CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(02): 196-207
DOI: 10.1055/a-1663-8208
Coagulation and Fibrinolysis

Pharmacological Inhibition of Factor XIIa Attenuates Abdominal Aortic Aneurysm, Reduces Atherosclerosis, and Stabilizes Atherosclerotic Plaques

Amy K. Searle*
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Medicine, Monash University, Melbourne, Australia
,
Yung-Chih Chen*
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Medicine, Monash University, Melbourne, Australia
3   Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
,
Maria Wallert*
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
James D. McFadyen
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Medicine, Monash University, Melbourne, Australia
3   Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
4   Clinical Haematology, Alfred Hospital, Melbourne, Australia
,
Ana C. Maluenda
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Jonathan Noonan
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
3   Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
,
Peter Kanellakis
5   Atherosclerosis and Cell Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Maria T. K. Zaldivia
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Angela Huang
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Hadi Lioe
6   CSL Limited, Bio21 Institute, Parkville, Melbourne, Australia
,
Mark Biondo
6   CSL Limited, Bio21 Institute, Parkville, Melbourne, Australia
,
Marc W. Nolte
7   CSL Behring Innovation GmbH, Marburg, Germany
,
Paolo Rossato
7   CSL Behring Innovation GmbH, Marburg, Germany
,
Alex Bobik
2   Department of Medicine, Monash University, Melbourne, Australia
5   Atherosclerosis and Cell Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
8   Department of Immunology, Monash University, Melbourne, Australia
,
Con Panousis
6   CSL Limited, Bio21 Institute, Parkville, Melbourne, Australia
,
Xiaowei Wang**
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Medicine, Monash University, Melbourne, Australia
3   Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
,
Hamid Hosseini**
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Karlheinz Peter**
1   Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Medicine, Monash University, Melbourne, Australia
3   Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
8   Department of Immunology, Monash University, Melbourne, Australia
9   Department of Cardiology, Alfred Hospital, Melbourne, Australia
› Author Affiliations
Funding This work was supported by the National Heart Foundation of Australia for A.K.S., Y.-C.C., J.D.M., and X.W.; by the German Research Foundation for M.W.; and by the National Health and Medical Research Council for J.D.M. and K.P. CSL Limited supported this research with an unrestricted research fund.


Abstract

Background 3F7 is a monoclonal antibody targeting the enzymatic pocket of activated factor XII (FXIIa), thereby inhibiting its catalytic activity. Given the emerging role of FXIIa in promoting thromboinflammation, along with its apparent redundancy for hemostasis, the selective inhibition of FXIIa represents a novel and highly attractive approach targeting pathogenic processes that cause thromboinflammation-driven cardiovascular diseases.

Methods The effects of FXIIa inhibition were investigated using three distinct mouse models of cardiovascular disease—angiotensin II-induced abdominal aortic aneurysm (AAA), an ApoE−/− model of atherosclerosis, and a tandem stenosis model of atherosclerotic plaque instability. 3F7 or its isotype control, BM4, was administered to mice (10 mg/kg) on alternate days for 4 to 8 weeks, depending on the experimental model. Mice were examined for the development and size of AAAs, or the burden and instability of atherosclerosis and associated markers of inflammation.

Results Inhibition of FXIIa resulted in a reduced incidence of larger AAAs, with less acute aortic ruptures and an associated fibro-protective phenotype. FXIIa inhibition also decreased stable atherosclerotic plaque burden and achieved plaque stabilization associated with increased deposition of fibrous structures, a >2-fold thicker fibrous cap, increased cap-to-core ratio, and reduction in localized and systemic inflammatory markers.

Conclusion Inhibition of FXIIa attenuates disease severity across three mouse models of thromboinflammation-driven cardiovascular diseases. Specifically, the FXIIa-inhibiting monoclonal antibody 3F7 reduces AAA severity, inhibits the development of atherosclerosis, and stabilizes vulnerable plaques. Ultimately, clinical trials in patients with cardiovascular diseases such as AAA and atherosclerosis are warranted to demonstrate the therapeutic potential of FXIIa inhibition.

Author Contributions

Y.-C.C., C.P., X.W., H.H., and K.P. designed the study. A.K.S., Y.-C.C., M.W., J.D.M., A.C.M., J.N., P.K., M.T.K.Z., A.H., H.L., M.B., M.W.N., P.R., X.W., and H.H. were involved in the acquisition and interpretation or analysis of data. A.K.S. drafted the manuscript. Y.-C.C., M.W., J.D.M., J.N., A.B., C.P., X.W., and K.P. provided critical review of the intellectual content of the manuscript. All authors approved the final version of the manuscript prior to submission.


