Semin Thromb Hemost 2021; 47(08): 962-971
DOI: 10.1055/s-0041-1733925
Review Article

Hemostatic Biomarkers and Cancer Prognosis: Where Do We Stand?

Anna Falanga
1   Division of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
2   Department of Medicine and Surgery, University of Milan Bicocca, Milan, Italy
,
Marina Marchetti
1   Division of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
,
Laura Russo
1   Division of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
› Author Affiliations
Funding The HYPERCAN study (ClinicalTrials.gov # NCT0262281) is funded by AIRC “5xMILLE” n. 12237 grant from the “Italian Association for Cancer Research (AIRC).”

Abstract

Cancer patients are characterized by hypercoagulable state and an increased rate of thrombotic events, the most common being venous thromboembolism. Several hemostatic pathways that are significantly implicated in mechanisms of thromboembolic disease are also involved in growth, invasion, and metastatic spread of malignant cells as well in tumor-induced neo-angiogenesis. This close connection between cancer and the hemostatic system has prompted numerous studies on the role of alterations in the level plasma biomarkers of the different compartments of hemostasis in predicting cancer prognosis. In this review, we collect the results of several exemplificative studies that have evaluated clotting activation biomarkers in relation to different cancer outcomes with a final emphasis on current research and forthcoming directions in this field.



Publication History

Article published online:
27 August 2021

© 2021. Thieme. All rights reserved.

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

 
  • References

  • 1 Falanga A, Marchetti M, Vignoli A. Coagulation and cancer: biological and clinical aspects. J Thromb Haemost 2013; 11 (02) 223-233
  • 2 Shoji M, Hancock WW, Abe K. et al. Activation of coagulation and angiogenesis in cancer: immunohistochemical localization in situ of clotting proteins and vascular endothelial growth factor in human cancer. Am J Pathol 1998; 152 (02) 399-411
  • 3 Falanga A, Marchetti M, Russo L. The mechanisms of cancer-associated thrombosis. Thromb Res 2015; 135 (Suppl. 01) S8-S11
  • 4 Timp JF, Braekkan SK, Versteeg HH, Cannegieter SC. Epidemiology of cancer-associated venous thrombosis. Blood 2013; 122 (10) 1712-1723
  • 5 Khorana AA, McCrae KR. Risk stratification strategies for cancer-associated thrombosis: an update. Thromb Res 2014; 133 (Suppl. 02) S35-S38
  • 6 Falanga A, Russo L. Epidemiology, risk and outcomes of venous thromboembolism in cancer. Hamostaseologie 2012; 32 (02) 115-125
  • 7 Falanga A, Panova-Noeva M, Russo L. Procoagulant mechanisms in tumour cells. Best Pract Res Clin Haematol 2009; 22 (01) 49-60
  • 8 Adams GN, Rosenfeldt L, Frederick M. et al. Colon cancer growth and dissemination relies upon thrombin, stromal PAR-1, and fibrinogen. Cancer Res 2015; 75 (19) 4235-4243
  • 9 Rak J, Milsom C, Magnus N, Yu J. Tissue factor in tumour progression. Best Pract Res Clin Haematol 2009; 22 (01) 71-83
  • 10 Rickles FR, Falanga A. Activation of clotting factors in cancer. Cancer Treat Res 2009; 148: 31-41
  • 11 Zhang PP, Sun JW, Wang XY, Liu XM, Li K. Preoperative plasma D-dimer levels predict survival in patients with operable non-small cell lung cancer independently of venous thromboembolism. Eur J Surg Oncol 2013; 39 (09) 951-956
  • 12 Buccheri G, Torchio P, Ferrigno D. Plasma levels of D-dimer in lung carcinoma: clinical and prognostic significance. Cancer 2003; 97 (12) 3044-3052
  • 13 Taguchi O, Gabazza EC, Yasui H, Kobayashi T, Yoshida M, Kobayashi H. Prognostic significance of plasma D-dimer levels in patients with lung cancer. Thorax 1997; 52 (06) 563-565
  • 14 Komurcuoglu B, Ulusoy S, Gayaf M, Guler A, Ozden E. Prognostic value of plasma D-dimer levels in lung carcinoma. Tumori 2011; 97 (06) 743-748
