scholar
Yau, J. W. et al. Corn trypsin inhibitor coating attenuates the prothrombotic properties of catheters in vitro and in vivo. Acta Biomater. 8, 4092–4100 (2012).
CASPubMedGoogle Scholar
Frank, R. D. et al. Role of contact system activation in hemodialyzer-induced thrombogenicity. Kidney Int. 60, 1972–1981 (2001).
CASPubMedGoogle Scholar
Bernacca, G. M., Gulbransen, M. J., Wilkinson, R. & Wheatley, D. J. In vitro blood compatibility of surface-modified polyurethanes. Biomaterials 19, 1151–1165 (1998).
CASPubMedGoogle Scholar
Tillman, B. & Gailani, D. Inhibition of factors XI and XII for prevention of thrombosis induced by artificial surfaces. Semin. Thromb. Hemost. 44, 60–69 (2018).
CASPubMedGoogle Scholar
Angiolillo, D. J. & Capodanno, D. Uncoupling thrombosis and hemostasis by inhibiting factor XI. N. Engl. J. Med. 392, 400–403 (2025).
PubMedGoogle Scholar
Ngo, A. T. P. et al. Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice. J. Thromb. Haemost. 19, 1001–1017 (2021).
CASPubMedPubMed CentralGoogle Scholar
Puy, C. et al. Coagulation factor XI regulates endothelial cell permeability and barrier function in vitro and in vivo. Blood 144, 1821–1833 (2024).
CASPubMedGoogle Scholar
Pallares Robles, A. et al. Association of FXI activity with thrombo-inflammation, extracellular matrix, lipid metabolism and apoptosis in venous thrombosis. Sci. Rep. 12, 9761 (2022).
CASPubMedPubMed CentralGoogle Scholar
Kossmann, S. et al. Platelet-localized FXI promotes a vascular coagulation-inflammatory circuit in arterial hypertension. Sci. Transl. Med. 9, eaah4923 (2017).
PubMedGoogle Scholar
Cao, Y. et al. Liver-heart cross-talk mediated by coagulation factor XI protects against heart failure. Science 377, 1399–1406 (2022).
CASPubMedPubMed CentralGoogle Scholar
Ji, Y. et al. Association of coagulation factor XI level with cardiovascular events and cardiac function in community-dwelling adults: from ARIC and CHS. Circulationhttps://doi.org/10.1161/CIRCULATIONAHA.124.070278 (2024).
ArticlePubMedPubMed CentralGoogle Scholar
Lova, A. et al. Congenital factor XI deficiency and risk of heart failure in humans. J. Thromb. Haemost. 21, 2626–2629 (2023).
PubMedGoogle Scholar
Liu, T. et al. Pharmacokinetics, pharmacodynamics, and safety of fesomersen, a novel antisense inhibitor of factor XI, in healthy Chinese, Japanese, and Caucasian volunteers. Clin. Transl. Sci. 17, e13784 (2024).
CASPubMedPubMed CentralGoogle Scholar
Zhang, H. et al. Inhibition of the intrinsic coagulation pathway factor XI by antisense oligonucleotides: a novel antithrombotic strategy with lowered bleeding risk. Blood 116, 4684–4692 (2010).
CASPubMedGoogle Scholar
Crooke, S. T. et al. The effects of 2’-O-methoxyethyl containing antisense oligonucleotides on platelets in human clinical trials. Nucleic Acid. Ther. 27, 121–129 (2017).
CASPubMedPubMed CentralGoogle Scholar
Crooke, S. T., Wang, S., Vickers, T. A., Shen, W. & Liang, X. H. Cellular uptake and trafficking of antisense oligonucleotides. Nat. Biotechnol. 35, 230–237 (2017).
CASPubMedGoogle Scholar
Geary, R. S. 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. 45, 789–801 (2006).
CASPubMedGoogle Scholar
Shemesh, C. S. et al. Assessment of the drug interaction potential of unconjugated and GalNAc3-conjugated 2’-MOE-ASOs. Mol. Ther. Nucleic Acids 9, 34–47 (2017).
CASPubMedPubMed CentralGoogle Scholar
Wang, Y., Yu, R. Z., Henry, S. & Geary, R. S. Pharmacokinetics and clinical pharmacology considerations of GalNAc3-conjugated antisense oligonucleotides. Expert. Opin. Drug. Metab. Toxicol. 15, 475–485 (2019).
CASPubMedGoogle Scholar
Verhamme, P. et al. Single intravenous administration of TB-402 for the prophylaxis of venous thromboembolism after total knee replacement: a dose-escalating, randomized, controlled trial. J. Thromb. Haemost. 9, 664–671 (2011).
