Semin Thromb Hemost
DOI: 10.1055/s-0043-57011
Review Article

Targeting the Contact Pathway of Coagulation for the Prevention and Management of Medical Device-Associated Thrombosis

Abhishek Goel
1   Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
,
Harsha Tathireddy
1   Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
,
Si-Han Wang
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
,
Helen H. Vu
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
,
Cristina Puy
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
,
Monica T. Hinds
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
,
David Zonies
3   Department of Surgery, Oregon Health and Science University, Portland, Oregon
,
Owen J.T. McCarty
1   Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
,
Joseph J. Shatzel
1   Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
2   Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
› Author Affiliations
Funding This work was supported by the National Institutes of Health (R01HL151367 to J.J.S, R01HL101972 and R01HL144113 to O.J.T.M).

Abstract

Hemorrhage remains a major complication of anticoagulants, with bleeding leading to serious and even life-threatening outcomes in rare settings. Currently available anticoagulants target either multiple coagulation factors or specifically coagulation factor (F) Xa or thrombin; however, inhibiting these pathways universally impairs hemostasis. Bleeding complications are especially salient in the medically complex population who benefit from medical devices. Extracorporeal devices—such as extracorporeal membrane oxygenation, hemodialysis, and cardiac bypass—require anticoagulation for optimal use. Nonetheless, bleeding complications are common, and with certain devices, highly morbid. Likewise, pharmacologic prophylaxis to prevent thrombosis is not commonly used with many medical devices like central venous catheters due to high rates of bleeding. The contact pathway members FXI, FXII, and prekallikrein serve as a nexus, connecting biomaterial surface-mediated thrombin generation and inflammation, and may represent safe, druggable targets to improve medical device hemocompatibility and thrombogenicity. Recent in vivo and clinical data suggest that selectively targeting the contact pathway of coagulation through the inhibition of FXI and FXII can reduce the incidence of medical device-associated thrombotic events, and potentially systemic inflammation, without impairing hemostasis. In the following review, we will outline the current in vivo and clinical data encompassing the mechanism of action of drugs targeting the contact pathway. This new class of inhibitors has the potential to herald a new era of effective and low-risk anticoagulation for the management of patients requiring the use of medical devices.



Publication History

Article published online:
12 April 2023

© 2023. Thieme. All rights reserved.

