Skip to main content

Advertisement

ADVERTISEMENT

EP Review

Factor XIa Inhibitors: The Quest for the Perfect Antithrombin Agents

Linda Moulton, RN, MS, 

Faculty: Order and Disorder EP Training Program, Critical Care ED/CCE Consulting, Calistoga, California

July 2023
© 2023 HMP Global. All Rights Reserved.
Any views and opinions expressed are those of the author(s) and/or participants and do not necessarily reflect the views, policy, or position of EP Lab Digest or HMP Global, their employees, and affiliates. 

EP LAB DIGEST. 2023;23(7):1,5-7.

There have been major advancements in the quest to make safer anticoagulant agents for the treatment of clots in atrial fibrillation (AF) and coronary artery disease (CAD). Direct oral anticoagulants (DOACs) have replaced vitamin K antagonists (VKAs) in many cases; DOACs have a safer bleeding profile than VKAs, as proven in multiple trials. However, there are still unacceptable levels of bleeding with DOAC use, leading to prescribing hesitancy and underuse.1 There are also issues with using DOACs with medical devices and mechanical valves. Thus, the search continues for safer anticoagulants.

Thrombosis can occur with CAD, AF, stroke, peripheral arterial disease (PAD), and venous thromboembolism (VTE). Heart disease and stroke are responsible for 25% of deaths worldwide.2 Currently, we have direct inhibitors of factor Xa (FXa) of the coagulation cascade, with rivaroxaban, apixaban, endoxaban, and betrixaban.

In addition, dabigatran, hirudins, and argatroban are direct inhibitors of thrombin. These drugs target thrombin within the common pathway and via FXa. A new suggested approach to anticoagulation is targeting factor XI (FXI) in the intrinsic pathway. To understand the difference between current therapies and the new suggested approach, one needs to review the difference between thrombosis formation and hemostasis, and the organization of the coagulation cascade.

Coagulation Cascade Review and FXI

The coagulation cascade is usually labeled and explained by discussing the 3 sections: the intrinsic or contact pathway, the extrinsic or tissue factor pathway, and the common pathway. The intrinsic or contact pathway is activated by factors in the bloodstream; endothelial injury leads to eventual thrombosis formation down in the common pathway.

The extrinsic pathway or tissue factor pathway is activated when injury occurs outside the circulation.  Tissue thromboplastin is released, the common pathway in engaged, and a clot is formed. The common pathway is where both intrinsic and extrinsic pathways lead. Prothrombin leads to thrombin, fibrinogen to fibrin, and fibrin to clot.

Thrombosis is an intravascular process. Plaque triggers “pathologic blood clotting” via platelet activation and coagulation. FXI is triggered by tissue factor (TF) or via the intrinsic pathway. The end result is clot generation. Clots form in the blood vessel lumen and thrombosis compromises blood flow. Thrombosis can also be initiated by contact of blood with valves, extracorporeal circuits, or any implanted devices. An additional factor of note is that thrombin feedback to FXI causes an expansion of the thrombin effect by repeating the cycle (Figure).

Moulton Factor XIa Inhibitors Figure 1
Figure. The coagulation cascade. Notice the feedback of thrombin to FXI, enhancing the thrombin effect. With FXI inhibition, the thrombin effect is lessened.

The goal of hemostasis is to seal the leak and stop the bleeding, which minimizes blood loss. Hemostasis is an extravascular process; tissue factor from surrounding vessels initiates this. Vascular injury leads to TF, triggering major thrombin generation and back activation of FXI by thrombin. This latter activity is not necessarily needed for hemostasis. Production of thrombin and fibrin are the end steps in both hemostasis and thrombosis.

FXI is produced by the liver and is a part of the contact or intrinsic pathway. FXI can be activated by FXIIa or by feedback from thrombin in the common pathway. An increase in thrombin depends on activation of FXI. Therefore, FXI may actually be more important for thrombosis formation and is possibly not needed for hemostasis at all.3,4

Patients with FXI deficiency do not seem to have spontaneous bleeding and these patients have a lower stroke risk; they have little or no bleeding tendency. They will have some bleeding after trauma to certain tissues: the oropharynx and urinary tract. However, the central nervous system, gastrointestinal system, joints, and muscles are not affected. On the other hand, patients with elevated FXI levels have a higher stroke risk as well as greater risk for venous or arterial thrombosis.5-7 So, as seen in deficiency, FXI inhibition should not pose the bleeding risk of DOACs.