* Equally contributing first authors.


** Equally contributing senior authors.


Supplementary Material



Publication History

Received: 30 April 2021

Accepted: 04 October 2021

Accepted Manuscript online:
07 October 2021

Article published online:
21 January 2022

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 The World Health Organisation. The top 10 causes of death worldwide 2016. World Health Organization Fact Sheets. Published May 24, 2018. Accessed October 27, 2021 at: http://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death
  • 2 Libby P, Buring JE, Badimon L. et al. Atherosclerosis. Nat Rev Dis Primers 2019; 5 (01) 56
  • 3 Koch M, Zernecke A. The hemostatic system as a regulator of inflammation in atherosclerosis. IUBMB Life 2014; 66 (11) 735-744
  • 4 Borissoff JI, Spronk HMH, ten Cate H. The hemostatic system as a modulator of atherosclerosis. N Engl J Med 2011; 364 (18) 1746-1760
  • 5 Borissoff JI, Heeneman S, Kilinç E. et al. Early atherosclerosis exhibits an enhanced procoagulant state. Circulation 2010; 122 (08) 821-830
  • 6 Vorlova S, Koch M, Manthey HD. et al. Coagulation factor XII induces pro-inflammatory cytokine responses in macrophages and promotes atherosclerosis in mice. Thromb Haemost 2017; 117 (01) 176-187
  • 7 Shnerb Ganor R, Harats D, Schiby G. et al. Factor XI deficiency protects against atherogenesis in apolipoprotein E/factor XI double knockout mice. Arterioscler Thromb Vasc Biol 2016; 36 (03) 475-481
  • 8 Olie RH, van der Meijden PEJ, Ten Cate H. The coagulation system in atherothrombosis: implications for new therapeutic strategies. Res Pract Thromb Haemost 2018; 2 (02) 188-198
  • 9 Moran CS, Seto S-W, Krishna SM. et al. Parenteral administration of factor Xa/IIa inhibitors limits experimental aortic aneurysm and atherosclerosis. Sci Rep 2017; 7: 43079
  • 10 Müller F, Mutch NJ, Schenk WA. et al. Platelet polyphosphates are proinflammatory and procoagulant mediators in vivo. Cell 2009; 139 (06) 1143-1156
  • 11 Maas C, Govers-Riemslag JWP, Bouma B. et al. Misfolded proteins activate factor XII in humans, leading to kallikrein formation without initiating coagulation. J Clin Invest 2008; 118 (09) 3208-3218
  • 12 von Brühl M-L, Stark K, Steinhart A. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209 (04) 819-835
  • 13 van der Meijden PEJ, Munnix ICA, Auger JM. et al. Dual role of collagen in factor XII-dependent thrombus formation. Blood 2009; 114 (04) 881-890
  • 14 Renné T, Stavrou EX. Roles of factor XII in Innate Immunity. Front Immunol 2019; 10: 2011
  • 15 Worm M, Köhler EC, Panda R. et al. The factor XIIa blocking antibody 3F7: a safe anticoagulant with anti-inflammatory activities. Ann Transl Med 2015; 3 (17) 247
  • 16 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
  • 17 Chen Y-C, Bui AV, Diesch J. et al. A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling. Circ Res 2013; 113 (03) 252-265
  • 18 Chen YC, Rivera J, Peter K. Tandem stenosis to induce atherosclerotic plaque instability in the mouse. Methods Mol Biol 2015; 1339: 333-338
  • 19 Koay YC, Chen Y-C, Wali JA. et al. Plasma levels of TMAO can be increased with “healthy” and “unhealthy” diets and do not correlate with the extent of atherosclerosis but with plaque instability. Cardiovasc Res 2021; 117 (02) 435-449
  • 20 Spinosa M, Su G, Salmon MD. et al. Resolvin D1 decreases abdominal aortic aneurysm formation by inhibiting NETosis in a mouse model. J Vasc Surg 2018; 68 (6S): 93S-103S
  • 21 Meher AK, Spinosa M, Davis JP. et al. A novel role of IL-1β in NETosis and abdominal aortic aneurysms. Arterioscler Thromb Vasc Biol 2018; 38 (04) 843-853
