Patients with cancer have an increased risk for venous thrombosis. Interestingly, different cancer types have different rates of thrombosis, with pancreatic cancer having one of the highest rates. However, the mechanisms responsible for the increase in venous thrombosis in patients with cancer are not understood. Tissue factor (TF) is a transmembrane receptor and primary initiator of blood coagulation. Tumor cells express TF and spontaneously release TF-positive microparticles (MPs) into the blood. MPs are small membrane vesicles that are highly procoagulant. It has been proposed that these circulating tumor-derived, TF-positive MPs may explain the increased rates of venous thrombosis seen in patients with cancer. In animal models, increased levels of tumor-derived, TF-positive MPs are associated with activation of coagulation. Moreover, these MPs bind to sites of vascular injury and enhance thrombosis. We and others have found that patients with cancer have elevated levels of circulating TF-positive MPs. These MPs are derived from tumors because they express tumor markers and are decreased by tumor resection. Importantly, several studies have shown that increased levels of TF-positive MPs correlate with venous thrombosis in patients with cancer. Taken together, these results suggest that TF-positive MPs may be a useful biomarker to identify patients with cancer who are at high risk for thrombosis.

Cancer and its treatment are frequently complicated by the development of venous thromboembolism (VTE),1,2  which is a collective term for deep vein thrombosis (DVT) and pulmonary embolism (PE). The development of VTE in patients with cancer is a condition included in the spectrum of prothrombotic conditions in cancer that are encompassed by Trousseau’s syndrome.1 

Thrombosis increases the morbidity and mortality of patients with cancer.3-5  Pancreatic and brain cancer have the highest rates of VTE among all cancer types, with cumulative incidences reported at 5.3% to 26% and 1.6% to 26%, respectively.5-7  Hematologic malignancies are also known to have a high rate of VTE. Reported incidence values for VTE in certain types of lymphoma are as high as 59.5%, and for multiple myeloma they are as high as 58%, with highly prothrombotic chemotherapy regimens.8  In comparison, lung and colorectal cancers have lower rates of VTE (from 1.6% to 13.6% and from 3.1% to 10.2%, respectively), and breast and prostate cancer have the lowest rates (from 0.4% to 8.1% and from 0.5% to 1.3%, respectively).5-7  In fact, cancer type was used in the development of a risk model to predict the development of VTE in patients with cancer.9  The epidemiology and treatment of cancer-induced thrombosis is discussed in detail in 2 other reviews in this series: one by Cannegieter and colleagues,10  and another by Lee and colleagues.11  Tissue factor (TF) is a transmembrane receptor that binds plasma factor VII/VIIa and triggers blood coagulation after vascular injury and in various diseases.12-17  TF is expressed by many cancer cells, particularly in cancers of epithelial origin.18-20  Cancer cells also spontaneously release small membrane microparticles (MPs) bearing TF.21,22  An early study proposed that these MPs may explain the prothrombotic state associated with malignancy.23,24 

In this review, we summarize studies analyzing the role of TF-positive, tumor-derived MPs (TMPs) in the activation of coagulation and thrombosis in mouse models, as well as the association between TF-positive MPs and the development of VTE in patients with cancer.

Venous clots form on the surface of a largely intact vascular endothelium because of a combination of changes in the vessel wall, blood flow disturbances, and thrombogenic factors in the blood itself.2,12,25-28  The most common site for the development of venous thrombi is valve pockets.26,27,29-32  A number of different mouse models have been used to study thrombosis.33  In general, animal models of thrombosis that do not produce extensive damage to the vessel wall are better models of venous thrombosis than models that expose vessel wall TF. Studies with healthy mice have shown that leukocyte TF contributes to thrombosis in the inferior vena cava (IVC) stenosis model, whereas vessel wall TF drives thrombosis in the IVC ligation and electrolytic injury models.34-38  In small vessels, thrombosis has been shown to be dependent on TF expression by neutrophils, hematopoietic cell-derived TF-positive MPs, and vessel wall TF.36,37 

Cells release 3 types of membrane vesicles: exosomes (50-100 nM), MPs (0.1-1 µm), and apoptotic bodies (1-3 µm).39  This review focuses on TF-positive MPs.

