Semin Thromb Hemost 2021; 47(06): 669-676
DOI: 10.1055/s-0040-1718926
Review Article

Cancer-Related Venous Thromboembolism: From Pathogenesis to Risk Assessment

José Costa
1   Department of Hematology and Transfusion Medicine, Centro Hospitalar de Trás-os-Montes e Alto Douro, Lordelo, Portugal
,
António Araújo
2   Department of Medical Oncology, Centro Hospitalar Universitário do Porto, Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
› Author Affiliations

Abstract

Cancer-related venous thromboembolism (VTE) remains a major health problem, accounting for at least 18% of all cases of VTE. Cancer patients with VTE have worse prognosis than those without VTE. Prophylaxis reduces VTE risk, but it is not feasible for all outpatients with cancer due to an increased bleeding risk. The factors involved in the pathogenesis of cancer-related VTE are direct coagulation activation, platelet activation, induction of inflammatory responses, and inhibition of fibrinolysis. Direct coagulation activation can be due to cancer procoagulant (a cysteine protease), microvesicles, or other prothrombotic abnormalities. Risk factors for developing VTE in cancer patients can be divided into four groups: tumor-related risk factors, patient-related risk factors, treatment-related risk factors, and biomarkers. Cancers of the pancreas, kidney, ovary, lung, and stomach have the highest rates of VTE. Patient-related risk factors such as age, obesity, or the presence of medical comorbidities can contribute to VTE. Platinum-based chemotherapies and antiangiogenesis treatments have also been associated with VTE. Biomarkers identified as risk factors include high platelet count, high leukocyte count, P-selectin, prothrombin fragments, D-dimer, and C-reactive protein. Based on the known risk factors, risk assessment models were developed to stratify patients who would benefit from thromboprophylaxis. The Khorana model was the first and is still the most widely used model. Because of its low sensitivity for certain tumor types, four new models have been developed in recent years. In this review, we describe the current knowledge about the pathogenesis and risk factors for cancer-related VTE, hoping to contribute to further research on the still many obscure aspects of this topic.



Publication History

Article published online:
14 May 2021

© 2021. Thieme. All rights reserved.

