Semin Thromb Hemost 2019; 45(04): 334-341
DOI: 10.1055/s-0039-1688493
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Venous Thromboembolism in Brain Tumors: Risk Factors, Molecular Mechanisms, and Clinical Challenges

Julia Riedl
1   Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
,
Cihan Ay
1   Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
2   I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
› Author Affiliations
Further Information

Publication History

Publication Date:
30 April 2019 (online)

Abstract

Venous thromboembolism (VTE) is a common complication in patients with primary brain tumors, with up to 20% of patients per year having a VTE event. Clinical risk factors for VTE include glioblastoma subtype, paresis, or surgery. Furthermore, specific factors playing a role in tumor biology were recently identified to predispose to prothrombotic risk. For instance, mutations in the isocitrate dehydrogenase 1 (IDH1) gene, which occurs in a subgroup of glioma, correlate with risk of VTE, with low incidence in patients with presence of an IDH1 mutation compared with those with IDH1 wild-type status. In addition, expression of the glycoprotein podoplanin on brain tumors was associated with both intratumoral thrombi and high risk of VTE. As podoplanin has the ability to activate platelets, a mechanistic role of podoplanin-mediated platelet activation in VTE development has been suggested. From a clinical point of view, the management of patients with primary brain tumors and VTE is challenging. Anticoagulation is required to treat patients; however, it is associated with increased risk of intracranial hemorrhage. This review focuses on describing the epidemiology, risk factors, and mechanisms of brain tumor-associated thrombosis and discusses clinical challenges in the prevention and treatment of VTE in patients with brain tumors.