CASPubMedGoogle Scholar
Verhamme, P. et al. Single-dose TB-402 or rivaroxaban for the prevention of venous thromboembolism after total hip replacement. A randomised, controlled trial. Thromb. Haemost. 109, 1091–1098 (2013).
CASPubMedGoogle Scholar
Thomas, D. et al. BAY 1213790, a fully human IgG1 antibody targeting coagulation factor XIa: first evaluation of safety, pharmacodynamics, and pharmacokinetics. Res. Pract. Thromb. Haemost. 3, 242–253 (2019).
CASPubMedPubMed CentralGoogle Scholar
Yi, B. A. et al. Pharmacokinetics and pharmacodynamics of abelacimab (MAA868), a novel dual inhibitor of factor XI and factor XIa. J. Thromb. Haemost. 20, 307–315 (2022).
CASPubMedGoogle Scholar
Koch, A. W. et al. MAA868, a novel FXI antibody with a unique binding mode, shows durable effects on markers of anticoagulation in humans. Blood 133, 1507–1516 (2019).
CASPubMedGoogle Scholar
Ruff, C. T. et al. Abelacimab versus rivaroxaban in patients with atrial fibrillation. N. Engl. J. Med. 392, 361–371 (2025).
CASPubMedGoogle Scholar
Lorentz, C. U. et al. Contact activation inhibitor and factor XI antibody, AB023, produces safe, dose-dependent anticoagulation in a phase 1 first-in-human trial. Arterioscler. Thromb. Vasc. Biol. 39, 799–809 (2019).
CASPubMedPubMed CentralGoogle Scholar
Lorentz, C. U. et al. The contact activation inhibitor AB023 in heparin-free hemodialysis: results of a randomized phase 2 clinical trial. Blood 138, 2173–2184 (2021).
CASPubMedPubMed CentralGoogle Scholar
Thomas, D. et al. First evaluation of the safety, pharmacokinetics, and pharmacodynamics of BAY 2433334, a small molecule targeting coagulation factor XIa. J. Thromb. Haemost. 19, 2407–2416 (2021).
CASPubMedPubMed CentralGoogle Scholar
Kubitza, D. et al. Pharmacokinetics, pharmacodynamics and safety of BAY 2433334, a novel activated factor XI inhibitor, in healthy volunteers: a randomized phase 1 multiple-dose study. Br. J. Clin. Pharmacol. 88, 3447–3462 (2022).
CASPubMedGoogle Scholar
Heitmeier, S. et al. Pharmacological profile of asundexian, a novel, orally bioavailable inhibitor of factor XIa. J. Thromb. Haemost. 20, 1400–1411 (2022).
CASPubMedPubMed CentralGoogle Scholar
Wong, P. C. 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. 20, 399–408 (2022).
CASPubMedGoogle Scholar
Perera, V. et al. First-in-human study of milvexian, an oral, direct, small molecule factor XIa inhibitor. Clin. Transl. Sci. 15, 330–342 (2022).
CASPubMedGoogle Scholar
Pollack, C. V. Jr., Kurz, M. A. & Hayward, N. J. EP-7041, a factor XIa inhibitor as a potential antithrombotic strategy in extracorporeal membrane oxygenation: a brief report. Crit. Care Explor. 2, e0196 (2020).
PubMedPubMed CentralGoogle Scholar
Zhang, J. Y. et al. Pharmacokinetics, pharmacodynamics, and safety of frunexian in healthy Chinese volunteer adults: a randomized dose-escalation phase I study. Clin. Transl. Sci. 17, e13787 (2024).
CASPubMedPubMed CentralGoogle Scholar
Pinto, D. J. P. et al. Discovery of a parenteral small molecule coagulation factor XIa inhibitor clinical candidate (BMS-962212). J. Med. Chem. 60, 9703–9723 (2017).
CASPubMedGoogle Scholar
Perera, V. et al. First-in-human study to assess the safety, pharmacokinetics and pharmacodynamics of BMS-962212, a direct, reversible, small molecule factor XIa inhibitor in non-Japanese and Japanese healthy subjects. Br. J. Clin. Pharmacol. 84, 876–887 (2018).
CASPubMedPubMed CentralGoogle Scholar
Wong, P. C. & Quan, M. L. Improved efficacy/safety profile of factor XIa inhibitor BMS-724296 versus factor Xa inhibitor apixaban and thrombin inhibitor dabigatran in cynomolgus monkeys. Res. Pract. Thromb. Haemost. 5, e12524 (2021).