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Revenko AS, Gao D, Crosby JR. et al. Selective depletion of plasma prekallikrein or coagulation factor XII inhibits thrombosis in mice without increased risk of bleeding. Blood 2011; 118 (19) 5302-5311
  • 2 DeLoughery EP, Olson SR, Puy C, McCarty OJT, Shatzel JJ. The safety and efficacy of novel agents targeting factors XI and XII in early phase human trials. Semin Thromb Hemost 2019; 45 (05) 502-508
  • 3 Eikelboom JW, Connolly SJ, Brueckmann M. et al; RE-ALIGN Investigators. Dabigatran versus warfarin in patients with mechanical heart valves. N Engl J Med 2013; 369 (13) 1206-1214
  • 4 Kato C, Oakes M, Kim M. et al. Anticoagulation strategies in extracorporeal circulatory devices in adult populations. Eur J Haematol 2021; 106 (01) 19-31
  • 5 Jawitz OK, Wang TY, Lopes RD. et al. Rationale and design of PROACT Xa: a randomized, multicenter, open-label, clinical trial to evaluate the efficacy and safety of apixaban versus warfarin in patients with a mechanical On-X Aortic Heart Valve. Am Heart J 2020; 227: 91-99
  • 6 Andreas M, Moayedifar R, Wieselthaler G. et al. Increased thromboembolic events with dabigatran compared with vitamin K antagonism in left ventricular assist device patients: a randomized controlled pilot trial. Circ Heart Fail 2017; 10 (05) e003709
  • 7 Lorentz CU, Verbout NG, Wallisch M. 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 2019; 39 (04) 799-809
  • 8 Puy C, Rigg RA, McCarty OJ. The hemostatic role of factor XI. Thromb Res 2016; 141 (Suppl. 02) S8-S11
  • 9 Raghunathan V, Zilberman-Rudenko J, Olson SR, Lupu F, McCarty OJT, Shatzel JJ. The contact pathway and sepsis. Res Pract Thromb Haemost 2019; 3 (03) 331-339
  • 10 Von dem Borne PA, Bajzar L, Meijers JC, Nesheim ME, Bouma BN. Thrombin-mediated activation of factor XI results in a thrombin-activatable fibrinolysis inhibitor-dependent inhibition of fibrinolysis. J Clin Invest 1997; 99 (10) 2323-2327
  • 11 Merlo C, Wuillemin WA, Redondo M. et al. Elevated levels of plasma prekallikrein, high molecular weight kininogen and factor XI in coronary heart disease. Atherosclerosis 2002; 161 (02) 261-267
  • 12 Adenaeuer A, Barco S, Trinchero A, Lackner KJ, Lämmle B, Rossmann H. Definite diagnosis of plasma prekallikrein deficiency should not be based exclusively on shortening of the aPTT upon prolonged pre-incubation. Int J Lab Hematol 2022; 44 (04) e179-e180
  • 13 Abraham R, Viswanathan GK, Dass J. et al. Prekallikrein deficiency: challenges in laboratory testing. Int J Lab Hematol 2022; 44 (04) e185-e186
  • 14 Deroux A, Boccon-Gibod I, Fain O. et al. Hereditary angioedema with normal C1 inhibitor and factor XII mutation: a series of 57 patients from the French National Center of Reference for Angioedema. Clin Exp Immunol 2016; 185 (03) 332-337
  • 15 Labarrere CA, Dabiri AE, Kassab GS. Thrombogenic and inflammatory reactions to biomaterials in medical devices. Front Bioeng Biotechnol 2020; 8: 123
  • 16 Annich GM. Extracorporeal life support: the precarious balance of hemostasis. J Thromb Haemost 2015; 13 (Suppl. 01) S336-S342
  • 17 Wong TE, Nguyen T, Shah SS, Brogan TV, Witmer CM. Antithrombin concentrate use in pediatric extracorporeal membrane oxygenation: a multicenter cohort study. Pediatr Crit Care Med 2016; 17 (12) 1170-1178
  • 18 Jaffer IH, Fredenburgh JC, Hirsh J, Weitz JI. Medical device-induced thrombosis: what causes it and how can we prevent it?. J Thromb Haemost 2015; 13 (Suppl. 01) S72-S81
  • 19 Byrnes JR, Wolberg AS. Red blood cells in thrombosis. Blood 2017; 130 (16) 1795-1799
  • 20 Palta S, Saroa R, Palta A. Overview of the coagulation system. Indian J Anaesth 2014; 58 (05) 515-523
  • 21 Aleman MM, Walton BL, Byrnes JR, Wolberg AS. Fibrinogen and red blood cells in venous thrombosis. Thromb Res 2014; 133 (0 1, Suppl 1): S38-S40
  • 22 Jaffer IH, Weitz JI. The blood compatibility challenge. Part 1: Blood-contacting medical devices: the scope of the problem. Acta Biomater 2019; 94: 2-10
  • 23 Vogler EA, Siedlecki CA. Contact activation of blood-plasma coagulation. Biomaterials 2009; 30 (10) 1857-1869
  • 24 Badv M, Bayat F, Weitz JI, Didar TF. Single and multi-functional coating strategies for enhancing the biocompatibility and tissue integration of blood-contacting medical implants. Biomaterials 2020; 258: 120291
  • 25 Dal Sasso E, Bagno A, Scuri STG, Gerosa G, Iop L. The biocompatibility challenges in the total artificial heart evolution. Annu Rev Biomed Eng 2019; 21: 85-110
