Vous êtes sur la page 1sur 4

Thrombosis Research 135, Suppl.

1 (2015) S8S11

The mechanisms of cancer-associated thrombosis


Anna Falanga *, Marina Marchetti, Laura Russo
Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy

A R T I C L E I N F O A B S T R A C T

Keywords: Patients with cancer may display many types of hemostatic disorders that signicantly contribute to mor-
Thrombosis bidity and mortality in this disease. A complex coagulopathy develops in parallel with malignancy and is
Cancer characterized by activation of clotting mechanisms to different extent in different patients and in different
Hemorrhage types of tumor. The pathogenesis of hemostatic alterations in cancer is multifactorial; however, the tumor
Hypercoagulable state
tissue capacity to interact with and activate the host hemostatic system plays an important role. New molec-
Tissue factor
ular pathways of regulation of these properties have been recently demonstrated. Intervention strategies to
prevent and treat venous thromboembolism (VTE) in cancer patients have been addressed by large RCTs and
guidelines for VTE management have been updated.
In this review, we will present an updated overview of the complex coagulopathy associated to malignancy
and of recent advances in the thrombotic risk assessment of cancer patients.
2015 Elsevier Ltd. All rights reserved.

Introduction Tissue Factor [TF], Cancer Procoagulant, Factor VII), which contribute
to the occurrence of the overt symptomatic coagulopathy in vivo
Patients with cancer may present many types of hemostatic [2]. The shedding of procoagulant microparticles is also regulated
disorders with increased frequency of related complications. A tight by oncogenic events and further adds to the pathogenesis of the
relationship exists between malignant disease, the occurrence of cancer-associated hypercoagulable state. Finally, the changes in the
coagulation abnormalities and thrombosis. The relationship relies stromal cells of the tumor niche induced by tumor TF shade new
on the evidence that cancer induces a prothrombotic switch of lights on the hemostasis/cancer interaction.
the host hemostatic system, and vice versa, that blood clotting In addition, many clinical factors inuence the thrombotic risk
activation, positively stimulates tumor growth and dissemination. of cancer patients, who represent a high-risk category of subjects.
Abnormalities in circulating thrombotic marker levels is a common Clinical factors include general cardiovascular risk factors, cancer-
laboratory nding in these patients, also in the absence of overt specic risk factors, and anti-cancer therapies (see Table 1). Recent
thrombo-hemorragic disease, and demonstrates that activation of data conrm that the risk of developing venous thromboembolism
blood coagulation and brinolysis parallels the development and (VTE) is increased up to seven-fold in these patients as compared
spread of cancer [1]. to the general population [3]. To help clinicians in the prevention
The clinical manifestations of hemostasis deregulation may vary and management of thrombotic events in cancer patients, a number
from a subclinical asymptomatic hypercoagulable state to manifest of guidelines have been released from national and international
thrombosis of the large vessels or, further, to systemic disseminated scientic societies [4,5].
intravascular coagulation with severe bleeding. Therefore, prevent- This paper wishes to provide the most recent discoveries on the
ing these complications is clinically relevant because signicantly coagulopathy of cancer patients, the mechanisms underlying this
contribute to the morbidity and mortality of these patients. phenomenon, and nally the risk assessment to predict thrombosis
The pathogenesis of cancer-associated coagulopathy is complex in each single patient.
and involves various mechanisms. Most importantly, tumor cells
gain the capacity to activate the host hemostatic system, and this Pathogenesis of coagulation in cancer
phenomenon is driven by the same oncogenes responsible for the
neoplastic transformation. Indeed, by this process, cancer tissues The pathogenesis of the coagulation in cancer is complex and
become capable to express different procoagulant proteins (i.e involves several clinical and biological factors

Clinical factors
* Correspondence to: Anna Falanga, MD, Department of Immunohematology and
Transfusion Medicine & the Hemostasis and Thrombosis Center, Hospital Papa
Multiple clinical risk factors (i.e. patients-, cancer-, and
Giovanni XXIII, Piazza OMS, I-24127, Bergamo, Italy. Tel.: +39 035 2673663; fax:
+39 035 2674832. treatment-related) concur to blood clotting activation and to
E-mail address: annafalanga@yahoo.com (A. Falanga). thrombotic manifestations in cancer [6,7]. Regarding cancer-related