CASPubMedPubMed CentralGoogle Scholar
Al-Horani, R. A., Ponnusamy, P., Mehta, A. Y., Gailani, D. & Desai, U. R. Sulfated pentagalloylglucoside is a potent, allosteric, and selective inhibitor of factor XIa. J. Med. Chem. 56, 867–878 (2013).
CASPubMedPubMed CentralGoogle Scholar
Decrem, Y. et al. Ir-CPI, a coagulation contact phase inhibitor from the tick Ixodes ricinus, inhibits thrombus formation without impairing hemostasis. J. Exp. Med. 206, 2381–2395 (2009).
CASPubMedPubMed CentralGoogle Scholar
Chen, W., Carvalho, L. P., Chan, M. Y., Kini, R. M. & Kang, T. S. Fasxiator, a novel factor XIa inhibitor from snake venom, and its site-specific mutagenesis to improve potency and selectivity. J. Thromb. Haemost. 13, 248–261 (2015).
CASPubMedGoogle Scholar
Li, D. et al. Identification of an anticoagulant peptide that inhibits both fXIa and fVIIa/tissue factor from the blood-feeding nematode Ancylostoma caninum. Biochem. Biophys. Res. Commun. 392, 155–159 (2010).
CASPubMedGoogle Scholar
Ma, D. et al. Desmolaris, a novel factor XIa anticoagulant from the salivary gland of the vampire bat (Desmodus rotundus) inhibits inflammation and thrombosis in vivo. Blood 122, 4094–4106 (2013).
CASPubMedPubMed CentralGoogle Scholar
Assumpção, T. C. et al. In vitro mode of action and anti-thrombotic activity of boophilin, a multifunctional Kunitz protease inhibitor from the midgut of a tick vector of babesiosis, Rhipicephalus microplus. PLoS Negl. Trop. Dis. 10, e0004298 (2016).
PubMedPubMed CentralGoogle Scholar
Manzato, V. M. et al. Boophilin D1, a Kunitz type protease inhibitor, as a source of inhibitors for the ZIKA virus NS2B-NS3 protease. Biochimie 214, 96–101 (2023).
CASPubMedGoogle Scholar
Donkor, D. A. et al. Selection and characterization of a DNA aptamer inhibiting coagulation factor XIa. Sci. Rep. 7, 2102 (2017).
PubMedPubMed CentralGoogle Scholar
Woodruff, R. S. et al. Generation and characterization of aptamers targeting factor XIa. Thromb. Res. 156, 134–141 (2017).
CASPubMedPubMed CentralGoogle Scholar
Gan, L. et al. Inhibition of Factor XI using a GalNAc-siRNA RBD5049: a novel antithrombotic drug with high potency and long duration [abstract]. Eur. Heart J. 44, ehad655.2843 (2023).
Google Scholar
Büller, H. R. et al. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N. Engl. J. Med. 372, 232–240 (2015).
PubMedGoogle Scholar
Weitz, J. I. et al. Effect of osocimab in preventing venous thromboembolism among patients undergoing knee arthroplasty: the FOXTROT randomized clinical trial. JAMA 323, 130–139 (2020).
CASPubMedPubMed CentralGoogle Scholar
Verhamme, P. et al. Abelacimab for prevention of venous thromboembolism. N. Engl. J. Med. 385, 609–617 (2021).
CASPubMedGoogle Scholar
Weitz, J. I. et al. Milvexian for the prevention of venous thromboembolism. N. Engl. J. Med. 385, 2161–2172 (2021).
CASPubMedPubMed CentralGoogle Scholar
Jain, S. S. et al. Milvexian vs apixaban fibrillation: the LIBREXIA atrial fibrillation trial rationale and design. Am. Heart J. 277, 145–158 (2024).
CASPubMedGoogle Scholar
Falanga, A. & Marchetti, M. Cancer-associated thrombosis: enhanced awareness and pathophysiologic complexity. J. Thromb. Haemost. 21, 1397–1408 (2023).
CASPubMedGoogle Scholar
de Winter, M. A. et al. Estimating bleeding risk in patients with cancer-associated thrombosis: evaluation of existing risk scores and development of a new risk score. Thromb. Haemost. 122, 818–829 (2022).
PubMedGoogle Scholar
Kraaijpoel, N. et al. Clinical impact of bleeding in cancer-associated venous thromboembolism: results from the hokusai VTE cancer study. Thromb. Haemost. 118, 1439–1449 (2018).