  • 26 Manivasagam VK, Sabino RM, Kantam P, Popat K. Surface modification strategies to improve titanium hemocompatibility: a comprehensive review. Mater Adv 2021; 2 (18) 5824-5842
  • 27 Spijker HT, Graaff R, Boonstra PW, Busscher HJ, van Oeveren W. On the influence of flow conditions and wettability on blood material interactions. Biomaterials 2003; 24 (26) 4717-4727
  • 28 Douglass M, Garren M, Devine R, Mondal A, Handa H. Bio-inspired hemocompatible surface modifications for biomedical applications. Prog Mater Sci 2022; 130: 100997
  • 29 Fisher C, Shao H, Ho CH. Improved hemocompatibility of polysulfone hemodialyzers with Endexo® surface modifying molecules. J Biomed Mater Res B Appl Biomater 2022; 110 (06) 1335-1343
  • 30 Fallon ME, Le HH, Bates NM. et al. Hemocompatibility of micropatterned biomaterial surfaces is dependent on topographical feature size. Front Physiol 2022; 13: 983187
  • 31 Ngo BKD, Grunlan MA. Protein resistant polymeric biomaterials. ACS Macro Lett 2017; 6 (09) 992-1000
  • 32 Gifford R, Kehoe JJ, Barnes SL, Kornilayev BA, Alterman MA, Wilson GS. Protein interactions with subcutaneously implanted biosensors. Biomaterials 2006; 27 (12) 2587-2598
  • 33 Wolberg AS, Campbell RA. Thrombin generation, fibrin clot formation and hemostasis. Transfus Apheresis Sci 2008; 38 (01) 15-23
  • 34 Klopfleisch R, Jung F. The pathology of the foreign body reaction against biomaterials. J Biomed Mater Res A 2017; 105 (03) 927-940
  • 35 Richenbacher WE. Complications of mechanical ventricular assistance. In: Mechanical Circulatory Support. CRC Press; 2020: 183-198
  • 36 Bazzan M, Vaccarino A, Marletto F. Systemic lupus erythematosus and thrombosis. Thromb J 2015; 13 (01) 16
  • 37 Hong JK, Gao L, Singh J. et al. Evaluating medical device and material thrombosis under flow: current and emerging technologies. Biomater Sci 2020; 8 (21) 5824-5845
  • 38 Kohs TCL, Lorentz CU, Johnson J. et al. Development of coagulation factor XII antibodies for inhibiting vascular device-related thrombosis. Cell Mol Bioeng 2020; 14 (02) 161-175
  • 39 Wu Y. Contact pathway of coagulation and inflammation. Thromb J 2015; 13: 17
  • 40 Grover SP, Mackman N. Intrinsic pathway of coagulation and thrombosis: insights from animal models. Arterioscler Thromb Vasc Biol 2019; 39 (03) 331-338
  • 41 de Maat S, Maas C. Factor XII: form determines function. J Thromb Haemost 2016; 14 (08) 1498-1506
  • 42 Shamanaev A, Ivanov I, Sun M-F. et al. Model for surface-dependent factor XII activation: the roles of factor XII heavy chain domains. Blood Adv 2022; 6 (10) 3142-3154
  • 43 Wu W, Roder H, Walsh PN. Conformational changes facilitate FXI autoactivation to FXIa. Blood 2010; 116 (21) 19
  • 44 Shamanaev A, Litvak M, Gailani D. Recent advances in factor XII structure and function. Curr Opin Hematol 2022; 29 (05) 233-243
  • 45 Naudin C, Burillo E, Blankenberg S, Butler L, Renné T. Factor XII contact activation. Semin Thromb Hemost 2017; 43 (08) 814-826
  • 46 Ivanov I, Matafonov A, Gailani D. Single-chain factor XII: a new form of activated factor XII. Curr Opin Hematol 2017; 24 (05) 411-418
  • 47 Tillman BF, Gruber A, McCarty OJT, Gailani D. Plasma contact factors as therapeutic targets. Blood Rev 2018; 32 (06) 433-448
  • 48 Kleinschnitz C, Stoll G, Bendszus M. et al. Targeting coagulation factor XII provides protection from pathological thrombosis in cerebral ischemia without interfering with hemostasis. J Exp Med 2006; 203 (03) 513-518
  • 49 Renné T, Stavrou EX. Roles of factor XII in Innate Immunity. Front Immunol 2019; 10: 2011
  • 50 Barbieri CM, Wang X, Wu W. et al. Factor XIIa as a novel target for thrombosis: target engagement requirement and efficacy in a rabbit model of microembolic signals. J Pharmacol Exp Ther 2017; 360 (03) 466-475
  • 51 Kenne E, Nickel KF, Long AT. et al. Factor XII: a novel target for safe prevention of thrombosis and inflammation. J Intern Med 2015; 278 (06) 571-585
  • 52 Larsson M, Rayzman V, Nolte MW. et al. A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk. Sci Transl Med 2014; 6 (222) 222ra17
  • 53 Wallisch M, Lorentz CU, Lakshmanan HHS. et al. Antibody inhibition of contact factor XII reduces platelet deposition in a model of extracorporeal membrane oxygenator perfusion in nonhuman primates. Res Pract Thromb Haemost 2020; 4 (02) 205-216