  • 15 Chen Y, Yu H, Wu C. et al. Prognostic value of plasma D-dimer levels in patients with small-cell lung cancer. Biomed Pharmacother 2016; 81: 210-217
  • 16 Altiay G, Ciftci A, Demir M. et al. High plasma D-dimer level is associated with decreased survival in patients with lung cancer. Clin Oncol (R Coll Radiol) 2007; 19 (07) 494-498
  • 17 Zhu LR, Li J, Chen P, Jiang Q, Tang XP. Clinical significance of plasma fibrinogen and D-dimer in predicting the chemotherapy efficacy and prognosis for small cell lung cancer patients. Clin Transl Oncol 2016; 18 (02) 178-188
  • 18 Moik F, Zöchbauer-Müller S, Posch F, Pabinger I, Ay C. Systemic inflammation and activation of haemostasis predict poor prognosis and response to chemotherapy in patients with advanced lung cancer. Cancers (Basel) 2020;12(06):
  • 19 Ma X, Li Y, Zhang J, Huang J, Liu L. Prognostic role of D-dimer in patients with lung cancer: a meta-analysis. Tumour Biol 2014; 35 (03) 2103-2109
  • 20 Zhou YX, Yang ZM, Feng J, Shan YJ, Wang WL, Mei YQ. High plasma D-dimer level is associated with decreased survival in patients with lung cancer: a meta-analysis. Tumour Biol 2013; 34 (06) 3701-3704
  • 21 Zhang Y, Cao J, Deng Y. et al. Pretreatment plasma fibrinogen level as a prognostic biomarker for patients with lung cancer. Clinics (São Paulo) 2020; 75: e993
  • 22 Hong T, Shen D, Chen X, Wu X, Hua D. Preoperative plasma fibrinogen, but not D-dimer might represent a prognostic factor in non-metastatic colorectal cancer: A prospective cohort study. Cancer Biomark 2017; 19 (01) 103-111
  • 23 Oya M, Akiyama Y, Okuyama T, Ishikawa H. High preoperative plasma D-dimer level is associated with advanced tumor stage and short survival after curative resection in patients with colorectal cancer. Jpn J Clin Oncol 2001; 31 (08) 388-394
  • 24 Tang L, Liu K, Wang J, Wang C, Zhao P, Liu J. High preoperative plasma fibrinogen levels are associated with distant metastases and impaired prognosis after curative resection in patients with colorectal cancer. J Surg Oncol 2010; 102 (05) 428-432
  • 25 Lee S, Huh SJ, Oh SY. et al. Clinical significance of coagulation factors in operable colorectal cancer. Oncol Lett 2017; 13 (06) 4669-4674
  • 26 Blackwell K, Hurwitz H, Liebérman G. et al. Circulating D-dimer levels are better predictors of overall survival and disease progression than carcinoembryonic antigen levels in patients with metastatic colorectal carcinoma. Cancer 2004; 101 (01) 77-82
  • 27 Kilic M, Yoldas O, Keskek M. et al. Prognostic value of plasma D-dimer levels in patients with colorectal cancer. Colorectal Dis 2008; 10 (03) 238-241
  • 28 Motavaf E, Sunesen KG, Stender MT, Thorlacius-Ussing O. Prognostic value of preoperative D-dimer and carcinoembryonic antigen levels in patients undergoing intended curative resection for colorectal cancer: a prospective cohort study. Int J Colorectal Dis 2014; 29 (11) 1427-1432
  • 29 Pedrazzani C, Cerullo G, Marrelli D. et al. Is circulating D-dimer level a better prognostic indicator than CEA in resectable colorectal cancer? Our experience on 199 cases. Int J Biol Markers 2010; 25 (03) 171-176
  • 30 Lu SL, Ye ZH, Ling T. et al. High pretreatment plasma D-dimer predicts poor survival of colorectal cancer: insight from a meta-analysis of observational studies. Oncotarget 2017; 8 (46) 81186-81194
  • 31 Pedrazzani C, Mantovani G, Salvagno GL. et al. Elevated fibrinogen plasma level is not an independent predictor of poor prognosis in a large cohort of Western patients undergoing surgery for colorectal cancer. World J Gastroenterol 2016; 22 (45) 9994-10001
  • 32 Lee JH, Hyun JH, Kim DY. et al. The role of fibrinogen as a predictor in preoperative chemoradiation for rectal cancer. Ann Surg Oncol 2015; 22 (01) 209-215