0049-3848/$ see front matter 2015 Elsevier Ltd. All rights reserved.
A. Falanga et al. / Thrombosis Research 135, Suppl. 1 (2015) S8S11 S9

features, VTE risk varies greatly depending on the primary cancer tissue procoagulant activities, much relevance is gaining the role of
site and the stage of the disease. Patients with pancreatic, stomach, the tumor-derived MP. These small membrane vescicles carry high
lung cancer, and hematologic malignancies, including lymphomas concentrations of procoagulant phosphatidylserine and TF [10].
and myeloma, have the strongest association with VTE. Additionally, In the last decade, molecular studies of experimental models
advanced, metastatic cancers are associated with an increased risk of human cancers (including hepatoma, brain and colon cancer)
of VTE compared to localized tumors. To the some guise, active have demonstrated that oncogene and repressor gene-mediated
anticancer treatments, including chemotherapy, hormonal therapy, neoplastic transformation (i.e. activation of Met, loss of PTEN,
anti-angiogenic agents, combination regimens, and surgery, sig- induction of K-ras and loss of p53) activate clotting as an integral
nicantly increase the thrombotic risk. In particular, the use of feature of neoplastic transformation. Moreover, a mutation of EGFR
systemic chemotherapy seems to increase the risk of VTE beyond to gene renders cancer cells hypersensitive to the action of coagulation
the expected from the disease itself, and is associated with up to proteins, such as TF. As a result, a microenvironment promoting
6-fold increase in VTE risk. tumor growth is generated [2]. These data conrm that a reciprocal
cancer-thrombosis connection exists, by which cancer cells support
Biological factors clot formation, and clotting proteins support cancer growth and
dissemination. In the development of human cancers, cycles of
Away from clinical factors, biological pathways likely play an microenvironmental and genetic changes are increasingly manifest.
important role in the pathogenesis of hemostatic alterations in Indolent or dormant phenotype of cells lacking tumor initiating
cancer. The principal mechanisms of thrombosis include the expres- capacity may be altered in the presence of procoagulant TF, which
sion of hemostatic proteins by tumor cells (i.e. TF), the production induces several changes in the tumor microenvironment, capable to
by tumor and/or host cells of microparticles (MP), inammatory affect the fate of these cells from a dormant to a fully malignant
cytokines (i.e. TNF-, IL-1), and proangiogenic factors (VEGF, phenotype [11].
bFGF), and the tumor cell expression of adhesion molecules to Finally, much relevant to the occurrence of the prothrombotic
bind platelets, endothelial cells and leucocytes (Fig. 1). The same state of cancer is the contribution of host endothelial cells, platelets
properties also contribute to tumor progression [8]. TF, constitu- and leukocytes. Recent data support the role of platelets in the
tively expressed on the malignant cell surface, plays a fundamental maintenance of the vascular integrity within tumors, acting as
role in thrombin generation in cancer, but also contributes to guardian of tumor vasculature; therefore platelets may be a target
tumor progression by directly inducing VEGF expression by both for the specic destabilization of tumor vessels [12]. Extensive
malignant and host vascular cells. This property regulates tumor experimental evidence also shows the presence of large quantities
neo-vascularization and provides an important link between coagu- of neutrophils in the tumor where they release extracellular DNA
lation, inammation, thrombosis and tumor growth and metastasis traps (NETs) and affect tumor growth and neoangiogenesis [13].
[8]. Another emerging mechanism of tumor-promoted clotting ac-
tivation is the heparanase activity, which associated with a high Assessment of the thrombotic risk in cancer patients
metastatic potential of tumor cells. Heparanase, besides degrading
the extracellular matrix, upregulates the expression of TF by inter- The role of most of clinical risk factors (Table 1) in cancer-
acting with the tissue factor pathway inhibitor (TFPI) on the surface associated thrombosis is well known. Differently, considerable
of endothelial and tumor cells, thereby causing the dissociation of effort has been made and is still ongoing to identify the predictive
TFPI with a subsequent increase in TF activity [9]. Among tumor value of VTE events of several circulating thrombotic biomarkers.

Fig. 1. Cancer-hemostatic system interactions. Tumor cells can activate the hemostatic system in multiple ways. Tumor cells release procoagulant activities, and
microparticles (MP), by which activate the coagulation cascade. Tumor cells also activate the host haemostatic cells (endothelial cells, leukocytes, and platelets), by either
release of soluble factors or by direct adhesion contact, thus eliciting the expression of a procoagulant phenotype of these cells. In addition, the neutrophils can release
neutrophil extracellular trap (NETs) and the adhesion of a large quantity of NETs to the vasculature may initiate thrombosis by providing a scaffold for platelet adhesion,
activation and thrombin generation.
S10 A. Falanga et al. / Thrombosis Research 135, Suppl. 1 (2015) S8S11

Table 1 Conclusions
Thrombotic risk factors in cancer patients.