PubMedGoogle Scholar
Otani, H. et al. Difficulty swallowing and food bolus obstruction in advanced cancer: association with the cachexia-related quality of life. Ann. Palliat. Med. 12, 717–728 (2023).
PubMedGoogle Scholar
Agnelli, G. et al. Apixaban and dalteparin for the treatment of venous thromboembolism in patients with different sites of cancer. Thromb. Haemost. 122, 796–807 (2022).
PubMedGoogle Scholar
Farge, D. et al. 2022 international clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer, including patients with COVID-19. Lancet Oncol. 23, e334–e347 (2022).
CASPubMedPubMed CentralGoogle Scholar
Farge, D. et al. 2019 international clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer. Lancet Oncol. 20, e566–e581 (2019).
PubMedGoogle Scholar
Stevens, S. M. et al. Antithrombotic therapy for VTE disease: second update of the CHEST guideline and expert panel report. Chest 160, e545–e608 (2021).
CASPubMedGoogle Scholar
Mäder, J. et al. Effect of factor XI inhibition on tumor cell-induced coagulation activation. J. Thromb. Haemost. 22, 199–212 (2024).
PubMedGoogle Scholar
Aggarwal, R. et al. Development and validation of the DOAC score: a novel bleeding risk prediction tool for patients with atrial fibrillation on direct-acting oral anticoagulants. Circulation 148, 936–946 (2023).
CASPubMedPubMed CentralGoogle Scholar
White, R. H., Chew, H. & Wun, T. Targeting patients for anticoagulant prophylaxis trials in patients with cancer: who is at highest risk? Thromb. Res. 120, S29–S40 (2007).
PubMedGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05171049 (2024).
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05171075 (2024).
Gibb, S., Engelhardt, S., von Dincklage, F. & Kuhn, S. O. Incidence and onset of central venous catheter-related thrombosis in critically ill surgical patients: a prospective observational single-center study. J. Clin. Anesth. 97, 111556 (2024).
PubMedGoogle Scholar
Pfeffer, M. A. et al. Factor XI inhibition for the prevention of catheter-associated thrombosis in patients with cancer undergoing central line placement: a phase 2 clinical trial. Arterioscler. Thromb. Vasc. Biol. 44, 290–299 (2024).
CASPubMedGoogle Scholar
Hughes, S., Szeki, I., Nash, M. J. & Thachil, J. Anticoagulation in chronic kidney disease patients – the practical aspects. Clin. Kidney J. 7, 442–449 (2014).
CASPubMedPubMed CentralGoogle Scholar
Reinecke, H. et al. Dilemmas in the management of atrial fibrillation in chronic kidney disease. J. Am. Soc. Nephrol. 20, 705–711 (2009).
CASPubMedGoogle Scholar
Potpara, T. S., Ferro, C. J. & Lip, G. Y. H. Use of oral anticoagulants in patients with atrial fibrillation and renal dysfunction. Nat. Rev. Nephrol. 14, 337–351 (2018).
CASPubMedGoogle Scholar
Kaw, D. & Malhotra, D. Platelet dysfunction and end-stage renal disease. Semin. Dial. 19, 317–322 (2006).
PubMedGoogle Scholar
Claudel, S. E., Miles, L. A. & Murea, M. Anticoagulation in hemodialysis: a narrative review. Semin. Dial. 34, 103–115 (2021).
PubMedGoogle Scholar
Dahal, K., Kunwar, S., Rijal, J., Schulman, P. & Lee, J. Stroke, major bleeding, and mortality outcomes in warfarin users with atrial fibrillation and chronic kidney disease: a meta-analysis of observational studies. Chest 149, 951–959 (2016).
PubMedGoogle Scholar
De Vriese, A. S., Caluwé, R. & Raggi, P. The atrial fibrillation conundrum in dialysis patients. Am. Heart J. 174, 111–119 (2016).
PubMedGoogle Scholar
Reinecke, H. et al. A randomized controlled trial comparing apixaban with the vitamin K antagonist phenprocoumon in patients on chronic hemodialysis: the AXADIA-AFNET 8 study. Circulation 147, 296–309 (2023).
CASPubMedGoogle Scholar
Pokorney, S. D. et al. Apixaban for patients with atrial fibrillation on hemodialysis: a multicenter randomized controlled trial. Circulation 146, 1735–1745 (2022).
PubMedGoogle Scholar
Walsh, M. et al. Phase 2 study of the factor XI antisense inhibitor IONIS-FXIRx in patients with ESRD. Kidney Int. Rep. 7, 200–209 (2022).
