CC BY 4.0 · TH Open 2023; 07(01): e1-e13
DOI: 10.1055/s-0042-1758855
Original Article

Edoxaban, a Factor Xa-Specific Direct Oral Anticoagulant, Significantly Suppresses Tumor Growth in Colorectal Cancer Colon26-Inoculated BALB/c Mice

Keiichi Hiramoto
1   Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
,
Nobuyuki Akita
2   Department of Clinical Engineering, Faculty of Medical Engineering, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
,
Junji Nishioka
3   Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
,
Koji Suzuki
1   Department of Molecular Pathobiology, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka-city, Mie, Japan
› Author Affiliations
Funding This work was supported by the Japan Society for the Promotion of Science (grant numbers 16K08633 and 19K08850).

Abstract

Introduction Certain low-molecular-weight heparins have been reported to reduce tumor growth and metastasis in tumor cell-inoculated mouse models and cancer patients. Recently, direct oral anticoagulants (DOACs) have been widely used in patients with thromboembolism. This study was aimed at investigating the effect of DOACs, which target thrombin or factor Xa, on tumor growth in a syngeneic mouse model comprising BALB/c mice inoculated with colon cancer Colon26 cells.

Materials and Methods DOACs targeting thrombin (dabigatran etexilate [DABE]) or factor Xa (rivaroxaban [RVX] and edoxaban [EDX]) were orally administered daily to male BALB/c mice inoculated with Colon26 cells, followed by analyses of tumor growth and plasma levels of coagulation- and tumor-related factors such as tissue factor (TF), plasminogen activator inhibitor-1 (PAI-1), interleukin-6 (IL-6), and matrix metalloproteinase-2 (MMP-2).

Results Colon26 cells expressed significant amounts of functionally active TF. Tumor growth in Colon26-inoculated mice was significantly suppressed in DABE- or RVX-treated mice (p <0.05) and was suppressed more significantly in EDX-treated mice (p <0.01). Therefore, the antitumor mechanism of action of EDX was investigated next. Plasma levels of TF, PAI-1, IL-6, and MMP-2 were elevated in Colon26-inoculated mice but were significantly reduced in EDX-treated mice (p <0.01). The expression of protease-activated receptor (PAR)1, PAR2, signal transducer and activator of transcription-3 (STAT3), cyclin D1, and Ki67 was increased in tumor tissue of Colon26-inoculated mice but (except for PAR1) was significantly decreased in tumor tissues of EDX-treated mice (p <0.01). In addition, apoptotic cells and p53 protein levels were significantly increased in tumor tissues of EDX-treated mice.

Conclusion The data suggest that among the tested DOACs, EDX significantly suppresses tumor cell proliferation via the factor Xa-PAR2 pathway, which is activated by coagulation and inflammation in Colon26-inoculated mice and induces tumor cell apoptosis.