  • 33 Li M, Wu Y, Zhang J, Huang L, Wu X, Yuan Y. Prognostic value of pretreatment plasma fibrinogen in patients with colorectal cancer: a systematic review and meta-analysis. Medicine (Baltimore) 2019; 98 (37) e16974
  • 34 Ay C, Dunkler D, Pirker R. et al. High D-dimer levels are associated with poor prognosis in cancer patients. Haematologica 2012; 97 (08) 1158-1164
  • 35 Moik F, Posch F, Grilz E. et al. Haemostatic biomarkers for prognosis and prediction of therapy response in patients with metastatic colorectal cancer. Thromb Res 2020; 187: 9-17
  • 36 Lee JH, Ryu KW, Kim S, Bae JM. Preoperative plasma fibrinogen levels in gastric cancer patients correlate with extent of tumor. Hepatogastroenterology 2004; 51 (60) 1860-1863
  • 37 Liu L, Zhang X, Yan B. et al. Elevated plasma D-dimer levels correlate with long term survival of gastric cancer patients. PLoS One 2014; 9 (03) e90547
  • 38 Yu X, Hu F, Yao Q, Li C, Zhang H, Xue Y. Serum fibrinogen levels are positively correlated with advanced tumor stage and poor survival in patients with gastric cancer undergoing gastrectomy: a large cohort retrospective study. BMC Cancer 2016; 16: 480
  • 39 Blackwell K, Haroon Z, Broadwater G. et al. Plasma D-dimer levels in operable breast cancer patients correlate with clinical stage and axillary lymph node status. J Clin Oncol 2000; 18 (03) 600-608
  • 40 Wen J, Yang Y, Ye F. et al. The preoperative plasma fibrinogen level is an independent prognostic factor for overall survival of breast cancer patients who underwent surgical treatment. Breast 2015; 24 (06) 745-750
  • 41 Krenn-Pilko S, Langsenlehner U, Stojakovic T. et al. An elevated preoperative plasma fibrinogen level is associated with poor disease-specific and overall survival in breast cancer patients. Breast 2015; 24 (05) 667-672
  • 42 Liu YL, Lu Q, Liang JW. et al. High plasma fibrinogen is correlated with poor response to trastuzumab treatment in HER2 positive breast cancer. Medicine (Baltimore) 2015; 94 (05) e481
  • 43 Man YN, Wang YN, Hao J. et al. Pretreatment plasma D-dimer, fibrinogen, and platelet levels significantly impact prognosis in patients with epithelial ovarian cancer independently of venous thromboembolism. Int J Gynecol Cancer 2015; 25 (01) 24-32
  • 44 Sakurai M, Satoh T, Matsumoto K. et al. High pretreatment plasma D-dimer levels are associated with poor prognosis in patients with ovarian cancer independently of venous thromboembolism and tumor extension. Int J Gynecol Cancer 2015; 25 (04) 593-598
  • 45 Zhao K, Deng H, Qin Y, Liao W, Liang W. Prognostic significance of pretreatment plasma fibrinogen and platelet levels in patients with early-stage cervical cancer. Gynecol Obstet Invest 2015; 79 (01) 25-33
  • 46 Yamada Y, Kawaguchi R, Iwai K. et al. Preoperative plasma D-dimer level is a useful prognostic marker in ovarian cancer. J Obstet Gynaecol 2020; 40 (01) 102-106
  • 47 Liu P, Wang Y, Tong L. et al. Elevated preoperative plasma D-dimer level is a useful predictor of chemoresistance and poor disease outcome for serous ovarian cancer patients. Cancer Chemother Pharmacol 2015; 76 (06) 1163-1171
  • 48 Xu L, He F, Wang H, Gao B, Wu H, Zhao S. A high plasma D-dimer level predicts poor prognosis in gynecological tumors in East Asia area: a systematic review and meta-analysis. Oncotarget 2017; 8 (31) 51551-51558
  • 49 Wu J, Fu Z, Liu G, Xu P, Xu J, Jia X. Clinical significance of plasma D-dimer in ovarian cancer: a meta-analysis. Medicine (Baltimore) 2017; 96 (25) e7062
  • 50 Shu YJ, Weng H, Bao RF. et al. Clinical and prognostic significance of preoperative plasma hyperfibrinogenemia in gallbladder cancer patients following surgical resection: a retrospective and in vitro study. BMC Cancer 2014; 14: 566
  • 51 Erdem S, Amasyali AS, Aytac O, Onem K, Issever H, Sanli O. Increased preoperative levels of plasma fibrinogen and D dimer in patients with renal cell carcinoma is associated with poor survival and adverse tumor characteristics. Urol Oncol 2014; 32 (07) 1031-1040