PubMedGoogle Scholar
Winkelmayer, W. C. et al. A phase II randomized controlled trial evaluated antithrombotic treatment with fesomersen in patients with kidney failure on hemodialysis. Kidney Int. 106, 145–153 (2024).
CASPubMedGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT03358030 (2023).
Weitz, J. I. et al. Anticoagulation with osocimab in patients with kidney failure undergoing hemodialysis: a randomized phase 2 trial. Nat. Med. 30, 435–442 (2024).
CASPubMedPubMed CentralGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05027074 (2025).
Staerk, L., Sherer, J. A., Ko, D., Benjamin, E. J. & Helm, R. H. Atrial fibrillation: epidemiology, pathophysiology, and clinical outcomes. Circ. Res. 120, 1501–1517 (2017).
CASPubMedPubMed CentralGoogle Scholar
Lee, E. et al. Mortality and causes of death in patients with atrial fibrillation: a nationwide population-based study. PLoS ONE 13, e0209687 (2018).
PubMedPubMed CentralGoogle Scholar
Michaud, G. F. & Stevenson, W. G. Atrial fibrillation. N. Engl. J. Med. 384, 353–361 (2021).
PubMedGoogle Scholar
January, C. T. et al. 2019 AHA/ACC/HRS focused update of the 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J. Am. Coll. Cardiol. 74, 104–132 (2019).
PubMedGoogle Scholar
Hindricks, G. et al. 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): the task force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur. Heart J. 42, 373–498 (2021).
PubMedGoogle Scholar
Chai-Adisaksopha, C., Crowther, M., Isayama, T. & Lim, W. The impact of bleeding complications in patients receiving target-specific oral anticoagulants: a systematic review and meta-analysis. Blood 124, 2450–2458 (2014).
CASPubMedGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05712200 (2025).
Piccini, J. P. et al. Safety of the oral factor XIa inhibitor asundexian compared with apixaban in patients with atrial fibrillation (PACIFIC-AF): a multicentre, randomised, double-blind, double-dummy, dose-finding phase 2 study. Lancet 399, 1383–1390 (2022).
CASPubMedGoogle Scholar
Piccini, J. P. et al. Asundexian versus apixaban in patients with atrial fibrillation. N. Engl. J. Med. 392, 23–32 (2025).
CASPubMedGoogle Scholar
Vassart, J. et al. Asundexian in atrial fibrillation: can pharmacodynamic data explain the failure? Thromb. Res. 236, 236–239 (2024).
CASPubMedGoogle Scholar
Visser, M., Heitmeier, S., Ten Cate, H. & Spronk, H. M. H. Role of factor XIa and plasma kallikrein in arterial and venous thrombosis. Thromb. Haemost. 120, 883–993 (2020).
PubMedGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05757869 (2025).
Lopes, R. D. et al. Apixaban for reduction in stroke and other thromboembolic events in atrial fibrillation (ARISTOTLE) trial: design and rationale. Am. Heart J. 159, 331–339 (2010).
CASPubMedGoogle Scholar
Kleindorfer, D. O. et al. 2021 guideline for the prevention of stroke in patients with stroke and transient ischemic attack: a guideline from the American Heart Association/American Stroke Association. Stroke 52, e364–e467 (2021).
PubMedGoogle Scholar
Capodanno, D. & Angiolillo, D. J. Oral antithrombotic therapy for the prevention of recurrent cerebrovascular events. Eur. Heart J. Cardiovasc. Pharmacother. 8, 383–391 (2022).
PubMedGoogle Scholar
Vermeer, S. E. et al. Silent brain infarcts and the risk of dementia and cognitive decline. N. Engl. J. Med. 348, 1215–1222 (2003).
PubMedGoogle Scholar
Durrani, R., Hill, M. D. & Smith, E. E. Preventing covert brain infarct-related cognitive impairment and dementia. Can. J. Neurol. Sci. 47, 456–463 (2020).
PubMedGoogle Scholar
Shoamanesh, A. et al. Factor XIa inhibition with asundexian after acute non-cardioembolic ischaemic stroke (PACIFIC-Stroke): an international, randomised, double-blind, placebo-controlled, phase 2b trial. Lancet 400, 997–1007 (2022).
CASPubMedGoogle Scholar
Sharma, M. et al. Safety and efficacy of factor XIa inhibition with milvexian for secondary stroke prevention (AXIOMATIC-SSP): a phase 2, international, randomised, double-blind, placebo-controlled, dose-finding trial. Lancet Neurol. 23, 46–59 (2024).