Supplementary Material



Publication History

Received: 30 June 2022

Accepted: 10 October 2022

Article published online:
07 January 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Dammacco F, Vacca A, Procaccio P, Ria R, Marech I, Racanelli V. Cancer-related coagulopathy (Trousseau's syndrome): review of the literature and experience of a single center of internal medicine. Clin Exp Med 2013; 13 (02) 85-97
  • 2 Zelaya H, Rothmeier AS, Ruf W. Tissue factor at the crossroad of coagulation and cell signaling. J Thromb Haemost 2018; 16 (10) 1941-1952
  • 3 Lee AYY, Levine MN, Baker RI. et al; Randomized Comparison of Low-Molecular-Weight Heparin versus Oral Anticoagulant Therapy for the Prevention of Recurrent Venous Thromboembolism in Patients with Cancer (CLOT) Investigators. Low-molecular-weight heparin versus a coumarin for the prevention of recurrent venous thromboembolism in patients with cancer. N Engl J Med 2003; 349 (02) 146-153
  • 4 Schulman S, Kearon C, Kakkar AK. et al; RE-COVER Study Group. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med 2009; 361 (24) 2342-2352
  • 5 Perzborn E, Roehrig S, Straub A, Kubitza D, Misselwitz F. The discovery and development of rivaroxaban, an oral, direct factor Xa inhibitor. Nat Rev Drug Discov 2011; 10 (01) 61-75
  • 6 Lassen MR, Davidson BL, Gallus A, Pineo G, Ansell J, Deitchman D. The efficacy and safety of apixaban, an oral, direct factor Xa inhibitor, as thromboprophylaxis in patients following total knee replacement. J Thromb Haemost 2007; 5 (12) 2368-2375
  • 7 Büller HR, Décousus H, Grosso MA. et al; Hokusai-VTE Investigators. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med 2013; 369 (15) 1406-1415
  • 8 Kakkar AK, Levine MN, Kadziola Z. et al. Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the fragmin advanced malignancy outcome study (FAMOUS). J Clin Oncol 2004; 22 (10) 1944-1948
  • 9 Klerk CPW, Smorenburg SM, Otten HM. et al. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol 2005; 23 (10) 2130-2135
  • 10 Mousa SA, Petersen LJ. Anti-cancer properties of low-molecular-weight heparin: preclinical evidence. Thromb Haemost 2009; 102 (02) 258-267
  • 11 Montroy J, Lalu MM, Auer RC. et al. The efficacy and safety of low molecular weight heparin administration to improve survival of cancer patients: a systematic review and meta-analysis. Thromb Haemost 2020; 120 (05) 832-846
  • 12 Altinbas M, Coskun HS, Er O. et al. A randomized clinical trial of combination chemotherapy with and without low-molecular-weight heparin in small cell lung cancer. J Thromb Haemost 2004; 2 (08) 1266-1271
  • 13 Lecumberri R, López Vivanco G, Font A. et al. Adjuvant therapy with bemiparin in patients with limited-stage small cell lung cancer: results from the ABEL study. Thromb Res 2013; 132 (06) 666-670
  • 14 Bock F, Shahzad K, Vergnolle N, Isermann B. Activated protein C based therapeutic strategies in chronic diseases. Thromb Haemost 2014; 111 (04) 610-617
  • 15 Gomi K, Zushi M, Honda G. et al. Antithrombotic effect of recombinant human thrombomodulin on thrombin-induced thromboembolism in mice. Blood 1990; 75 (07) 1396-1399
  • 16 Akita N, Ma N, Okamoto T. et al. Host protein C inhibitor inhibits tumor growth, but promotes tumor metastasis, which is closely correlated with hypercoagulability. Thromb Res 2015; 135 (06) 1203-1208
  • 17 Amada E, Fukuda K, Kumagai K, Kawakubo H, Kitagawa Y. Soluble recombinant human thrombomodulin suppresses inflammation-induced gastrointestinal tumor growth in a murine peritonitis model. Mol Cell Biochem 2020; 475 (1-2): 195-203
  • 18 Shimizu T, Abu Lila AS, Nishio M. et al. Modulation of antitumor immunity contributes to the enhanced therapeutic efficacy of liposomal oxaliplatin in mouse model. Cancer Sci 2017; 108 (09) 1864-1869
  • 19 Bohmann F, Mirceska A, Pfeilschifter J. et al. No influence of dabigatran anticoagulation on hemorrhagic transformation in an experimental model of ischemic stroke. Kleinschnitz C, ed. PLoS ONE 2012; 7: e40804
  • 20 Hara T, Fukuda D, Tanaka K. et al. Rivaroxaban, a novel oral anticoagulant, attenuates atherosclerotic plaque progression and destabilization in ApoE-deficient mice. Atherosclerosis 2015; 242 (02) 639-646
  • 21 Morishima Y, Honda Y. A direct oral factor Xa inhibitor edoxaban ameliorates neointimal hyperplasia following vascular injury and thrombosis in apolipoprotein E-deficient mice. J Thromb Thrombolysis 2018; 46 (01) 95-101
  • 22 Crowley LC, Marfell BJ, Waterhouse NJ. Detection of DNA fragmentation in apoptotic cells by TUNEL. Cold Spring Harb Protoc 2016; ;2016(10); DOI: 10.1101/pdb.prot087221.