  • 52 Falanga A, Marchetti M, Massi D. et al. Thrombophilic status may predict prognosis in patients with metastatic BRAFV600-mutated melanoma who are receiving BRAF inhibitors. J Am Acad Dermatol 2016; 74 (06) 1254-1256.e4
  • 53 Liu R, Zheng S, Yuan Q. et al. The prognostic significance of combined pretreatment fibrinogen and neutrophil-lymphocyte ratio in various cancers: a systematic review and meta-analysis. Dis Markers 2020; 2020: 4565379
  • 54 Mengele K, Napieralski R, Magdolen V. et al. Characteristics of the level-of-evidence-1 disease forecast cancer biomarkers uPA and its inhibitor PAI-1. Expert Rev Mol Diagn 2010; 10 (07) 947-962
  • 55 Duffy MJ, McGowan PM, Harbeck N, Thomssen C, Schmitt M. uPA and PAI-1 as biomarkers in breast cancer: validated for clinical use in level-of-evidence-1 studies. Breast Cancer Res 2014; 16 (04) 428
  • 56 Duffy MJ, Harbeck N, Nap M. et al. Clinical use of biomarkers in breast cancer: updated guidelines from the European Group on Tumor Markers (EGTM). Eur J Cancer 2017; 75: 284-298
  • 57 Dovnik NF, Takac I. Prognostic significance of uPA/PAI-1 level, HER2 status, and traditional histologic factors for survival in node-negative breast cancer patients. Radiol Oncol 2016; 51 (01) 65-73
  • 58 Stephens RW, Nielsen HJ, Christensen IJ. et al. Plasma urokinase receptor levels in patients with colorectal cancer: relationship to prognosis. J Natl Cancer Inst 1999; 91 (10) 869-874
  • 59 Fernebro E, Madsen RR, Fernö M. et al. Prognostic importance of the soluble plasminogen activator receptor, suPAR, in plasma from rectal cancer patients. Eur J Cancer 2001; 37 (04) 486-491
  • 60 Riisbro R, Christensen IJ, Nielsen HJ. et al. Preoperative plasma soluble urokinase plasminogen activator receptor as a prognostic marker in rectal cancer patients. An EORTC-Receptor and Biomarker Group collaboration. Int J Biol Markers 2005; 20 (02) 93-102
  • 61 Riisbro R, Stephens RW, Brünner N. et al. Soluble urokinase plasminogen activator receptor in preoperatively obtained plasma from patients with gynecological cancer or benign gynecological diseases. Gynecol Oncol 2001; 82 (03) 523-531
  • 62 Ferroni P, Roselli M, Portarena I. et al. Plasma plasminogen activator inhibitor-1 (PAI-1) levels in breast cancer - relationship with clinical outcome. Anticancer Res 2014; 34 (03) 1153-1161
  • 63 Versteeg HH. Tissue factor: old and new links with cancer biology. Semin Thromb Hemost 2015; 41 (07) 747-755
  • 64 Ruf W, Yokota N, Schaffner F. Tissue factor in cancer progression and angiogenesis. Thromb Res 2010; 125 (Suppl. 02) S36-S38
  • 65 Falanga A, Barbui T, Rickles FR. Hypercoagulability and tissue factor gene upregulation in hematologic malignancies. Semin Thromb Hemost 2008; 34 (02) 204-210
  • 66 Ueno T, Toi M, Koike M, Nakamura S, Tominaga T. Tissue factor expression in breast cancer tissues: its correlation with prognosis and plasma concentration. Br J Cancer 2000; 83 (02) 164-170
  • 67 Tieken C, Verboom MC, Ruf W. et al. Tissue factor associates with survival and regulates tumour progression in osteosarcoma. Thromb Haemost 2016; 115 (05) 1025-1033
  • 68 Falanga A, Tartari CJ, Marchetti M. Microparticles in tumor progression. Thromb Res 2012; 129 (Suppl. 01) S132-S136
  • 69 Marchetti M, Tartari CJ, Russo L. et al. Phospholipid-dependent procoagulant activity is highly expressed by circulating microparticles in patients with essential thrombocythemia. Am J Hematol 2014; 89 (01) 68-73
  • 70 Garnier D, Magnus N, D'Asti E. et al. Genetic pathways linking hemostasis and cancer. Thromb Res 2012; 129 (Suppl. 01) S22-S29
  • 71 Claussen C, Rausch AV, Lezius S. et al. Microvesicle-associated tissue factor procoagulant activity for the preoperative diagnosis of ovarian cancer. Thromb Res 2016; 141: 39-48