 
  • References

  • 1 Heit JA, Silverstein MD, Mohr DN, Petterson TM, O'Fallon WM, Melton III LJ. Risk factors for deep vein thrombosis and pulmonary embolism: a population-based case-control study. Arch Intern Med 2000; 160 (06) 809-815
  • 2 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
  • 3 Marras LC, Geerts WH, Perry JR. The risk of venous thromboembolism is increased throughout the course of malignant glioma: an evidence-based review. Cancer 2000; 89 (03) 640-646
  • 4 Riedl J, Preusser M, Nazari PM. , et al. Podoplanin expression in primary brain tumors induces platelet aggregation and increases risk of venous thromboembolism. Blood 2017; 129 (13) 1831-1839
  • 5 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
  • 6 Simanek R, Vormittag R, Hassler M. , et al. Venous thromboembolism and survival in patients with high-grade glioma. Neuro-oncol 2007; 9 (02) 89-95
  • 7 Smith TR, Lall RR, Graham RB. , et al. Venous thromboembolism in high grade glioma among surgical patients: results from a single center over a 10 year period. J Neurooncol 2014; 120 (02) 347-352
  • 8 Semrad TJ, O'Donnell R, Wun T. , et al. Epidemiology of venous thromboembolism in 9489 patients with malignant glioma. J Neurosurg 2007; 106 (04) 601-608
  • 9 Cote DJ, Dawood HY, Smith TR. Venous thromboembolism in patients with high-grade glioma. Semin Thromb Hemost 2016; 42 (08) 877-883
  • 10 Jo JT, Schiff D, Perry JR. Thrombosis in brain tumors. Semin Thromb Hemost 2014; 40 (03) 325-331
  • 11 Streiff MB, Segal J, Grossman SA, Kickler TS, Weir EG. ABO blood group is a potent risk factor for venous thromboembolism in patients with malignant gliomas. Cancer 2004; 100 (08) 1717-1723
  • 12 Brandes AA, Scelzi E, Salmistraro G. , et al. Incidence of risk of thromboembolism during treatment high-grade gliomas: a prospective study. Eur J Cancer 1997; 33 (10) 1592-1596
  • 13 Yust-Katz S, Mandel JJ, Wu J. , et al. Venous thromboembolism (VTE) and glioblastoma. J Neurooncol 2015; 124 (01) 87-94
  • 14 O'Donnell J, Laffan MA. The relationship between ABO histo-blood group, factor VIII and von Willebrand factor. Transfus Med 2001; 11 (04) 343-351
  • 15 Vormittag R, Simanek R, Ay C. , et al. High factor VIII levels independently predict venous thromboembolism in cancer patients: the cancer and thrombosis study. Arterioscler Thromb Vasc Biol 2009; 29 (12) 2176-2181
  • 16 Pépin M, Kleinjan A, Hajage D. , et al. ADAMTS-13 and von Willebrand factor predict venous thromboembolism in patients with cancer. J Thromb Haemost 2016; 14 (02) 306-315
  • 17 Streiff MB, Ye X, Kickler TS. , et al. A prospective multicenter study of venous thromboembolism in patients with newly-diagnosed high-grade glioma: hazard rate and risk factors. J Neurooncol 2015; 124 (02) 299-305
  • 18 Rodas RA, Fenstermaker RA, McKeever PE. , et al. Correlation of intraluminal thrombosis in brain tumor vessels with postoperative thrombotic complications: a preliminary report. J Neurosurg 1998; 89 (02) 200-205
  • 19 Unruh D, Schwarze SR, Khoury L. , et al. Mutant IDH1 and thrombosis in gliomas. Acta Neuropathol 2016; 132 (06) 917-930
  • 20 Mir Seyed Nazari P, Riedl J, Preusser M. , et al. Combination of isocitrate dehydrogenase 1 (IDH1) mutation and podoplanin expression in brain tumors identifies patients at high or low risk of venous thromboembolism. J Thromb Haemost 2018; 16 (06) 1121-1127
  • 21 Senders JT, Goldhaber NH, Cote DJ. , et al. Venous thromboembolism and intracranial hemorrhage after craniotomy for primary malignant brain tumors: a National Surgical Quality Improvement Program analysis. J Neurooncol 2018; 136 (01) 135-145
  • 22 Perry JR. Anticoagulation of malignant glioma patients in the era of novel antiangiogenic agents. Curr Opin Neurol 2010; 23 (06) 592-596
  • 23 Li X, Huang R, Xu Z. Risk of adverse vascular events in newly diagnosed glioblastoma multiforme patients treated with bevacizumab: a systematic review and meta-analysis. Sci Rep 2015; 5: 14698
  • 24 D'Asti E, Rak J. Biological basis of personalized anticoagulation in cancer: oncogene and oncomir networks as putative regulators of coagulopathy. Thromb Res 2016; 140 (Suppl. 01) S37-S43
  • 25 Tawil N, Chennakrishnaiah S, Bassawon R, Johnson R, D'Asti E, Rak J. Single cell coagulomes as constituents of the oncogene-driven coagulant phenotype in brain tumours. Thromb Res 2018; 164 (Suppl. 01) S136-S142
  • 26 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
  • 27 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
  • 28 Thaler J, Ay C, Kaider A. , et al. Biomarkers predictive of venous thromboembolism in patients with newly diagnosed high-grade gliomas. Neuro-oncol 2014; 16 (12) 1645-1651
  • 29 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
  • 30 Khorana AA, Francis CW, Culakova E, Lyman GH. Risk factors for chemotherapy-associated venous thromboembolism in a prospective observational study. Cancer 2005; 104 (12) 2822-2829
