Semin Thromb Hemost 2021; 47(02): 139-149
DOI: 10.1055/s-0041-1722863
Review Article

Advances in the Management of Cancer-Associated Thrombosis

Sukhraj Pal Singh Dhami
1   Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
,
Sean Patmore
1   Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
,
Jamie M. O'Sullivan
1   Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
› Author Affiliations

Abstract

The association between cancer and venous thromboembolism (VTE) has been established for more than 150 years. Nevertheless, cancer-associated thrombosis still remains a major clinical challenge and is associated with significant morbidity and mortality for patients with cancer. The clinical presentation of cancer-associated thrombosis can be distinct from that of a patient without an underlying malignancy. Moreover, specific cancer types, including pancreatic cancer and hematological malignancies, as well as advanced stage disease can confer a significant thrombotic risk. This risk is further augmented by specific anticancer treatment modalities. The pathophysiology of cancer-associated thrombosis is complex and multifactorial. However, understanding the biological mechanisms underpinning VTE risk may provide insight into novel targeted prophylaxis in cancer patients. Over the last decade, low-molecular-weight heparin has been the preferred anticoagulant agent for patients with cancer-associated thrombosis due to improved efficacy compared with vitamin K antagonists. However, the advent of direct oral anticoagulants (DOACs) has added to the repertoire of ammunition now at the disposal of clinicians to aid in the management of cancer-associated thrombosis. Several randomized controlled trials have now been published, demonstrating DOAC as a noninferior alternative for both the treatment and prevention of cancer-associated thrombosis. Notwithstanding this, limitations for their widespread use remain, with the potential for increased bleeding risk, drug interactions, and poor DOAC metabolism. This review discusses the evidence base for the incidence and risk factors associated with VTE in cancer, development, and refinement of risk prediction models and novel advances in the therapeutic management of cancer-associated thrombosis.

Authors' Contributions

All authors were involved in the writing and review processes of the manuscript.




Publication History

Article published online:
26 February 2021

© 2021. Thieme. All rights reserved.