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

 
  • References

  • 1 Trousseau A. Phlegmasia alba dolens. In: Clinique Médicale de l'Hotel-Dieu de Paris. Vol. 3.. Paris: JB Baillière et Fils; 1865: 654-712
  • 2 Ay C, Pabinger I, Cohen AT. Cancer-associated venous thromboembolism: burden, mechanisms, and management. Thromb Haemost 2017; 117 (02) 219-230
  • 3 Heit JA, O'Fallon WM, Petterson TM. et al. Relative impact of risk factors for deep vein thrombosis and pulmonary embolism: a population-based study. Arch Intern Med 2002; 162 (11) 1245-1248
  • 4 Cronin-Fenton DP, Søndergaard F, Pedersen LA. et al. Hospitalisation for venous thromboembolism in cancer patients and the general population: a population-based cohort study in Denmark, 1997-2006. Br J Cancer 2010; 103 (07) 947-953
  • 5 Riedl J, Kaider A, Reitter EM. et al. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb Haemost 2014; 111 (04) 670-678
  • 6 Chew HK, Wun T, Harvey D, Zhou H, White RH. Incidence of venous thromboembolism and its effect on survival among patients with common cancers. Arch Intern Med 2006; 166 (04) 458-464
  • 7 Sørensen HT, Mellemkjaer L, Olsen JH, Baron JA. Prognosis of cancers associated with venous thromboembolism. N Engl J Med 2000; 343 (25) 1846-1850
  • 8 Tafur AJ, Wysokinski WE, McBane RD. et al. Cancer effect on periprocedural thromboembolism and bleeding in anticoagulated patients. Ann Oncol 2012; 23 (08) 1998-2005
  • 9 Shaw JR, Douketis J, Le Gal G, Carrier M. Periprocedural interruption of anticoagulation in patients with cancer-associated venous thromboembolism: an analysis of thrombotic and bleeding outcomes. J Thromb Haemost 2019; 17 (07) 1171-1178
  • 10 Kaźmierczak M, Lewandowski K, Wojtukiewicz MZ. et al. Cancer procoagulant in patients with adenocarcinomas. Blood Coagul Fibrinolysis 2005; 16 (08) 543-547
  • 11 Uno K, Homma S, Satoh T. et al. Tissue factor expression as a possible determinant of thromboembolism in ovarian cancer. Br J Cancer 2007; 96 (02) 290-295
  • 12 Thaler J, Preusser M, Ay C. et al. Intratumoral tissue factor expression and risk of venous thromboembolism in brain tumor patients. Thromb Res 2013; 131 (02) 162-165
  • 13 Freyssinet JM. Cellular microparticles: what are they bad or good for?. J Thromb Haemost 2003; 1 (07) 1655-1662
  • 14 Chen Y, Li G, Liu M-L. Microvesicles as emerging biomarkers and therapeutic targets in cardiometabolic diseases. Genomics Proteomics Bioinformatics 2018; 16 (01) 50-62
  • 15 Zwicker JI, Liebman HA, Neuberg D. et al. Tumor-derived tissue factor-bearing microparticles are associated with venous thromboembolic events in malignancy. Clin Cancer Res 2009; 15 (22) 6830-6840
  • 16 Zwicker JI, Liebman HA, Bauer KA. et al. Prediction and prevention of thromboembolic events with enoxaparin in cancer patients with elevated tissue factor-bearing microparticles: a randomized-controlled phase II trial (the Microtec study). Br J Haematol 2013; 160 (04) 530-537
  • 17 Thaler J, Ay C, Mackman N. et al. Microparticle-associated tissue factor activity, venous thromboembolism and mortality in pancreatic, gastric, colorectal and brain cancer patients. J Thromb Haemost 2012; 10 (07) 1363-1370
  • 18 Zhou L, Qi XL, Xu MX, Mao Y, Liu ML, Song HM. Microparticles: new light shed on the understanding of venous thromboembolism. Acta Pharmacol Sin 2014; 35 (09) 1103-1110
  • 19 Falati S, Liu Q, Gross P. et al. Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin. J Exp Med 2003; 197 (11) 1585-1598
  • 20 Minnema MC, Fijnheer R, De Groot PG, Lokhorst HM. Extremely high levels of von Willebrand factor antigen and of procoagulant factor VIII found in multiple myeloma patients are associated with activity status but not with thalidomide treatment. J Thromb Haemost 2003; 1 (03) 445-449
  • 21 Nickel KF, Ronquist G, Langer F. et al. The polyphosphate-factor XII pathway drives coagulation in prostate cancer-associated thrombosis. Blood 2015; 126 (11) 1379-1389
  • 22 Toth B, Liebhardt S, Steinig K. et al. Platelet-derived microparticles and coagulation activation in breast cancer patients. Thromb Haemost 2008; 100 (04) 663-669
  • 23 Mitrugno A, Tassi Yunga S, Sylman JL. et al. The role of coagulation and platelets in colon cancer-associated thrombosis. Am J Physiol Cell Physiol 2019; 316 (02) C264-C273