CASPubMedPubMed CentralGoogle Scholar
US National Library of Medicine. ClinicalTrials.govwww.clinicaltrials.gov/study/NCT05686070 (2025).
US National Library of Medicine. ClinicalTrials.govwww.clinicaltrials.gov/study/NCT05702034 (2025).
Capodanno, D., Alfonso, F., Levine, G. N., Valgimigli, M. & Angiolillo, D. J. ACC/AHA versus ESC guidelines on dual antiplatelet therapy: JACC guideline comparison. J. Am. Coll. Cardiol. 72, 2915–2931 (2018).
PubMedGoogle Scholar
Wiviott, S. D. et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N. Engl. J. Med. 357, 2001–2015 (2007).
CASPubMedGoogle Scholar
Wallentin, L. et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N. Engl. J. Med. 361, 1045–1057 (2009).
CASPubMedGoogle Scholar
Bahit, M. C. & Gibson, C. M. Thrombin as target for prevention of recurrent events after acute coronary syndromes. Thromb. Res. 235, 116–121 (2024).
CASPubMedGoogle Scholar
Eikelboom, J. W. et al. Rivaroxaban with or without aspirin in stable cardiovascular disease. N. Engl. J. Med. 377, 1319–1330 (2017).
CASPubMedGoogle Scholar
Mega, J. L. et al. Rivaroxaban in patients with a recent acute coronary syndrome. N. Engl. J. Med. 366, 9–19 (2012).
CASPubMedGoogle Scholar
Morrow, D. A. et al. Vorapaxar in the secondary prevention of atherothrombotic events. N. Engl. J. Med. 366, 1404–1413 (2012).
CASPubMedGoogle Scholar
Capodanno, D. et al. Defining strategies of modulation of antiplatelet therapy in patients with coronary artery disease: a consensus document from the Academic Research Consortium. Circulation 147, 1933–1944 (2023).
PubMedGoogle Scholar
Rao, S. V. et al. A multicenter, phase 2, randomized, placebo-controlled, double-blind, parallel-group, dose-finding trial of the oral factor XIa inhibitor asundexian to prevent adverse cardiovascular outcomes after acute myocardial infarction. Circulation 146, 1196–1206 (2022).
CASPubMedGoogle Scholar
US National Library of Medicine. ClinicalTrials.govclinicaltrials.gov/study/NCT05754957 (2025).
Patel, S. Long-acting factor XI inhibition and periprocedural bleeding: a secondary analysis from AZALEA-TIMI 71 [abstract]. ESC 365esc365.escardio.org/presentation/290273 (2024).
Download references
Author information
Authors and Affiliations
Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
Davide Capodanno
Duke University Medical Center, Durham, NC, USA
John H. Alexander, Renato D. Lopes, Manesh R. Patel & Jonathan P. Piccini
INECO Neurociencias, Rosario, Argentina
M. Cecilia Bahit
BAIM Institute for Clinical Research, Boston, MA, USA
M. Cecilia Bahit
Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
John W. Eikelboom, Shamir R. Mehta & Jeffrey I. Weitz
Hamilton Health Sciences, Hamilton, Ontario, Canada
John W. Eikelboom, Shamir R. Mehta & Jeffrey I. Weitz
Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
C. Michael Gibson
St. Michael’s Hospital, Unity Health Toronto, Toronto, Ontario, Canada
Shaun G. Goodman
Peter Munk Cardiac Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
Shaun G. Goodman
Canadian VIGOUR Centre, University of Alberta, Edmonton, Alberta, Canada
Shaun G. Goodman
Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University Medical School, Newcastle upon Tyne, UK
Vijay Kunadian
Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
Gregory Y. H. Lip
Liverpool John Moores University, Liverpool, UK
Gregory Y. H. Lip
Liverpool Heart and Chest Hospital, Liverpool, UK
Gregory Y. H. Lip
Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
Gregory Y. H. Lip
Icahn School of Medicine at Mount Sinai, New York, NY, USA
Roxana Mehran
New York University School of Medicine, New York, NY, USA
Sunil V. Rao
TIMI Study Group, Division of Cardiovascular Medicine, Brigham & Women’s Hospital, Boston, MA, USA
Christian T. Ruff
Harvard Medical School, Boston, MA, USA
Christian T. Ruff
Université Paris-Cité, INSERM-UMR1148, Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, French Alliance for Cardiovascular Trials, Paris, France
P. Gabriel Steg
Institut Universitaire de France, Paris, France
P. Gabriel Steg
Division of Cardiology, University of Florida College of Medicine, Jacksonville, FL, USA
Dominick J. Angiolillo
Authors
Davide Capodanno
View author publications