Cells undergoing apoptosis release apoptotic bodies.40  Exosomes are released from the cell by the fusion of intracellular multivesicular bodies with the plasma membrane.40  A recent study reported that cancer cells undergoing epithelial-to-mesenchymal transition release TF-positive exosomes.41  MPs are released from most cell types upon activation, and platelets are the major cellular source of MPs in blood. Indeed, MPs were originally described as “platelet dust.”2,42-44  Importantly, tumor cells spontaneously release MPs.21,22 

MPs are formed by the outward blebbing of the plasma membrane and are subsequently released as small phospholipid vesicles after proteolytic cleavage of the cytoskeleton.40,42  There is selective packaging of surface proteins into MPs,40  including TF12,21,45  and adhesion molecules.46,47  MPs are procoagulant because of the presence of TF2,12  and the exposure of negatively charged phospholipids, such as phosphatidylserine (PS).48  The negatively charged surface facilitates the assembly of positively charged coagulation protein complexes.13,46,49 

Several methods have been used for the quantification of TF-positive MPs in human plasma, including antigen- and activity-based assays.50,51  We believe that functional assays are the best way to measure TF in MPs because they are the most sensitive methods for detecting very low levels of active TF in plasma. In addition, the specificity of some TF antigen-based assays has been questioned.52-54  One study failed to observe a correlation between MP TF activity and 2 antigen-based assays (flow cytometry and a commercial TF enzyme-linked immunosorbent assay).55  In contrast, our laboratory found a strong correlation between MP TF activity and plasma TF protein levels using an “in-house” human TF enzyme-linked immunosorbent assay.56  Several functional assays have been developed that measure TF activity in isolated MPs, using either a single-stage clotting assay or a 2-stage factor Xa generation assay in the presence or absence of an inhibitory anti-TF antibody.55-59  Thaler and colleagues measured levels of MP TF activity in patients with cancer, and found a good correlation between 2 assays.60  We observed comparable levels of TF activity in MPs isolated from lipopolysaccharide-stimulated human whole blood, using 2 centrifugation speeds (either 20 000 × g for 20 minutes [primarily MPs] or 100 000 × g for 1 hour [MPs and exosomes]), suggesting that the majority of vesicle TF is on MPs.59 

Light-scatter flow cytometry is the most commonly used method for the quantification of MPs in clinical samples.61  Several studies have used flow cytometry to detect TF-positive MPs in patients with cancer.55,62,63  However, the low sensitivity of flow cytometry makes it difficult to reliably detect the low levels of TF-positive MPs in clinical samples. A recent publication reported that the level of TF-positive MPs measured using a functional MP TF activity assay correlated with the development of VTE in patients with cancer, whereas no correlation was found using flow cytometry to measure TF-positive MPs.64  Interestingly, we failed to detect TF-positive MPs by flow cytometry in samples from LPS-treated whole blood that had high levels of MP TF activity.59  Impedance-based flow cytometry is an alternative method for measuring levels of TF-positive MPs65 ; it has been used to detect TF-positive MPs in plasma from patients with cancer.66,67 

Animal studies have found that TMPs are released from a variety of tumors in vivo.23,53,68  An early study found that guinea pigs bearing hepatocarcinoma tumors had increased procoagulant activity in cell-free ascites fluid that could be pelleted by ultracentrifugation. The presence of membrane vesicles less than 1 µm in size in this pellet was confirmed by electron microscopy.23  The procoagulant activity of the MPs released from these tumor cells in vitro was later determined to be TF-dependent.24  A more recent study found that severe combined immune deficiency mice bearing TF-expressing human colorectal tumors had increased levels of tumor-derived human TF protein in plasma.68  Further, circulating TF levels correlated with the size of the tumor in these mice.68  Circulating TF-positive TMPs have also been found to be associated with the activation of coagulation in mice.21,41,53  Specifically, increased circulating levels of human TF protein in nude mice bearing orthotopic human pancreatic tumors was associated with activation of coagulation, as monitored by plasma levels of thrombin-antithrombin (TAT) complex.21  In vitro studies showed that the plasma from tumor-bearing nude mice generated increased thrombin compared with control plasma and that this increase could be inhibited by an anti-human TF monoclonal antibody.21  A similar elevation in plasma TAT complex levels was seen in severe combined immune deficiency mice bearing TF-positive tumors formed by the human squamous cell carcinoma cell line A431.41  We found that inhibition of human TF with a species-specific monoclonal antibody reduced TAT levels in nude mice bearing an orthotopic tumor derived from human pancreatic adenocarcinoma cell line HPAF-II.53  However, the relative contribution of TF expression by the TMPs and that of the tumor cells themselves to the activation of coagulation could not be determined.53 