  • 72 Bharthuar A, Khorana AA, Hutson A. et al. Circulating microparticle tissue factor, thromboembolism and survival in pancreaticobiliary cancers. Thromb Res 2013; 132 (02) 180-184
  • 73 Thaler J, Ay C, Mackman N. et al. Microparticle-associated tissue factor activity in patients with pancreatic cancer: correlation with clinicopathological features. Eur J Clin Invest 2013; 43 (03) 277-285
  • 74 Bogdanov VY, Balasubramanian V, Hathcock J, Vele O, Lieb M, Nemerson Y. Alternatively spliced human tissue factor: a circulating, soluble, thrombogenic protein. Nat Med 2003; 9 (04) 458-462
  • 75 Unruh D, Sagin F, Adam M. et al. Levels of alternatively spliced tissue factor in the plasma of patients with pancreatic cancer may help predict aggressive tumor phenotype. Ann Surg Oncol 2015; 22 (Suppl. 03) S1206-S1211
  • 76 Kocatürk B, Van den Berg YW, Tieken C. et al. Alternatively spliced tissue factor promotes breast cancer growth in a β1 integrin-dependent manner. Proc Natl Acad Sci U S A 2013; 110 (28) 11517-11522
  • 77 Kocatürk B, Tieken C, Vreeken D. et al. Alternatively spliced tissue factor synergizes with the estrogen receptor pathway in promoting breast cancer progression. J Thromb Haemost 2015; 13 (09) 1683-1693
  • 78 Amirkhosravi A, Meyer T, Amaya M. et al. The role of tissue factor pathway inhibitor in tumor growth and metastasis. Semin Thromb Hemost 2007; 33 (07) 643-652
  • 79 Zhai LL, Cai CY, Wu Y, Tang ZG. Correlation and prognostic significance of MMP-2 and TFPI-2 differential expression in pancreatic carcinoma. Int J Clin Exp Pathol 2015; 8 (01) 682-691
  • 80 Xu C, Wang H, He H. et al. Low expression of TFPI-2 associated with poor survival outcome in patients with breast cancer. BMC Cancer 2013; 13: 118
  • 81 Wang S, Xiao X, Zhou X. et al. TFPI-2 is a putative tumor suppressor gene frequently inactivated by promoter hypermethylation in nasopharyngeal carcinoma. BMC Cancer 2010; 10: 617
  • 82 Wu D, Xiong L, Wu S, Jiang M, Lian G, Wang M. TFPI-2 methylation predicts poor prognosis in non-small cell lung cancer. Lung Cancer 2012; 76 (01) 106-111
  • 83 Balasenthil S, Huang Y, Liu S. et al. A plasma biomarker panel to identify surgically resectable early-stage pancreatic cancer. J Natl Cancer Inst 2017; 109 (08) djw341
  • 84 Luengo-Gil G, Calvo MI, Martín-Villar E. et al. Antithrombin controls tumor migration, invasion and angiogenesis by inhibition of enteropeptidase. Sci Rep 2016; 6: 27544
  • 85 Unsal E, Atalay F, Atikcan S, Yilmaz A. Prognostic significance of hemostatic parameters in patients with lung cancer. Respir Med 2004; 98 (02) 93-98
  • 86 Koh SC, Khalil R, Lim FK, Ilancheran A, Choolani M. The association between fibrinogen, von Willebrand Factor, antithrombin III, and D-dimer levels and survival outcome by 36 months from ovarian cancer. Clin Appl Thromb Hemost 2006; 12 (01) 3-8
  • 87 Wilts IT, Hutten BA, Meijers JCM, Spek CA, Büller HR, Kamphuisen PW. Association between protein C levels and mortality in patients with advanced prostate, lung and pancreatic cancer. Thromb Res 2017; 154: 1-6
  • 88 Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood 2014; 124 (10) 1553-1562
  • 89 Ruf W, Schaffner F. Role of the protein C receptor in cancer progression. Thromb Res 2014; 133 (Suppl. 02) S85-S89
  • 90 Thålin C, Lundström S, Seignez C. et al. Citrullinated histone H3 as a novel prognostic blood marker in patients with advanced cancer. PLoS One 2018; 13 (01) e0191231
  • 91 Garley M, Jabłońska E, Dąbrowska D. NETs in cancer. Tumour Biol 2016; 37 (11) 14355-14361
  • 92 Yang C, Sun W, Cui W. et al. Procoagulant role of neutrophil extracellular traps in patients with gastric cancer. Int J Clin Exp Pathol 2015; 8 (11) 14075-14086
  • 93 Demers M, Krause DS, Schatzberg D. et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci U S A 2012; 109 (32) 13076-13081