You can also search for this author inPubMedGoogle Scholar
2. John H. Alexander
View author publications
You can also search for this author inPubMedGoogle Scholar
3. M. Cecilia Bahit
View author publications
You can also search for this author inPubMedGoogle Scholar
4. John W. Eikelboom
View author publications
You can also search for this author inPubMedGoogle Scholar
5. C. Michael Gibson
View author publications
You can also search for this author inPubMedGoogle Scholar
6. Shaun G. Goodman
View author publications
You can also search for this author inPubMedGoogle Scholar
7. Vijay Kunadian
View author publications
You can also search for this author inPubMedGoogle Scholar
8. Gregory Y. H. Lip
View author publications
You can also search for this author inPubMedGoogle Scholar
9. Renato D. Lopes
View author publications
You can also search for this author inPubMedGoogle Scholar
10. Roxana Mehran
View author publications
You can also search for this author inPubMedGoogle Scholar
11. Shamir R. Mehta
View author publications
You can also search for this author inPubMedGoogle Scholar
12. Manesh R. Patel
View author publications
You can also search for this author inPubMedGoogle Scholar
13. Jonathan P. Piccini
View author publications
You can also search for this author inPubMedGoogle Scholar
14. Sunil V. Rao
View author publications
You can also search for this author inPubMedGoogle Scholar
15. Christian T. Ruff
View author publications
You can also search for this author inPubMedGoogle Scholar
16. P. Gabriel Steg
View author publications
You can also search for this author inPubMedGoogle Scholar
17. Jeffrey I. Weitz
View author publications
You can also search for this author inPubMedGoogle Scholar
18. Dominick J. Angiolillo
View author publications
You can also search for this author inPubMedGoogle Scholar
Contributions
D.C. and D.J.A. researched data for the article and wrote the manuscript. All the authors discussed its content and reviewed/edited it before submission.
Corresponding author
Correspondence to Dominick J. Angiolillo.
Ethics declarations
Competing interests
D.C. has received consulting fees or honoraria from Bristol Myers Squibb, Daiichi Sankyo, Novo Nordisk, Sanofi and Terumo. J.H.A. reports research grants through Duke University from Artivion/CryoLife, Bayer, Bristol Myers Squibb, CSL Behring, Ferring and Humacyte; and honoraria or consulting payments from AbbVie, AtriCure, Artivion/CryoLife, Bayer, Bristol Myers Squibb, Curis, Eli Lilly, Ferring, GlaxoSmithKline, Janssen, Novostia, Pfizer, Portola, Theravance and Veralox. M.C.B. has received consulting fees or honoraria from Anthos, Bristol Myers Squibb, CSL Behring, Johnson & Johnson, MSD and Pfizer. J.W.E. has received payments and/or grant support from Anthos, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Daiichi Sankyo, Janssen, Johnson & Johnson, Merck, Pfizer, PhaseBio and Servier. C.M.G. has received grants from CSL Behring, Janssen, Johnson & Johnson and SCAD Alliance; has received personal fees from AMAG Pharmaceuticals, Amarin, Angel Medical Corporation, Anthos Therapeutics, AstraZeneca (during the conduct of the study), Bayer, Bioclinica, Boston Clinical Research Institute, Boston Scientific, Bristol Myers Squibb, Caladrius Biosciences, Cardiovascular Clinical Science Foundation, Cardiovascular Research Foundation, CeleCor Therapeutics, CSL Behring, CytoSorbents, Duke Clinical Research Institute, Dyad Medical, Eidos Therapeutics, EXCITE International ($0 received), Gilead Sciences, Inari, Janssen, Johnson & Johnson, MD Magazine, MedImmune, Medtelligence, MedTrace, Merck, Microdrop, Microport, Novo Nordisk, Paratek, PERT Consortium, Pfizer, PhaseBio, PHRI, PLx Pharma, Revance Therapeutics, SmartMedics, Society for Cardiovascular Angiography and Interventions, Somahlution, UpToDate in Cardiovascular Medicine and WebMD; has received non-financial support from the Baim Institute; and has equity from Absolutys and Dyad Medical. S.G.G. has received research grant support (for work on steering committees or data and safety monitoring committees) and/or speaker or consulting honoraria (such as advisory boards) from Alnylam, Amgen, Anthos Therapeutics, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, CSL