General factors Cancer-specic factors Treatment-related factors Cancer patients are at high risk of developing thrombosis. The
Older age Site of cancer: brain, Surgery pathogenesis of blood coagulation activation in cancer is complex
Gender pancreas, kidney, Chemo- and and involves both clinical and biological factors. The thrombotic
Immobility stomach, lung, bladder, hormone-therapy risk varies according to the type and stage of malignancy, and is
Previous VTE gynecologic, hematologic Anti-angiogenic therapy increased by concomitant patient-related general thrombotic risk
Infections malignancies Erythropoiesis
Stage of cancer:
factors and anti-cancer therapies. However, the tumor cell-specic
Hypertension stimulating agents
Hyperlipidemia advanced stage Blood transfusions prothrombotic properties and the host cell inammatory response
Initial period of Central venous lines dramatically contribute to thrombosis in these patients. New path-
diagnosis ways of regulation of these properties have been recently identied.
This may lead to the denition of target-specic treatments to hit
both cancer and thrombosis. For the time being, the development
These include thrombinantithrombin complex [TAT], prothrombin of risk assessment models to predict thrombosis in cancer is impor-
fragment F1+2 [F1+2], brinopeptide A and B, plasminogen activator tant for identifying high-risk patients and predispose the adequate
inhibitor 1, D-dimer, and others [14]. Data from the Vienna preventive measures.
Cancer and Thrombosis (CAT) prospective study show a signicant
association with increased VTE risk of elevated P-selectin [15], or Acknowledgements
elevated D-dimer plus F1+2 levels [16]. High MP concentration
have been found to be increased in both solid and hematological The authors wish to thank Associazione Italiana per la Ricerca
malignancies and have been utilized to select patients to entry sul Cancro (A.I.R.C., grants IG10558 and 5 per mille 12237) for its
into a randomized clinical trial of thromboprophylaxis during support.
chemotherapy. The results of this small, but very original trial are
promising [17]. In this context, the thrombin generation assay, a Conict of interest statement
global test of coagulation activation, also is under evaluation to
predict VTE in cancer [18]. The authors have no conicts of interest to declare
The interaction and relative effects of the several risk factors
associated with VTE in cancer patients is highly complex, making References
the pretreatment risk assessment dicult. The updated ASCO
VTE Guidelines Panel recommends against the use of a single [1] Falanga A, Russo L. Epidemiology, risk and outcomes of venous thromboem-
risk factor or a single biomarkers to identify high-risk patients, bolism in cancer. Hamostaseologie 2012;32(2):11525.
[2] Magnus N, DAsti E, Meehan B, Garnier D, Rak J. Oncogenes and the
and instead recommends the use of a Risk Score models that
coagulation system forces that modulate dormant and aggressive states in
incorporate multiple variables to identify the high-risk patients [5]. cancer. Thromb Res 2014;133(Suppl 2):S19.
The combination of some of the clinical factors with laboratory [3] Timp JF, et al. Epidemiology of cancer-associated venous thrombosis. Blood
biomarkers allowed the development of the Khorana VTE risk 2013;122(10):171223.
assessment model (RAM) specic for cancer patients undergoing [4] Farge D, et al. International clinical practice guidelines for the treatment and
prophylaxis of venous thromboembolism in patients with cancer. J Thromb
chemotherapy [19]. This RAM is based on 5 predictive variables,
Haemost 2013;11(1):5670.
including cancer site, platelet count, hemoglobin level or the use of [5] Lyman GH, et al. Venous thromboembolism prophylaxis and treatment in
erythropoiesis-stimulating agents, leukocyte count, and body mass patients with cancer: American Society of Clinical Oncology clinical practice
index [19]. Subsequently, the Vienna CAT Study group proposed guideline update. J Clin Oncol 2013;31(17):2189204.
an expansion of this score by including two additional biomarkers [6] Falanga A, Russo L, Verzeroli C, Mechanisms of thrombosis in cancer. Thromb
(i.e. P-selectin and D-Dimer), which considerably improved the Res 2013;131(Suppl 1):S5962.
[7] Khorana AA, McCrae KR. Risk stratication strategies for cancer-associated
predictive value of VTE in the study population [14]. More recently,
thrombosis: an update. Tromb Res 2014;133(S2):S358.
another modi?ed Khoranas RAM, the Protecht score, was designed [8] Falanga A, Marchetti M, Vignoli A. Coagulation and cancer: biological and
by adding platinum- or gemcitabine-based chemotherapy to the 5 clinical aspects. J Thromb Haemost 2013;11(2):22333.
original predictive variables to increase the high-risk cancer patients [9] Nadir Y, Brenner B. Heparanase multiple effects in cancer. Thromb Res
identication [20]. In addition, in the setting of patients with 2014;133(Suppl 2):S904.
[10] Falanga A, Tartari CJ, Marchetti M. Microparticles in tumor progression.
multiple myeloma, Palumbo et al. published a RAM based on expert
Thromb Res 2012;129(Suppl 1):S1326.
recommendations, specic for the denition of patients at risk [11] Magnus N, et al. Tissue factor expression provokes escape from tumor
for thalidomide or lenalidomide treatment-associated thrombosis dormancy and leads to genomic alterations. Proc Natl Acad Sci U S A
[21]. Finally, the Ottawa Score is capable to distinguish between 2014;111(9):35449.
cancer patients with thrombosis at high or low risk of recurrent [12] Stegner D, Dutting S, Nieswandt B. Mechanistic explanation for platelet
contribution to cancer metastasis. Thromb Res 2014;133(Suppl 2):S14957.
VTE [22,23]. The ASCO panel recommends the use of RAMs to
[13] Demers M, Wagner DD. Neutrophil extracellular traps: A new link to cancer-
periodically assess the thrombotic risk of cancer patients during the associated thrombosis and potential implications for tumor progression.
different disease phases. Oncoimmunology, 2013;2(2):e22946.
The search for thrombotic markers capable to predict survival [14] Pabinger I, Thaler J, Ay C. Biomarkers for prediction of venous thromboem-
in cancer patients is currently active. Results of a large cohort bolism in cancer. Blood 2013;122(12):20118.
[15] Ay C, et al. High plasma levels of soluble P-selectin are predictive of venous
of individuals free of clinically evident cardiovascular and cancer
thromboembolism in cancer patients: results from the Vienna Cancer and
disease (MOLI-SANI project) show that elevated plasma D-dimer Thrombosis Study (CATS). Blood 2008;112(7):27038.
levels are independently associated with increased risk of death for [16] Ay C, et al. D-dimer and prothrombin fragment 1 + 2 predict venous
any cause [24]. Furthermore, in patients with ovarian masses, the thromboembolism in patients with cancer: results from the Vienna Cancer
preoperative D-dimer levels, alone or in combination with CA-125, and Thrombosis Study. J Clin Oncol 2009;27(25):41249.
[17] Zwicker JI, et al. Prediction and prevention of thromboembolic events with
signicantly predict for malignant versus benign ovarian lesions
enoxaparin in cancer patients with elevated tissue factor-bearing micropar-
[25]. ticles: a randomized-controlled phase II trial (the Microtec study). Br J
Haematol 2013;160(4):5307.
[18] Ay C, et al. Prediction of venous thromboembolism in patients with cancer
A. Falanga et al. / Thrombosis Research 135, Suppl. 1 (2015) S8S11 S11