  • 24 Heinmöller E, Weinel RJ, Heidtmann HH. et al. Studies on tumor-cell-induced platelet aggregation in human lung cancer cell lines. J Cancer Res Clin Oncol 1996; 122 (12) 735-744
  • 25 Heinmöller E, Schropp T, Kisker O, Simon B, Seitz R, Weinel RJ. Tumor cell-induced platelet aggregation in vitro by human pancreatic cancer cell lines. Scand J Gastroenterol 1995; 30 (10) 1008-1016
  • 26 Riedl J, Preusser M, Nazari PMS. et al. Podoplanin expression in primary brain tumors induces platelet aggregation and increases risk of venous thromboembolism. Blood 2017; 129 (13) 1831-1839
  • 27 Watson SP, Herbert JMS, Pollitt AY. GPVI and CLEC-2 in hemostasis and vascular integrity. J Thromb Haemost 2010; 8 (07) 1456-1467
  • 28 Riedl J, Pabinger I, Ay C. Platelets in cancer and thrombosis. Hamostaseologie 2014; 34 (01) 54-62
  • 29 Szotowski B, Antoniak S, Poller W, Schultheiss HP, Rauch U. Procoagulant soluble tissue factor is released from endothelial cells in response to inflammatory cytokines. Circ Res 2005; 96 (12) 1233-1239
  • 30 Bernardo A, Ball C, Nolasco L, Moake JF, Dong JF. Effects of inflammatory cytokines on the release and cleavage of the endothelial cell-derived ultralarge von Willebrand factor multimers under flow. Blood 2004; 104 (01) 100-106
  • 31 Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 2012; 32 (08) 1777-1783
  • 32 Casslén B, Bossmar T, Lecander I, Astedt B. Plasminogen activators and plasminogen activator inhibitors in blood and tumour fluids of patients with ovarian cancer. Eur J Cancer 1994; 30A (09) 1302-1309
  • 33 Yağci M, Sucak GT, Haznedar R. Fibrinolytic activity in multiple myeloma. Am J Hematol 2003; 74 (04) 231-237
  • 34 Khorana AA, Francis CW, Culakova E, Kuderer NM, Lyman GH. Frequency, risk factors, and trends for venous thromboembolism among hospitalized cancer patients. Cancer 2007; 110 (10) 2339-2346
  • 35 Blom JW, Vanderschoot JP, Oostindiër MJ, Osanto S, van der Meer FJ, Rosendaal FR. Incidence of venous thrombosis in a large cohort of 66,329 cancer patients: results of a record linkage study. J Thromb Haemost 2006; 4 (03) 529-535
  • 36 Blom JW, Osanto S, Rosendaal FR. The risk of a venous thrombotic event in lung cancer patients: higher risk for adenocarcinoma than squamous cell carcinoma. J Thromb Haemost 2004; 2 (10) 1760-1765
  • 37 Ahlbrecht J, Dickmann B, Ay C. et al. Tumor grade is associated with venous thromboembolism in patients with cancer: results from the Vienna Cancer and Thrombosis Study. J Clin Oncol 2012; 30 (31) 3870-3875
  • 38 Gran OV, Smith EN, Brækkan SK. et al. Joint effects of cancer and variants in the factor 5 gene on the risk of venous thromboembolism. Haematologica 2016; 101 (09) 1046-1053
  • 39 Kovac M, Kovac Z, Tomasevic Z. et al. Factor V Leiden mutation and high FVIII are associated with an increased risk of VTE in women with breast cancer during adjuvant tamoxifen - results from a prospective, single center, case control study. Eur J Intern Med 2015; 26 (01) 63-67
  • 40 Moore RA, Adel N, Riedel E. et al. High incidence of thromboembolic events in patients treated with cisplatin-based chemotherapy: a large retrospective analysis. J Clin Oncol 2011; 29 (25) 3466-3473
  • 41 Nalluri SR, Chu D, Keresztes R, Zhu X, Wu S. Risk of venous thromboembolism with the angiogenesis inhibitor bevacizumab in cancer patients: a meta-analysis. JAMA 2008; 300 (19) 2277-2285
  • 42 Rajkumar SV, Blood E, Vesole D, Fonseca R, Greipp PR. Eastern Cooperative Oncology Group. Phase III clinical trial of thalidomide plus dexamethasone compared with dexamethasone alone in newly diagnosed multiple myeloma: a clinical trial coordinated by the Eastern Cooperative Oncology Group. J Clin Oncol 2006; 24 (03) 431-436
  • 43 Douros A, Jobski K, Kollhorst B, Schink T, Garbe E. Risk of venous thromboembolism in cancer patients treated with epoetins or blood transfusions. Br J Clin Pharmacol 2016; 82 (03) 839-848
  • 44 Khorana AA, Francis CW, Blumberg N, Culakova E, Refaai MA, Lyman GH. Blood transfusions, thrombosis, and mortality in hospitalized patients with cancer. Arch Intern Med 2008; 168 (21) 2377-2381
  • 45 Kakkar VV, Howe CT, Nicolaides AN, Renney JT, Clarke MB. Deep vein thrombosis of the leg. Is there a “high risk” group?. Am J Surg 1970; 120 (04) 527-530