Behring, CYTE, Daiichi-Sankyo/American Regent, Eli Lilly, Esperion, Ferring Pharmaceuticals, HLS Therapeutics, Idorsia, JAMP Pharma, Merck, Novartis, Novo Nordisk, Pendopharm/Pharmascience, Pfizer, Regeneron, Roche, Sanofi, Servier, Tolmar Pharmaceuticals and Valeo Pharma; and salary support/honoraria from the Canadian Heart Failure Society, Canadian Heart Research Centre and MD Primer, Canadian VIGOUR Centre, Cleveland Clinic Coordinating Centre for Clinical Research, Duke Clinical Research Institute, Jewish General Hospital/CIUSSS Centre-Ouest-de-l’Ile-de-Montreal, New York University Clinical Coordinating Centre, PERFUSE Research Institute, Peter Munk Cardiac Centre Clinical Trials and Translation Unit, Ted Rogers Centre for Heart Research and TIMI Study Group (Brigham Health). R.D.L. reports research grants or contracts from Amgen, Bristol-Myers Squibb, GlaxoSmithKline, Medtronic, Pfizer, Sanofi-Aventis; funding for educational activities or lectures from Daiichi Sankyo, Novo Nordisk and Pfizer; and funding for consulting or other services from Bayer, Boehringer Ingelheim, Bristol-Myers Squibb and Novo Nordisk. M.R.P. reports research grants with Bayer, Idorsia, Medtronic, NHBLI and Novartis, and has received honoraria for scientific advisory boards and consulting from Bayer, Esperion and Novartis. J.P.P. is supported by a grant (R01AG074185) from the National Institutes of Aging; receives grants for clinical research from Abbott, American Heart Association, Boston Scientific, iRhythm and Philips; and serves as a consultant to Abbott, Boston Scientific, Kardium, Medtronic, Milestone Pharmaceuticals, Philips, Sanofi and Up-to-Date. S.V.R. reports research grants from American College of Cardiology and NHLBI. C.T.R. reports research grants through Brigham and Women’s Hospital from Anthos, AstraZeneca, Daiichi Sankyo, Janssen and Novartis; has received honoraria for scientific advisory boards and consulting from Anthos, Bayer, Bristol Myers Squibb, Daiichi Sankyo, Janssen and Pfizer; and is a member of the TIMI Study Group, which has received institutional research grant support through Brigham and Women’s Hospital from Abbott, Abiomed, Amgen, Anthos Therapeutics, ARCA Biopharma, AstraZeneca, Boehringer Ingelheim, Daiichi-Sankyo, Ionis Pharmaceuticals, Janssen Research and Development, LLC, MedImmune, Merck, Novartis, Pfizer, Regeneron Pharmaceuticals, Roche, Saghmos Therapeutics, Siemens Healthcare Diagnostics, Softcell Medical, The Medicines Company, Verve Therapeutics and Zora Biosciences. P.G.S. has received research grants from Bayer; is a consultant for Amarin, Amgen, AstraZeneca, Bayer, BMS, Boehringer Ingelheim, Janssen, Novartis, Novo Nordisk, Pfizer, PhaseBio, Regeneron, Sanofi and Servier; and is Chief Medical Officer for Bioquantis. D.J.A. has received consulting fees or honoraria from Abbott, Amgen, AstraZeneca, Bayer, Biosensors, Boehringer Ingelheim, Bristol Myers Squibb, Chiesi, CSL Behring, Daiichi-Sankyo, Eli Lilly, Faraday, Haemonetics, Janssen, Merck, Novartis, Novo Nordisk, PhaseBio, PLx Pharma, Pfizer and Sanofi; and his institution has received research grants from Amgen, AstraZeneca, Bayer, Biosensors, CeloNova, CSL Behring, Daiichi-Sankyo, Eisai, Eli Lilly, Faraday, Gilead, Idorsia, Janssen, Matsutani Chemical Industry Company, Merck, Novartis and the Scott R. MacKenzie Foundation. The other authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Cadiology thanks Jose A. Paramo, Hugo ten Cate and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
Reprints and permissions
About this article
Check for updates. Verify currency and authenticity via CrossMark
Cite this article
Capodanno, D., Alexander, J.H., Bahit, M.C. et al. Factor XI inhibitors for the prevention and treatment of venous and arterial thromboembolism. Nat Rev Cardiol (2025). https://doi.org/10.1038/s41569-025-01144-z
Download citation
Accepted:06 March 2025
Published:31 March 2025
DOI:https://doi.org/10.1038/s41569-025-01144-z
Share this article
Anyone you share the following link with will be able to read this content:
Get shareable link
Sorry, a shareable link is not currently available for this article.
Copy to clipboard
Provided by the Springer Nature SharedIt content-sharing initiative