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

 
  • References

  • 1 Silverstein MD, Heit JA, Mohr DN, Petterson TM, O'Fallon WM, Melton III LJ. Trends in the incidence of deep vein thrombosis and pulmonary embolism: a 25-year population-based study. Arch Intern Med 1998; 158 (06) 585-593
  • 2 Blom JW, Doggen CJM, Osanto S, Rosendaal FR. Malignancies, prothrombotic mutations, and the risk of venous thrombosis. JAMA 2005; 293 (06) 715-722
  • 3 Gussoni G, Frasson S, La Regina M, Di Micco P, Monreal M. RIETE Investigators. Three-month mortality rate and clinical predictors in patients with venous thromboembolism and cancer. Findings from the RIETE registry. Thromb Res 2013; 131 (01) 24-30
  • 4 Timp JF, Braekkan SK, Versteeg HH, Cannegieter SC. Epidemiology of cancer-associated venous thrombosis. Blood 2013; 122 (10) 1712-1723
  • 5 Lillicrap D. Introduction to a series of reviews on cancer-associated thrombotic disease. Blood 2013; 122 (10) 1687-1688
  • 6 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
  • 7 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
  • 8 Blom JW, Vanderschoot JPM, 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
  • 9 Horsted F, West J, Grainge MJ. Risk of venous thromboembolism in patients with cancer: a systematic review and meta-analysis. PLoS Med 2012; 9 (07) e1001275
  • 10 Sørensen HT, Mellemkjaer L, Olsen JH, Baron JA. Prognosis of cancers associated with venous thromboembolism. N Engl J Med 2000; 343 (25) 1846-1850
  • 11 Amer MH. Cancer-associated thrombosis: clinical presentation and survival. Cancer Manag Res 2013; 5: 165-178
  • 12 Grilz E, Königsbrügge O, Posch F. et al. Frequency, risk factors, and impact on mortality of arterial thromboembolism in patients with cancer. Haematologica 2018; 103 (09) 1549-1556
  • 13 Canale ML, Bisceglia I, Lestuzzi C, Parrini I. ANMCO Cardio-Oncology Task Force. Arterial thrombosis in cancer: spotlight on the neglected vessels. Anticancer Res 2019; 39 (09) 4619-4625
  • 14 Seinturier C, Bosson JL, Colonna M, Imbert B, Carpentier PH. Site and clinical outcome of deep vein thrombosis of the lower limbs: an epidemiological study. J Thromb Haemost 2005; 3 (07) 1362-1367
  • 15 Joffe HV, Goldhaber SZ. Upper-extremity deep vein thrombosis. Circulation 2002; 106 (14) 1874-1880
  • 16 Riva N, Ageno W. Cerebral and splanchnic vein thrombosis: advances, challenges, and unanswered questions. J Clin Med 2020; 9 (03) 743
  • 17 Martinelli I, De Stefano V. Rare thromboses of cerebral, splanchnic and upper-extremity veins. A narrative review. Thromb Haemost 2010; 103 (06) 1136-1144
  • 18 Dentali F, Squizzato A, Brivio L. et al. JAK2V617F mutation for the early diagnosis of Ph- myeloproliferative neoplasms in patients with venous thromboembolism: a meta-analysis. Blood 2009; 113 (22) 5617-5623
  • 19 O'Donnell JS, O'Sullivan JM, Preston RJS. Advances in understanding the molecular mechanisms that maintain normal haemostasis. Br J Haematol 2019; 186 (01) 24-36
  • 20 Han X, Guo B, Li Y, Zhu B. Tissue factor in tumor microenvironment: a systematic review. J Hematol Oncol 2014; 7 (01) 54
  • 21 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
  • 22 Khorana AA, Ahrendt SA, Ryan CK. et al. Tissue factor expression, angiogenesis, and thrombosis in pancreatic cancer. Clin Cancer Res 2007; 13 (10) 2870-2875
  • 23 Palumbo JS, Talmage KE, Massari JV. et al. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood 2005; 105 (01) 178-185
  • 24 Owens III AP, Mackman N. Microparticles in hemostasis and thrombosis. Circ Res 2011; 108 (10) 1284-1297
  • 25 Wang JG, Geddings JE, Aleman MM. et al. Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer. Blood 2012; 119 (23) 5543-5552
  • 26 Hron G, Kollars M, Weber H. et al. Tissue factor-positive microparticles: cellular origin and association with coagulation activation in patients with colorectal cancer. Thromb Haemost 2007; 97 (01) 119-123
  • 27 Bharthuar A, Khorana AA, Hutson A. et al. Association of elevated tissue factor (TF) with survival and thromboembolism (TE) in pancreaticobiliary cancers (PBC). J Clin Oncol 2010; 28 (15) 4126
  • 28 Tesselaar MET, Romijn FPHTM, van der Linden IK, Bertina RM, Osanto S. Microparticle-associated tissue factor activity in cancer patients with and without thrombosis. J Thromb Haemost 2009; 7 (08) 1421-1423
  • 29 Haubold K, Rink M, Spath B. et al. Tissue factor procoagulant activity of plasma microparticles is increased in patients with early-stage prostate cancer. Thromb Haemost 2009; 101 (06) 1147-1155
  • 30 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