A major benefit of FXI inhibition might be seen in patients with implantable devices and valves. With mechanical devices, coagulation begins in the contact or intrinsic pathway with activation of FXII and generation of tissue factor (Figure). The next step is activation of FXI to FXIa. The inhibition of FXI can stop the coagulation process and avert thrombosis formation downstream.8 Dabigatran, which has been contraindicated for use with valve patients, inhibits some thrombin creation, but fails to prevent device thrombosis. Other DOACs (apixaban, rivaroxaban, etc) inhibit FXa in the common pathway where thrombin is forming, but are associated with a higher bleeding risk. So, use of an FXI inhibitor that inhibits thrombin from upstream may be safer for these patients than dabigatran and other DOACs.9

Inhibitors of FXI Currently Under Investigation

The majority of research in FXI inhibition has been evaluating 4 types of potential inhibitors, each with a slightly different mode of action (Table). None of these are on the market or approved at this time by the US Food and Drug Administration.5,9-15

Moulton Factor XIa Inhibitors Table 1
Table. Inhibitors of FXI currently under investigation.

The first type is the second-generation antisense oligonucleotides. These work to reduce the hepatic syntheses of FXI. Most of the research has been with fesomersen (Ionis Pharmaceuticals), which can be subcutaneously administered. Fesomersen requires a few weeks to lower FXI levels into therapeutic range, has a long half-life, and could be used once a month.

There are monoclonal antibodies that inhibit or block FXI activation, FXIa activity, or both. The most researched of these are osocimab and abelacimab. Initial work has found them to be safe and well tolerated. The mode of administration is subcutaneous (sq) or intravenous (IV), with rapid onset of action for the IV mode. Drug half-life is long; thus, monthly maintenance via subcutaneous injection would be possible.16

Aptamers are short, single-stranded oligonucleotides that bind over large surface areas on a targeted protein, in this case, targeting FXI or FXIa, inhibiting activity. Thus far, there has been only early-stage testing of aptamers. However, their administration would be IV, with rapid onset of action and a short half-life, requiring daily administration. This could be advantageous for use in the critical care setting.5,17

Lastly, there are the small molecule FXIa inhibitors. The development of this category is entering the phase 3 trial realm. The small molecule agents target FXI active sites or hepatic allosteric sites on FXIa. Asundexian and milvexian have been found to be safe and well tolerated. Milvexian has been tested in patients with normal hepatic function and mild or moderate hepatic impairment.15,18-19 Coagulation studies during treatment have shown milvexian to increase activated partial thromboplastin time (APTT) without changing prothrombin time.20 Asundexian and milvexian have been administered intravenously and orally, have rapid onset of action (maximum plasma level at 2-4 hours), and a half-life between 12 and 17 hours, making them a once- or twice-daily possibility.12,19,21

The different administration profiles seen with the drug categories could fit with varying treatment goals. For the critically ill patient or those undergoing interventional procedures, aptamers may be best due to the IV administration and rapid onset and offset of effect. A weekly administered drug may work for hemodialysis patients. Monthly administration could work with cancer-associated thromboses during a monthly oncology visit. Short half-life IV or oral administration could work with peri-interventional needs. Another benefit may be the possibility of once- or twice-a-month injections, a feature that may increase drug compliance for some patients.13

Clinical Trials

Studies have been conducted on all the categories of inhibitors, some studies on a basic laboratory level and others as phase 1 and 2 studies. These have included prevention studies, safety studies, comparison studies, and dose-finding studies. The phase 2 trials initially began with use of the agents in orthopedic surgery patients, with hip and knee surgeries. This group has historically been the starting point for testing the efficacy of anticoagulant therapies, as deep vein thrombosis of the leg can easily be detected with venography. Such study results have shown reduced thrombosis risk, decreased bleeding events, and decreased risk of VTE.22 In other studies, fesomersen, osocimab, abelacimab, and milvexian have been compared with enoxaparin. Fesomersen and osocimab have also been studied vs placebo with end-stage kidney disease and hemodialysis patients.5,9,15

The most likely agents to be used in atrial fibrillation (AF) patients at risk for stroke are the small molecule FXIa inhibitors asundexian and milvexian, which can be administered orally. Clinical trials with asundexian include the PACIFIC-AF,23 PACIFIC-STROKE,24 and PACIFIC-AMI studies.12

The PACIFIC-AF study compared asundexian to apixaban in an AF population. The reduction in bleeding was similar but slightly favored asundexian. The PACIFIC-STROKE study enrolled patients with recent non-cardioembolic ischemic stroke and compared asundexian to placebo. The subjects were kept on antiplatelet therapy in both groups. Asundexian did not reduce the stroke rate and did not increase bleeding complications. The PACIFIC-AMI study was a dose-finding safety study for prevention of myocardial infarction (MI). Recent MI patients on dual antiplatelet therapy were randomized to placebo or 10, 20, or 50 mg of asundexian. A low rate of ischemic events was reported and no significant increase in bleeding was seen.