  • 94 Abdol Razak N, Elaskalani O, Metharom P. Pancreatic cancer-induced neutrophil extracellular traps: a potential contributor to cancer-associated thrombosis. Int J Mol Sci 2017; 18 (03) E487
  • 95 Leal AC, Mizurini DM, Gomes T. et al. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: implications for the establishment of cancer-associated thrombosis. Sci Rep 2017; 7 (01) 6438
  • 96 Li B, Liu Y, Hu T. et al. Neutrophil extracellular traps enhance procoagulant activity in patients with oral squamous cell carcinoma. J Cancer Res Clin Oncol 2019; 145 (07) 1695-1707
  • 97 Rosell A, Aguilera K, Hisada Y. et al. Prognostic value of circulating markers of neutrophil activation, neutrophil extracellular traps, coagulation and fibrinolysis in patients with terminal cancer. Sci Rep 2021; 11 (01) 5074
  • 98 D'Alessio A, Marchetti M, Tartari CJ. et al. Long term low molecular weight heparin anticoagulant therapy modulates thrombin generation and D-dimer in patients with cancer and venous thromboembolism. Cancer Invest 2017; 35 (07) 490-499
  • 99 Marchetti M, Castoldi E, Spronk HM. et al. Thrombin generation and activated protein C resistance in patients with essential thrombocythemia and polycythemia vera. Blood 2008; 112 (10) 4061-4068
  • 100 Reitter EM, Kaider A, Ay C. et al. Longitudinal analysis of hemostasis biomarkers in cancer patients during antitumor treatment. J Thromb Haemost 2016; 14 (02) 294-305
  • 101 Negaard HF, Iversen PO, Ostenstad B, Mowinckel MC, Sandset PM. Increased acquired activated protein C resistance in unselected patients with hematological malignancies. J Thromb Haemost 2008; 6 (09) 1482-1487
  • 102 Abu Saadeh F, Langhe R, Galvin DM. et al. Procoagulant activity in gynaecological cancer patients; the effect of surgery and chemotherapy. Thromb Res 2016; 139: 135-141
  • 103 Leiba M, Malkiel S, Budnik I. et al. Thrombin generation as a predictor of thromboembolic events in multiple myeloma patients. Blood Cells Mol Dis 2017; 65: 1-7
  • 104 Ay C, Dunkler D, Simanek R. et al. Prediction of venous thromboembolism in patients with cancer by measuring thrombin generation: results from the Vienna Cancer and Thrombosis Study. J Clin Oncol 2011; 29 (15) 2099-2103
  • 105 Roselli M, Ferroni P, Riondino S. et al. Impact of chemotherapy on activated protein C-dependent thrombin generation—association with VTE occurrence. Int J Cancer 2013; 133 (05) 1253-1258
  • 106 Tinholt M, Viken MK, Dahm AE. et al. Increased coagulation activity and genetic polymorphisms in the F5, F10 and EPCR genes are associated with breast cancer: a case-control study. BMC Cancer 2014; 14: 845
  • 107 Gezelius E, Flou Kristensen A, Bendahl PO. et al. Coagulation biomarkers and prediction of venous thromboembolism and survival in small cell lung cancer: a sub-study of RASTEN—a randomized trial with low molecular weight heparin. PLoS One 2018; 13 (11) e0207387
  • 108 Falanga A, Santoro A, Labianca R. et al; HYPERCAN Study Group. Hypercoagulation screening as an innovative tool for risk assessment, early diagnosis and prognosis in cancer: the HYPERCAN study. Thromb Res 2016; 140 (Suppl. 01) S55-S59
  • 109 Giaccherini C, Marchetti M, Masci G. et al; HYPERCAN Investigators. Thrombotic biomarkers for risk prediction of malignant disease recurrence in patients with early stage breast cancer. Haematologica 2020; 105 (06) 1704-1711
  • 110 Marchetti M, Giaccherini C, Masci G. et al; HYPERCAN Investigators. Thrombin generation predicts early recurrence in breast cancer patients. J Thromb Haemost 2020; 18 (09) 2220-2231
  • 111 Gomez-Rosas P, Pesenti M, Verzeroli C. et al; HYPERCAN Investigators. Validation of the role of thrombin generation potential by a fully automated system in the identification of breast cancer patients at high risk of disease recurrence. TH Open 2021; 5 (01) e56-e65