  • 22 Haller SJ, Crawford JD, Courchaine KM. et al. Intraluminal thrombus is associated with early rupture of abdominal aortic aneurysm. J Vasc Surg 2018; 67 (04) 1051.e1-1058.e1
  • 23 Koole D, Zandvoort HJA, Schoneveld A. et al. Intraluminal abdominal aortic aneurysm thrombus is associated with disruption of wall integrity. J Vasc Surg 2013; 57 (01) 77-83
  • 24 Sénémaud J, Caligiuri G, Etienne H, Delbosc S, Michel J-B, Coscas R. Translational relevance and recent advances of animal models of abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol 2017; 37 (03) 401-410
  • 25 Golledge J. Abdominal aortic aneurysm: update on pathogenesis and medical treatments. Nat Rev Cardiol 2019; 16 (04) 225-242
  • 26 Zernecke A, Winkels H, Cochain C. et al. Meta-analysis of leukocyte diversity in atherosclerotic mouse aortas. Circ Res 2020; 127 (03) 402-426
  • 27 Fernandez DM, Rahman AH, Fernandez NF. et al. Single-cell immune landscape of human atherosclerotic plaques. Nat Med 2019; 25 (10) 1576-1588
  • 28 Zhao G, Lu H, Chang Z. et al. Single-cell RNA sequencing reveals the cellular heterogeneity of aneurysmal infrarenal abdominal aorta. Cardiovasc Res 2021; 117 (05) 1402-1416
  • 29 Kuivaniemi H, Ryer EJ, Elmore JR, Tromp G. Understanding the pathogenesis of abdominal aortic aneurysms. Expert Rev Cardiovasc Ther 2015; 13 (09) 975-987
  • 30 Cameron SJ, Russell HM, Owens III AP. Antithrombotic therapy in abdominal aortic aneurysm: beneficial or detrimental?. Blood 2018; 132 (25) 2619-2628
  • 31 Vorp DA, Lee PC, Wang DHJ. et al. Association of intraluminal thrombus in abdominal aortic aneurysm with local hypoxia and wall weakening. J Vasc Surg 2001; 34 (02) 291-299
  • 32 Moran CS, Seto S-W, Biros E. et al. Factor XII blockade inhibits aortic dilatation in angiotensin II-infused apolipoprotein E-deficient mice. Clin Sci (Lond) 2020; 134 (09) 1049-1061
  • 33 Wilson JS, Virag L, Di Achille P, Karšaj I, Humphrey JD. Biochemomechanics of intraluminal thrombus in abdominal aortic aneurysms. J Biomech Eng 2013; 135 (02) 021011
  • 34 Hansson GK, Libby P, Tabas I. Inflammation and plaque vulnerability. J Intern Med 2015; 278 (05) 483-493
  • 35 Rashid I, Maghzal GJ, Chen Y-C. et al. Myeloperoxidase is a potential molecular imaging and therapeutic target for the identification and stabilization of high-risk atherosclerotic plaque. Eur Heart J 2018; 39 (35) 3301-3310
  • 36 Stavrou EX, Fang C, Bane KL. et al. Factor XII and uPAR upregulate neutrophil functions to influence wound healing. J Clin Invest 2018; 128 (03) 944-959
  • 37 Göbel K, Pankratz S, Asaridou C-M. et al. Blood coagulation factor XII drives adaptive immunity during neuroinflammation via CD87-mediated modulation of dendritic cells. Nat Commun 2016; 7: 11626
  • 38 Ramji DP, Davies TS. Cytokines in atherosclerosis: key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26 (06) 673-685
  • 39 Naudin C, Burillo E, Blankenberg S, Butler L, Renné T. Factor XII contact activation. Semin Thromb Hemost 2017; 43 (08) 814-826
  • 40 Kuijpers MJE, van der Meijden PEJ, Feijge MAH. et al. Factor XII regulates the pathological process of thrombus formation on ruptured plaques. Arterioscler Thromb Vasc Biol 2014; 34 (08) 1674-1680
  • 41 Ngo ATP, Jordan KR, Mueller PA. et al. Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice. J Thromb Haemost 2021; 19 (04) 1001-1017
  • 42 Noonan J, Bobik A, Peter K. The tandem stenosis mouse model: towards understanding, imaging, and preventing atherosclerotic plaque instability and rupture. Br J Pharmacol 2021; 1-19 DOI: 10.1111/bph.15356.