  • 54 Wilbs J, Kong XD, Middendorp SJ. et al. Cyclic peptide FXII inhibitor provides safe anticoagulation in a thrombosis model and in artificial lungs. Nat Commun 2020; 11 (01) 3890
  • 55 Matafonov A, Leung PY, Gailani AE. et al. Factor XII inhibition reduces thrombus formation in a primate thrombosis model. Blood 2014; 123 (11) 1739-1746
  • 56 Pollack Jr CV, Kurz MA, Hayward NJ. EP-7041, a factor XIa inhibitor as a potential antithrombotic strategy in extracorporeal membrane oxygenation: a brief report. Crit Care Explor 2020; 2 (09) e0196
  • 57 Schumacher WA, Seiler SE, Steinbacher TE. et al. Antithrombotic and hemostatic effects of a small molecule factor XIa inhibitor in rats. Eur J Pharmacol 2007; 570 (1-3): 167-174
  • 58 Cheng Q, Tucker EI, Pine MS. et al. A role for factor XIIa-mediated factor XI activation in thrombus formation in vivo. Blood 2010; 116 (19) 3981-3989
  • 59 Gruber A, Hanson SR. Factor XI-dependence of surface- and tissue factor-initiated thrombus propagation in primates. Blood 2003; 102 (03) 953-955
  • 60 Schultze AE, Walker DB, Turk JR, Tarrant JM, Brooks MB, Pettit SD. Current practices in preclinical drug development: gaps in hemostasis testing to assess risk of thromboembolic injury. Toxicol Pathol 2013; 41 (03) 445-453
  • 61 Fredenburgh JC, Weitz JI. Factor XI as a target for new anticoagulants. Hamostaseologie 2021; 41 (02) 104-110
  • 62 Büller HR, Bethune C, Bhanot S. et al; FXI-ASO TKA Investigators. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N Engl J Med 2015; 372 (03) 232-240
  • 63 Lippi G, Harenberg J, Mattiuzzi C, Favaloro EJ. Next generation antithrombotic therapy: focus on antisense therapy against coagulation factor XI. Semin Thromb Hemost 2015; 41 (02) 255-262
  • 64 Geary RS, Norris D, Yu R, Bennett CF. Pharmacokinetics, biodistribution and cell uptake of antisense oligonucleotides. Adv Drug Deliv Rev 2015; 87: 46-51
  • 65 Salomon O, Gailani D. A proposal for managing bleeding in patients on therapeutic factor XI(a) inhibitors. J Thromb Haemost 2022; 20 (01) 32-38
  • 66 Wang X, Li Q, Du F. et al. Reversal of antithrombotic effects of FXIa inhibitor milvexian (BMS-986177/JNJ-70033093) by non-specific agents in a rabbit AV-shunt model of thrombosis. In: Proceedings of the ISTH 2021 Congress, Philadelphia, PA; 2021:17–21
  • 67 Fischer PM. Design of small-molecule active-site inhibitors of the S1A family proteases as procoagulant and anticoagulant drugs. J Med Chem 2018; 61 (09) 3799-3822
  • 68 Walsh M, Bethune C, Smyth A. et al; CS4 Investigators. Phase 2 study of the factor XI antisense inhibitor IONIS-FXIRx in patients with ESRD. Kidney Int Rep 2021; 7 (02) 200-209
  • 69 Lorentz CU, Tucker EI, Verbout NG. et al. The contact activation inhibitor AB023 in heparin-free hemodialysis: results of a randomized phase 2 clinical trial. Blood 2021; 138 (22) 2173-2184
  • 70 Gailani D, Broze Jr GJ. Factor XI activation in a revised model of blood coagulation. Science 1991; 253 (5022): 909-912
  • 71 Lewandowska MD, Connors JM. Factor XI deficiency. Hematol Oncol Clin North Am 2021; 35 (06) 1157-1169
  • 72 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 (09) 1210-1215
  • 73 Georgi B, Mielke J, Chaffin M. et al. Leveraging human genetics to estimate clinical risk reductions achievable by inhibiting factor XI. Stroke 2019; 50 (11) 3004-3012
  • 74 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 (03) 411-415
  • 75 Meijers JCM, Tekelenburg WLH, 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
  • 76 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
  • 77 Kalinin DV. Factor XII(a) inhibitors: a review of the patent literature. Expert Opin Ther Pat 2021; 31 (12) 1155-1176
  • 78 Craig T, Magerl M, Levy DS. et al. Prophylactic use of an anti-activated factor XII monoclonal antibody, garadacimab, for patients with C1-esterase inhibitor-deficient hereditary angioedema: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2022; 399 (10328): 945-955
  • 79 Naito N, Ukita R, Wilbs J. et al. Combination of polycarboxybetaine coating and factor XII inhibitor reduces clot formation while preserving normal tissue coagulation during extracorporeal life support. Biomaterials 2021; 272: 120778
  • 80 Yau JW, Liao P, Fredenburgh JC. et al. Selective depletion of factor XI or factor XII with antisense oligonucleotides attenuates catheter thrombosis in rabbits. Blood 2014; 123 (13) 2102-2107
  • 81 Tucker EI, Marzec UM, White TC. et al. Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI. Blood 2009; 113 (04) 936-944