  • 23 Nishiyama M, Oshikawa K, Tsukada Y. et al. CHD8 suppresses p53-mediated apoptosis through histone H1 recruitment during early embryogenesis. Nat Cell Biol 2009; 11 (02) 172-182
  • 24 Nierodzik ML, Karpatkin S. Thrombin induces tumor growth, metastasis, and angiogenesis: evidence for a thrombin-regulated dormant tumor phenotype. Cancer Cell 2006; 10 (05) 355-362
  • 25 Wojtukiewicz MZ, Tang DG, Nelson KK, Walz DA, Diglio CA, Honn KV. Thrombin enhances tumor cell adhesive and metastatic properties via increased α IIb β 3 expression on the cell surface. Thromb Res 1992; 68 (03) 233-245
  • 26 Otsuki T, Fujimoto D, Hirono Y, Goi T, Yamaguchi A. Thrombin conducts epithelial–mesenchymal transition via protease–activated receptor–1 in human gastric cancer. Int J Oncol 2014; 45 (06) 2287-2294
  • 27 Morris DR, Ding Y, Ricks TK, Gullapalli A, Wolfe BL, Trejo J. Protease-activated receptor-2 is essential for factor VIIa and Xa-induced signaling, migration, and invasion of breast cancer cells. Cancer Res 2006; 66 (01) 307-314
  • 28 Jiang X, Bailly MA, Panetti TS, Cappello M, Konigsberg WH, Bromberg ME. Formation of tissue factor-factor VIIa-factor Xa complex promotes cellular signaling and migration of human breast cancer cells. J Thromb Haemost 2004; 2 (01) 93-101
  • 29 Nishibori M, Mori S, Takahashi HK. Physiology and pathophysiology of proteinase-activated receptors (PARs): PAR-2-mediated proliferation of colon cancer cell. J Pharmacol Sci 2005; 97 (01) 25-30
  • 30 Xie L, Duan Z, Liu C, Zheng Y, Zhou J. Protease-activated receptor 2 agonist increases cell proliferation and invasion of human pancreatic cancer cells. Exp Ther Med 2015; 9 (01) 239-244
  • 31 Iablokov V, Hirota CL, Peplowski MA. et al. Proteinase-activated receptor 2 (PAR2) decreases apoptosis in colonic epithelial cells. J Biol Chem 2014; 289 (49) 34366-34377
  • 32 Shanshan H, Lan X, Xia L, Huang W, Meifang Z, Ling Y. Inhibition of protease-activated receptor-2 induces apoptosis in cervical cancer by inhibiting signal transducer and activator of transcription-3 signaling. J Int Med Res 2019; 47 (03) 1330-1338
  • 33 Zain J, Huang YQ, Feng X, Nierodzik ML, Li JJ, Karpatkin S. Concentration-dependent dual effect of thrombin on impaired growth/apoptosis or mitogenesis in tumor cells. Blood 2000; 95 (10) 3133-3138
  • 34 Sébert M, Denadai-Souza A, Quaranta M. et al. Thrombin modifies growth, proliferation and apoptosis of human colon organoids: a protease-activated receptor 1- and protease-activated receptor 4-dependent mechanism. Br J Pharmacol 2018; 175 (18) 3656-3668
  • 35 Zhu T, Sennlaub F, Beauchamp MH. et al. Proangiogenic effects of protease-activated receptor 2 are tumor necrosis factor-alpha and consecutively Tie2 dependent. Arterioscler Thromb Vasc Biol 2006; 26 (04) 744-750
  • 36 Teleanu RI, Chircov C, Grumezescu AM, Teleanu DM. Tumor angiogenesis and anti-angiogenic strategies for cancer treatment. J Clin Med 2019; 9 (01) 84-104
  • 37 Cui C, Gao J, Li J, Yu M, Zhang H, Cui W. Value of TAT and PIC with D-dimer for cancer patients with metastasis. Int J Lab Hematol 2020; 42 (04) 387-393
  • 38 Furugohri T, Isobe K, Honda Y. et al. DU-176b, a potent and orally active factor Xa inhibitor: in vitro and in vivo pharmacological profiles. J Thromb Haemost 2008; 6 (09) 1542-1549
  • 39 Kim PY, Yeh CH, Dale BJ. et al. Mechanistic basis for the differential effects of rivaroxaban and apixaban on global tests of coagulation. TH Open 2018; 2 (02) e190-e201
  • 40 Yamamoto Y, Uchida T. Dielectric blood coagulometry as a means of evaluating the change in thrombin generation induced by direct oral anticoagulants. Thromb Res 2021; 197: 141-143
  • 41 Gerotziafas GT, Elalamy I, Depasse F, Perzborn E, Samama MM. In vitro inhibition of thrombin generation, after tissue factor pathway activation, by the oral, direct factor Xa inhibitor rivaroxaban. J Thromb Haemost 2007; 5 (04) 886-888
  • 42 Nhu QM, Shirey KA, Pennini ME, Stiltz J, Vogel SN. Proteinase-activated receptor 2 activation promotes an anti-inflammatory and alternatively activated phenotype in LPS-stimulated murine macrophages. Innate Immun 2012; 18 (02) 193-203
  • 43 Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res 2019; 79 (18) 4557-4566
  • 44 Buijs JT, Laghmani EH, van den Akker RFP. et al. The direct oral anticoagulants rivaroxaban and dabigatran do not inhibit orthotopic growth and metastasis of human breast cancer in mice. J Thromb Haemost 2019; 17 (06) 951-963
  • 45 Najidh S, Versteeg HH, Buijs JT. A systematic review on the effects of direct oral anticoagulants on cancer growth and metastasis in animal models. Thromb Res 2020; 187: 18-27