by measuring thrombin generation: results from the Vienna Cancer and [23] den Exter PL, Kooiman J, Huisman MV. Validation of the Ottawa prognostic
Thrombosis Study. J Clin Oncol 2011;29(15):2099103. score for the prediction of recurrent venous thromboembolism in patients
[19] Khorana AA, et al. Development and validation of a predictive model for with cancer-associated thrombosis. J Thromb Haemost 2013;11(5):9981000.
chemotherapyassociated thrombosis. Blood 2008;111(10):49027. [24] Di Castelnuovo A, et al. Association of D-dimer levels with all-cause mor-
[20] Verso M, et al. A modied Khorana risk assessment score for venous tality in a healthy adult population: ndings from the MOLI-SANI study.
thromboembolism in cancer patients receiving chemotherapy: the Protecht Haematologica 2013;98(9):147680.
score. Intern Emerg Med 2012;7(3):2912. [25] Amirkhosravi A, et al. Blood clotting activation analysis for preoperative
[21] Palumbo A, et al. Prevention of thalidomide- and lenalidomide-associated differentiation of benign versus malignant ovarian masses. Blood Coagul
thrombosis in myeloma. Leukemia 2008;22(2):41423. Fibrinolysis 2013;24(5):5107.
[22] Louzada ML, et al., Development of a clinical prediction rule for risk stratica-
tion of recurrent venous thromboembolism in patients with cancer-associated
venous thromboembolism. Circulation 2012;126(4):44854.

Vous aimerez peut-être aussi