  • 31 Thomas GM, Panicot-Dubois L, Lacroix R, Dignat-George F, Lombardo D, Dubois C. Cancer cell-derived microparticles bearing P-selectin glycoprotein ligand 1 accelerate thrombus formation in vivo. J Exp Med 2009; 206 (09) 1913-1927
  • 32 Zhao L, Bi Y, Kou J, Shi J, Piao D. Phosphatidylserine exposing-platelets and microparticles promote procoagulant activity in colon cancer patients. J Exp Clin Cancer Res 2016; 35 (01) 54
  • 33 Palacios-Acedo AL, Mège D, Crescence L, Dignat-George F, Dubois C, Panicot-Dubois L. Platelets, thrombo-inflammation, and cancer: collaborating with the enemy. Front Immunol 2019; 10: 1805
  • 34 Nieswandt B, Hafner M, Echtenacher B, Männel DN. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res 1999; 59 (06) 1295-1300
  • 35 Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci U S A 1968; 61 (01) 46-52
  • 36 Karpatkin S, Pearlstein E, Ambrogio C, Coller BS. Role of adhesive proteins in platelet tumor interaction in vitro and metastasis formation in vivo. J Clin Invest 1988; 81 (04) 1012-1019
  • 37 Plantureux L, Mège D, Crescence L, Dignat-George F, Dubois C, Panicot-Dubois L. Impacts of cancer on platelet production, activation and education and mechanisms of cancer-associated thrombosis. Cancers (Basel) 2018; 10 (11) 441
  • 38 Bauer AT, Suckau J, Frank K. et al. von Willebrand factor fibers promote cancer-associated platelet aggregation in malignant melanoma of mice and humans. Blood 2015; 125 (20) 3153-3163
  • 39 O'Sullivan JM, Preston RJS, Robson T, O'Donnell JS. Emerging roles for von Willebrand factor in cancer cell biology. Semin Thromb Hemost 2018; 44 (02) 159-166
  • 40 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
  • 41 Martinod K, Wagner DD. Thrombosis: tangled up in NETs. Blood 2014; 123 (18) 2768-2776
  • 42 Brill A, Fuchs TA, Savchenko AS. et al. Neutrophil extracellular traps promote deep vein thrombosis in mice. J Thromb Haemost 2012; 10 (01) 136-144
  • 43 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
  • 44 Mauracher L-M, Posch F, Martinod K. et al. Citrullinated histone H3, a biomarker of neutrophil extracellular trap formation, predicts the risk of venous thromboembolism in cancer patients. J Thromb Haemost 2018; 16 (03) 508-518
  • 45 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
  • 46 Park J, Wysocki RW, Amoozgar Z. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med 2016; 8 (361) 361ra138
  • 47 Nishigori N, Matsumoto M, Koyama F. et al. von Willebrand factor-rich platelet thrombi in the liver cause sinusoidal obstruction syndrome following oxaliplatin-based chemotherapy. PLoS One 2015; 10 (11) e0143136
  • 48 Wang J, Weiss I, Svoboda K, Kwaan HC. Thrombogenic role of cells undergoing apoptosis. Br J Haematol 2001; 115 (02) 382-391
  • 49 Swystun LL, Mukherjee S, Liaw PC. Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus. J Thromb Haemost 2011; 9 (11) 2313-2321
  • 50 Wood SJ, Holyoke E, Yardley J. An experimental study of the influence of adrenal steroids, growth hormone and anticoagulants on pulmonary metastasis formation in mice. Proc Am Assoc Cancer Res 1956; 2: 157
  • 51 Kreisler L. Effect of heparin on the growth of a transplantable lymphosarcoma in mice. Science 1952; 115 (2980): 145-146
  • 52 Stein PD, Beemath A, Meyers FA, Skaf E, Sanchez J, Olson RE. Incidence of venous thromboembolism in patients hospitalized with cancer. Am J Med 2006; 119 (01) 60-68
  • 53 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
  • 54 Alcalay A, Wun T, Khatri V. et al. Venous thromboembolism in patients with colorectal cancer: incidence and effect on survival. J Clin Oncol 2006; 24 (07) 1112-1118
  • 55 Rickles FR, Levine MN. Epidemiology of thrombosis in cancer. Acta Haematol 2001; 106 (1–2): 6-12
  • 56 Byrne M, Reynolds JV, O'Donnell JS. et al. Long-term activation of the pro-coagulant response after neoadjuvant chemoradiation and major cancer surgery. Br J Cancer 2010; 102 (01) 73-79
  • 57 Khorana AA, Dalal M, Lin J, Connolly GC. Incidence and predictors of venous thromboembolism (VTE) among ambulatory high-risk cancer patients undergoing chemotherapy in the United States. Cancer 2013; 119 (03) 648-655
  • 58 Haddad TC, Greeno EW. Chemotherapy-induced thrombosis. Thromb Res 2006; 118 (05) 555-568
  • 59 Deitcher SR, Gomes MPV. The risk of venous thromboembolic disease associated with adjuvant hormone therapy for breast carcinoma: a systematic review. Cancer 2004; 101 (03) 439-449
  • 60 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
  • 61 Palumbo A, Rajkumar SV, Dimopoulos MA. et al; International Myeloma Working Group. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia 2008; 22 (02) 414-423