  • 46 Agnelli G, Bolis G, Capussotti L. et al. A clinical outcome-based prospective study on venous thromboembolism after cancer surgery: the @RISTOS project. Ann Surg 2006; 243 (01) 89-95
  • 47 Lee AYY, Levine MN, Butler G. et al. Incidence, risk factors, and outcomes of catheter-related thrombosis in adult patients with cancer. J Clin Oncol 2006; 24 (09) 1404-1408
  • 48 Simanek R, Vormittag R, Ay C. et al. High platelet count associated with venous thromboembolism in cancer patients: results from the Vienna Cancer and Thrombosis Study (CATS). J Thromb Haemost 2010; 8 (01) 114-120
  • 49 Blix K, Jensvoll H, Brækkan SK, Hansen JB. White blood cell count measured prior to cancer development is associated with future risk of venous thromboembolism--the Tromsø study. PLoS One 2013; 8 (09) e73447
  • 50 Ay C, Vormittag R, Dunkler D. et al. D-dimer and prothrombin fragment 1 + 2 predict venous thromboembolism in patients with cancer: results from the Vienna Cancer and Thrombosis Study. J Clin Oncol 2009; 27 (25) 4124-4129
  • 51 Chen M, Geng JG. P-selectin mediates adhesion of leukocytes, platelets, and cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Arch Immunol Ther Exp (Warsz) 2006; 54 (02) 75-84
  • 52 Stender MT, Frøkjaer JB, Larsen TB, Lundbye-Christensen S, Thorlacius-Ussing O. Preoperative plasma D-dimer is a predictor of postoperative deep venous thrombosis in colorectal cancer patients: a clinical, prospective cohort study with one-year follow-up. Dis Colon Rectum 2009; 52 (03) 446-451
  • 53 Kanz R, Vukovich T, Vormittag R. et al. Thrombosis risk and survival in cancer patients with elevated C-reactive protein. J Thromb Haemost 2011; 9 (01) 57-63
  • 54 Stegenga ME, van der Crabben SN, Blümer RM. et al. Hyperglycemia enhances coagulation and reduces neutrophil degranulation, whereas hyperinsulinemia inhibits fibrinolysis during human endotoxemia. Blood 2008; 112 (01) 82-89
  • 55 Ferroni P, Roselli M, Riondino S, Guadagni F. Predictive value of HDL cholesterol for cancer-associated venous thromboembolism during chemotherapy. J Thromb Haemost 2014; 12 (12) 2049-2053
  • 56 Khemasuwan D, Divietro ML, Tangdhanakanond K, Pomerantz SC, Eiger G. Statins decrease the occurrence of venous thromboembolism in patients with cancer. Am J Med 2010; 123 (01) 60-65
  • 57 Prandoni P, Lensing AWA, Piccioli A. et al. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002; 100 (10) 3484-3488
  • 58 Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood 2008; 111 (10) 4902-4907
  • 59 Mansfield AS, Tafur AJ, Wang CE, Kourelis TV, Wysokinska EM, Yang P. Predictors of active cancer thromboembolic outcomes: validation of the Khorana score among patients with lung cancer. J Thromb Haemost 2016; 14 (09) 1773-1778
  • 60 Noble S, Alikhan R, Robbins A, Macbeth F, Hood K. Predictors of active cancer thromboembolic outcomes: validation of the Khorana score among patients with lung cancer: comment. J Thromb Haemost 2017; 15 (03) 590-591
  • 61 van Es N, Franke VF, Middeldorp S, Wilmink JW, Büller HR. The Khorana score for the prediction of venous thromboembolism in patients with pancreatic cancer. Thromb Res 2017; 150: 30-32
  • 62 Ay C, Dunkler D, Marosi C. et al. Prediction of venous thromboembolism in cancer patients. Blood 2010; 116 (24) 5377-5382
  • 63 Gerotziafas GT, Taher A, Abdel-Razeq H. et al; COMPASS–CAT Working Group. A predictive score for thrombosis associated with breast, colorectal, lung, or ovarian cancer: the Prospective COMPASS-Cancer Associated Thrombosis Study. Oncologist 2017; 22 (10) 1222-1231
  • 64 Anand LN, Spyropoulos AC, Elredge JB. et al. External validation of the COMPASS-Cancer Associated Thrombosis Study: a predictive score to identify patients with solid tumors on treatment who are at risk of venous thromboembolism. J Clin Oncol 2019; 37 (15 suppl): e18005
  • 65 Cella CA, Di Minno G, Carlomagno C. et al. Preventing venous thromboembolism in ambulatory cancer patients: the ONKOTEV Study. Oncologist 2017; 22 (05) 601-608
  • 66 Muñoz Martín AJ, Ortega I, Font C. et al. Multivariable clinical-genetic risk model for predicting venous thromboembolic events in patients with cancer. Br J Cancer 2018; 118 (08) 1056-1061
  • 67 Riondino S, Ferroni P, Zanzotto FM, Roselli M, Guadagni F. Predicting VTE in cancer patients: candidate biomarkers and risk assessment models. Cancers (Basel) 2019; 11 (01) 95