  • 31 Suzuki-Inoue K, Kato Y, Inoue O. , et al. Involvement of the snake toxin receptor CLEC-2, in podoplanin-mediated platelet activation, by cancer cells. J Biol Chem 2007; 282 (36) 25993-26001
  • 32 Suzuki-Inoue K, Tsukiji N, Shirai T, Osada M, Inoue O, Ozaki Y. Platelet CLEC-2: roles beyond hemostasis. Semin Thromb Hemost 2018; 44 (02) 126-134
  • 33 Uhrin P, Zaujec J, Breuss JM. , et al. Novel function for blood platelets and podoplanin in developmental separation of blood and lymphatic circulation. Blood 2010; 115 (19) 3997-4005
  • 34 Herzog BH, Fu J, Wilson SJ. , et al. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature 2013; 502 (7469): 105-109
  • 35 Birner P, Pusch S, Christov C. , et al. Mutant IDH1 inhibits PI3K/Akt signaling in human glioma. Cancer 2014; 120 (16) 2440-2447
  • 36 Suzuki-Inoue K, Inoue O, Ozaki Y. Novel platelet activation receptor CLEC-2: from discovery to prospects. J Thromb Haemost 2011; 9 (Suppl. 01) 44-55
  • 37 Suzuki-Inoue K, Fuller GL, García A. , et al. A novel Syk-dependent mechanism of platelet activation by the C-type lectin receptor CLEC-2. Blood 2006; 107 (02) 542-549
  • 38 Tsukiji N, Inoue O, Morimoto M. , et al. Platelets play an essential role in murine lung development through Clec-2/podoplanin interaction. Blood 2018; 132 (11) 1167-1179
  • 39 Lowe KL, Finney BA, Deppermann C. , et al. Podoplanin and CLEC-2 drive cerebrovascular patterning and integrity during development. Blood 2015; 125 (24) 3769-3777
  • 40 Rayes J, Watson SP, Nieswandt B. Functional significance of the platelet immune receptors GPVI and CLEC-2. J Clin Invest 2019; 129 (01) 12-23
  • 41 Suzuki-Inoue K, Inoue O, Ding G. , et al. Essential in vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2-deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets. J Biol Chem 2010; 285 (32) 24494-24507
  • 42 Hughes CE, Navarro-Núñez L, Finney BA, Mourão-Sá D, Pollitt AY, Watson SP. CLEC-2 is not required for platelet aggregation at arteriolar shear. J Thromb Haemost 2010; 8 (10) 2328-2332
  • 43 Shirai T, Inoue O, Tamura S. , et al. C-type lectin-like receptor 2 promotes hematogenous tumor metastasis and prothrombotic state in tumor-bearing mice. J Thromb Haemost 2017; 15 (03) 513-525
  • 44 Hitchcock JR, Cook CN, Bobat S. , et al. Inflammation drives thrombosis after Salmonella infection via CLEC-2 on platelets. J Clin Invest 2015; 125 (12) 4429-4446
  • 45 Payne H, Ponomaryov T, Watson SP, Brill A. Mice with a deficiency in CLEC-2 are protected against deep vein thrombosis. Blood 2017; 129 (14) 2013-2020
  • 46 Lavallée V-P, Chagraoui J, MacRae T. , et al. Transcriptomic landscape of acute promyelocytic leukemia reveals aberrant surface expression of the platelet aggregation agonist Podoplanin. Leukemia 2018; 32 (06) 1349-1357
  • 47 Cueni LN, Chen L, Zhang H. , et al. Podoplanin-Fc reduces lymphatic vessel formation in vitro and in vivo and causes disseminated intravascular coagulation when transgenically expressed in the skin. Blood 2010; 116 (20) 4376-4384
  • 48 Ernst A, Hofmann S, Ahmadi R. , et al. Genomic and expression profiling of glioblastoma stem cell-like spheroid cultures identifies novel tumor-relevant genes associated with survival. Clin Cancer Res 2009; 15 (21) 6541-6550
  • 49 Krishnan H, Rayes J, Miyashita T. , et al. Podoplanin: An emerging cancer biomarker and therapeutic target. Cancer Sci 2018; 109 (05) 1292-1299
  • 50 Shiina S, Ohno M, Ohka F. , et al. CAR T cells targeting podoplanin reduce orthotopic glioblastomas in mouse brains. Cancer Immunol Res 2016; 4 (03) 259-268
  • 51 Eisemann T, Costa B, Harter PN. , et al. Podoplanin expression is a prognostic biomarker but may be dispensable for the malignancy of glioblastoma. Neuro Oncol 2019; 21 (03) 326-336
  • 52 Lyman GH, Bohlke K, Khorana AA. , et al; American Society of Clinical Oncology. Venous thromboembolism prophylaxis and treatment in patients with cancer: american society of clinical oncology clinical practice guideline update 2014. J Clin Oncol 2015; 33 (06) 654-656
  • 53 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
  • 54 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
  • 55 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
  • 56 Al Megren M, De Wit C, Al Qahtani M, Le Gal G, Carrier M. Management of venous thromboembolism in patients with glioma. Thromb Res 2017; 156: 105-108
  • 57 Zwicker JI, Karp Leaf R, Carrier M. A meta-analysis of intracranial hemorrhage in patients with brain tumors receiving therapeutic anticoagulation. J Thromb Haemost 2016; 14 (09) 1736-1740
  • 58 Perry JR, Julian JA, Laperriere NJ. , et al. PRODIGE: a randomized placebo-controlled trial of dalteparin low-molecular-weight heparin thromboprophylaxis in patients with newly diagnosed malignant glioma. J Thromb Haemost 2010; 8 (09) 1959-1965
  • 59 Le Rhun E, Genbrugge E, Stupp R. , et al. Associations of anticoagulant use with outcome in newly diagnosed glioblastoma. Eur J Cancer 2018; 101 (101) 95-104