  • 62 Mulder FI, Candeloro M, Kamphuisen PW. et al; CAT-prediction collaborators. The Khorana score for prediction of venous thromboembolism in cancer patients: a systematic review and meta-analysis. Haematologica 2019; 104 (06) 1277-1287
  • 63 Pabinger I, Thaler J, Ay C. Biomarkers for prediction of venous thromboembolism in cancer. Blood 2013; 122 (12) 2011-2018
  • 64 Fotiou D, Gavriatopoulou M, Terpos E. Multiple myeloma and thrombosis: prophylaxis and risk prediction tools. Cancers (Basel) 2020; 12 (01) 191
  • 65 Di Nisio M, Porreca E, Candeloro M, De Tursi M, Russi I, Rutjes AW. Primary prophylaxis for venous thromboembolism in ambulatory cancer patients receiving chemotherapy. Cochrane Database Syst Rev 2016; 12 (12) CD008500
  • 66 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
  • 67 Meyer G, Marjanovic Z, Valcke J. et al. Comparison of low-molecular-weight heparin and warfarin for the secondary prevention of venous thromboembolism in patients with cancer: a randomized controlled study. Arch Intern Med 2002; 162 (15) 1729-1735
  • 68 Lee AYY, Bauersachs R, Janas MS. et al; CATCH Investigators. CATCH: a randomised clinical trial comparing long-term tinzaparin versus warfarin for treatment of acute venous thromboembolism in cancer patients. BMC Cancer 2013; 13: 284
  • 69 Lee AYY, Rickles FR, Julian JA. et al. Randomized comparison of low molecular weight heparin and coumarin derivatives on the survival of patients with cancer and venous thromboembolism. J Clin Oncol 2005; 23 (10) 2123-2129
  • 70 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
  • 71 Klerk CPW, Smorenburg SM, Otten H-M. et al. The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol 2005; 23 (10) 2130-2135
  • 72 Sideras K, Schaefer PL, Okuno SH. et al. Low-molecular-weight heparin in patients with advanced cancer: a phase 3 clinical trial. Mayo Clin Proc 2006; 81 (06) 758-767
  • 73 Borsig L. Antimetastatic activities of heparins and modified heparins. Experimental evidence. Thromb Res 2010; 125 (Suppl. 02) S66-S71
  • 74 Douxfils J, Ageno W, Samama CM. et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16 (02) 209-219
  • 75 Young AM, Marshall A, Thirlwall J. et al. Comparison of an oral factor xa inhibitor with low molecular weight heparin in patients with cancer with venous thromboembolism: results of a randomized trial (SELECT-D). J Clin Oncol 2018; 36 (20) 2017-2023
  • 76 Raskob GE, van Es N, Verhamme P. et al; Hokusai VTE Cancer Investigators. Edoxaban for the treatment of cancer-associated venous thromboembolism. N Engl J Med 2018; 378 (07) 615-624
  • 77 Khorana AA, Noble S, Lee AYY. et al. Role of direct oral anticoagulants in the treatment of cancer-associated venous thromboembolism: guidance from the SSC of the ISTH. J Thromb Haemost 2018; 16 (09) 1891-1894
  • 78 McBane II RD, Wysokinski WE, Le-Rademacher JG. et al. Apixaban and dalteparin in active malignancy-associated venous thromboembolism: the ADAM VTE trial. J Thromb Haemost 2020; 18 (02) 411-421
  • 79 Agnelli G, Becattini C, Meyer G. et al; Caravaggio Investigators. Apixaban for the treatment of venous thromboembolism associated with cancer. N Engl J Med 2020; 382 (17) 1599-1607
  • 80 Spencer FA, Emery C, Lessard D. et al. The Worcester Venous Thromboembolism study: a population-based study of the clinical epidemiology of venous thromboembolism. J Gen Intern Med 2006; 21 (07) 722-727
  • 81 Carrier M, Abou-Nassar K, Mallick R. et al; AVERT Investigators. Apixaban to prevent venous thromboembolism in patients with cancer. N Engl J Med 2019; 380 (08) 711-719
  • 82 Khorana AA, Soff GA, Kakkar AK. et al; CASSINI Investigators. Rivaroxaban for thromboprophylaxis in high-risk ambulatory patients with cancer. N Engl J Med 2019; 380 (08) 720-728
  • 83 Wang TF, Zwicker JI, Ay C. et al. The use of direct oral anticoagulants for primary thromboprophylaxis in ambulatory cancer patients: guidance from the SSC of the ISTH. J Thromb Haemost 2019; 17 (10) 1772-1778
  • 84 Hull RD, Pineo GF, Brant RF. et al; LITE Trial Investigators. Long-term low-molecular-weight heparin versus usual care in proximal-vein thrombosis patients with cancer. Am J Med 2006; 119 (12) 1062-1072
  • 85 Prins MH, Lensing AWA, Brighton TA. et al. Oral rivaroxaban versus enoxaparin with vitamin K antagonist for the treatment of symptomatic venous thromboembolism in patients with cancer (EINSTEIN-DVT and EINSTEIN-PE): a pooled subgroup analysis of two randomised controlled trials. Lancet Haematol 2014; 1 (01) e37-e46
  • 86 Schulman S, Kakkar AK, Goldhaber SZ. et al; RE-COVER II Trial Investigators. Treatment of acute venous thromboembolism with dabigatran or warfarin and pooled analysis. Circulation 2014; 129 (07) 764-772