AXIOMATIC-SSP is a phase 2 trial comparing various milvexian doses to placebo. The study objective is to prevent new ischemic stroke in patients after stroke or TIA, when milvexian is added to antiplatelet drugs (clopidogrel and aspirin).25    

New phase 3 trials with asundexian are beginning to enroll patients.9 OCEANIC-AF is enrolling patients with AF and randomizing them to apixaban or asundexian with the goal of preventing stroke or systemic embolism and comparing safety. The study began in December and has an enrollment goal of 18,000 participants with a 9- to 33-month follow-up.26 The second study, called OCEANIC-STROKE, is recruiting patients with a history of stroke and randomizing them to placebo or asundexian, while keeping both groups on dual antiplatelet therapy. Data will be collected on bleeding rates and whether there is a reduction in ischemic stroke or temporary stroke-like symptoms with asundexian. This study began in January 2023 with plans to enroll 9300 participants with a 3- to 31-month follow-up.27  

Reversal Agents

In the past, as new anticoagulants have been developed, there has been the need to develop or identify possible approaches for reversing their anticoagulant effect in the setting of trauma or during medical procedures where excessive bleeding is a risk. There is some work being done in this area. However, there is an established treatment regimen that has been used for patients with FXI deficiency in such situations. This involves treatment with antifibrinolytic agents and low doses of recombinant FVIIa.28 Thus, the introduction of FXI inhibitors into the clinical setting could well be safer than prior new drug introductions due to an already known reversal regimen to use in an emergency.

Future Directions

The inhibition of FXIa appears to be a promising approach for anticoagulation due to the reduced bleeding complications seen with their use. Inhibitors may be hemostasis-sparing drugs with better safety profiles. Also, we may soon be able to treat a wider range of patients with a high risk of bleeding. The beginning of these phase 3 trials brings us ever closer to the reality of seeing these agents used in the clinical setting. 

Disclosure: The author has completed and returned the ICMJE Form for Disclosure of Potential Conflicts of Interest. She reports no conflicts of interest regarding the content herein.

References

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

2. Lozano R, Naghavi M, Foreman K, 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. 2012;380(9859):2095-2128. doi:10.1016/S0140-6736(12)61728-0

3. Puy C, Rigg RA, McCarty OJT. The hemostatic role of factor XI. Thromb Res. 2016;141(Suppl 2):S8-S11. doi:10.1016/S0049-3848(16)30354-1

4. Emsley J, McEwan PA, Gailani D. Structure and function of factor XI. Blood. 2010;115(13):2569-2577. doi:10.1182/blood-2009-09-199182

5. Badimon JJ, Escolar G, Zafar MU. Factor XI/XIa inhibition: the arsenal in development for a new therapeutic target in cardio- and cerebrovascular disease. J Cardiovasc Dev Dis. 2022;9(12):437. doi:10.3390/jcdd9120437

6. Key NS. Epidemiologic and clinical data linking factors XI and XII to thrombosis. Hematology Am Soc Hematol Educ Program. 2014;2014(1):66-70. doi:10.1182/asheducation-2014.1.66

7. Gailani D. Making anticoagulation safer. Lancet. 2022;399(10333):1360-1361. doi:10.1016/S0140-6736(22)00563-3

8. Jaffer IH, Stafford AR, Fredendurgh JC, et al. Dabigatran is less effective than warfarin in attenuating mechanical heart valve-induced thrombin generation. J Am Heart Assoc. 2015;4(8):e002322. doi:10.1161/JAHA.115.002322

9. Chan NC, Weitz JI. Antithrombotic agents: new directions in antithrombotic therapy. Circ Res. 2019;124(3):426-436. doi:10.1161/CIRCRESAHA.118.313155

10. Salomon O, Gailani D. A proposal for managing bleeding in patients on therapeutic factor XI(a) inhibitors. J Thromb Haemost. 2022;20(1):32-38. doi:10.1111/jth.15579

11. Weitz JI, Eikelboom JW. What is the future of factor XI inhibitors? Circulation. 2022;146(25):1899-1902. doi:10.1161/CIRCULATIONAHA.122.061132

12. Rao SV, Kirsch B, Bhatt DL, 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 following acute myocardial infarction. Circulation. 2022;146(16):1196-1206. doi:10.1161/CIRCULATIONAHA.122.061612

13. Nopp S, Kraemmer D, Ay C. Factor XI inhibitors for prevention and treatment of venous thromboembolism: a review on the rationale and update on current evidence. Front Cardiovasc Med. 2022;9:903029. doi:10.3389/fcvm.2022.903029

14. Perera V, Wang Z, Luettgen J, et al. First-in-human study of milvexian, an oral, direct, small molecule factor XIa inhibitor. Clin Transl Sci. 2022;15(2):330-342. doi:10.1111/cts.13148

15. Perera V, Abelian G, Li D, et al. Single-dose pharmacokinetics of milvexian in participants with mild or moderate hepatic impairment compared with healthy participants. Clin Pharmacokinet. 2022;61(6):857-867. doi:10.1007/s40262-022-01110-9

16. Yi BA, Freedholm D, Widener N, et al. Pharmacokinetics and pharmacodynamics of abelacimab (MAA868), a novel dual inhibitor of factor XI and factor XIa. J Thromb Haemost. 2022;20(2):307-315. doi:10.1111/jth.15577

17. Woodruff RS, Ivanov I, Verhamme IM, Sun MF, Gailani D, Sullenger BA. Generation and characterization of aptamers targeting factor XIa. Thromb Res. 2017;156:134-141. doi:10.1016/j.thromres.2017.06.015

18. Perera V, Abelian G, Li D, et al. Single-dose pharmacokinetics of milvexian in participants with normal renal function and participants with moderate or severe renal impairment. Clin Pharmacokinet. 2022;61(10):1405-1416. doi:10.1007/s40262-022-01150-1

19. Weitz J, Strong J, Ageno W, et al. Milvexian for the prevention of venous thromboembolism. N Engl J Med. 2021;385(23):2161-2172. doi:10.1056/NEJMoa2113194

20. Wong PC, Crain EJ, Bozarth JM, 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. 2022;20(2):399-408. doi:10.1111/jth.15588

21. Heitmeier S, Visser M, Tersteegen A, et al. Pharmacological profile of asundexian, a novel, orally bioavailable inhibitor of factor XIa. J Thromb Haemost. 2022;20(6):1400-1411. doi:10.1111/jth.15700

22. Presume J, Ferreira J, Ribeiras R, et al. Achieving higher efficiency without compromising safety with factor XI inhibitors versus low molecular weight heparin for the prevention of venous thromboembolism in major orthopedic surgery-systematic review and meta-analysis. J Thromb Haemost. 2022;20(12):2930-2938. doi:10.1111/jth.15890

23. Piccini JP, Caso V, Connolly SJ, et al, PACIFIC-AF investigators. Safety of the oral factor XIa inhibitor asundexian compared with apixaban in patients with atrial fibrillation (PACIFIC-AF): a multicenter, randomized, double-blind, double-dummy, dose-finding phase 2 study. Lancet. 2022;399(10333):1383-1390. doi:10.1016/SO140-6736(22)00456-1

24. Shoamanesh A, Mundi H, Smith EE, et al. Factor XIa inhibition with asundexian after acute non-cardiometabolic ischemic stroke (PACIFIC-Stroke): an international, randomized, double-blind, placebo-controlled, phase 2b trial. Lancet. 2022;400(10357):997-1007. doi:10.1016/SO140-6736(22)01588-4

25. Sharma M, Molina CA, Toyoda K, et al. Rationale and design of the AXIOMATIC-SSP phase II trial: Antithrombotic treatment with factor XIa inhibition to optimize management of acute thromboembolic events for secondary stroke prevention. J Stroke Cerebrovasc Dis. 2022;31(10):106742. doi:10.1016/j.jstrokecerebrovasdis.2022.106742

26. A study to learn how well the study treatment asundexian works and how safe it is compared to apixaban to prevent stroke or systemic embolism in people with irregular and often rapid heartbeat (atrial fibrillation), and at risk for stroke (OCEANIC-AF). ClinicalTrials.gov. Accessed March 10, 2023. https://clinicaltrials.gov/ct2/show/NCT05643573

27. A study to learn more about asundexian (also called BAY2433334) for prevention of ischemic stroke in male and female participants aged 18 years and older who already had such a stroke due to a blood clot that formed outside the heart and traveled to the brain, or temporary stroke-like symptoms (OCEANIC-STROKE). ClinicalTrials.gov. Accessed March 10, 2023. https://clinicaltrials.gov/ct2/show/NCT05686070

28. Gailani D, Bane CE, Gruber A. Factor XI and contact activation as targets for antithrombotic therapy. J Thromb Haemost. 2015;13(8):1383-1395. doi:10.1111/jth.13005


Advertisement

Advertisement

Advertisement