Several studies have also shown that TMPs enhance the development of thrombosis in mice in vivo. Thomas and colleagues found that both endogenously generated and exogenously injected PANC02 TMPs, but not tumor cells, accumulated at the site of ferric chloride–induced mesenteric vessel injury and laser-induced cremaster arteriole injury.69  In addition, accumulation of endogenous TF-positive TMPs in these orthotopic PANC02 pancreatic tumor–bearing mice was shown to be P-selectin-dependent. Interestingly, this study found that both human and mouse pancreatic cell lines expressed P-selectin glycoprotein ligand 1.69  In contrast, we did not detect P-selectin glycoprotein ligand 1 expression by 4 different human pancreatic cell lines but found that they bound to an immobilized P-selectin-IgG chimera, which indicated that they expressed a P-selectin ligand (J.-G. Wang and N. Mackman, unpublished data, 2012). Injected exogenous TMPs enhanced thrombosis, and this enhancement was abolished with an anti-P-selectin antibody.69  We found that mice bearing human pancreatic tumors had increased thrombosis compared with control mice in a ferric chloride saphenous vein model.53  However, as discussed previously, this is not the best model for studying venous thrombosis.

The IVC stenosis model of venous thrombosis is particularly suited for the evaluation of the mechanisms of VTE initiation because thrombosis is triggered by endothelial cell activation and changes in blood flow, rather than denudation of the endothelium and vessel wall damage.35  It should be noted, however, that there is significant variability in thrombosis observed in the IVC stenosis model.35,53,70-72  One study reported increased incidence of thrombosis in this model in C57BL/6J mice bearing subcutaneous PANC02 tumors.73  In contrast, we did not observe an increase in IVC thrombosis in nude mice bearing TF-positive human HPAF-II pancreatic tumors.53  We did find that injection of exogenous TF-positive HPAF-II MPs enhanced thrombosis, although the amount of injected TMPs required to increase thrombosis was 40 times higher than that present in mice with HPAF-II tumors.53  At present, it is unclear the reasons for these different results, but it may be because of differences in experimental conditions, use of different mouse strains, tumor size, and species of the tumors (mouse vs human). Clearly, further studies are needed to understand how TF-positive TMPs contribute to thrombosis in different mouse models.

Cancer cells are well known to express TF and release TF-positive MPs.18-21,69,74  In addition, TF expression increases with histologic grade in different cancer types, including pancreatic cancer.19,20,75  Two studies have reported a correlation between the level of TF in pancreatic and brain tumors and VTE.20,75  In addition to its proposed role in cancer-associated thrombosis, TF has been shown to be involved in tumor growth and metastasis.76,77 

Similarly, TF-positive MPs released from cancer cells have been implicated in non-VTE-related cancer processes.58,78  Tesselaar and colleagues found that patients with advanced breast cancer, but not patients with early-stage cancer, had elevated levels of MP TF activity compared with healthy controls.58  Similarly, we did not detect elevated levels of MP TF activity in a group of 26 patients with early-stage breast cancer.79  Thaler and colleagues recently identified a correlation between increased MP TF activity and worsened cancer stage, grade, and survival in patients with metastatic nonresectable pancreatic cancer.78 

Numerous studies have analyzed levels of circulating TF-positive MPs in patients with cancer. Hron and colleagues used flow cytometry to detect increased levels of TF-positive MPs in patients with advanced colorectal cancer (n = 20) compared with those found in age-matched healthy control individuals.62  Levels of TF-positive MPs in the patients with cancer correlated with activation of coagulation, as determined by d-dimer levels.62  A study of patients with early-stage prostate cancer (n = 69) also observed increased MP TF activity in those with a modest correlation between MP TF and d-dimer levels.55  In patients with glioblastoma, plasma levels of TF-positive MPs, but not MP TF activity, correlated with d-dimer levels.80,81  An observational study of a single patient with giant cell lung carcinoma who had a high rate of thromboembolic events reported vesicle-associated plasma TF antigen levels that were 41 times higher than those seen in 16 healthy control indivduals.45 

Hron and colleagues also found that most of the circulating TF-positive MPs in their patients with advanced colorectal cancer coexpressed platelet antigen CD41a, indicating they were derived from platelets.62  Platelets do not appear to synthesize TF but have been shown to bind monocyte-derived, TF-positive MPs.82-84  Recently, we found that TMPs bind to platelets (J. E. Geddings, W. Bergmeier, N. Mackman, unpublished data). This suggests that TF present on platelet-derived MPs may be a result of the binding of TF-positive TMPs to platelets and their reprocessing to form platelet-derived MPs. We found an increase in TF activity in combined platelet and MP samples from patients with cancer compared with healthy control individuals.85  Therefore, levels of TF activity in a combined platelet and MP sample may be a more accurate measure of thrombotic risk in patients with cancer compared with the TF activity of MPs alone.

Cancer chemotherapy is known to be associated with increased thrombosis.7,8  We found that treatment of the human monocytic cell line THP-1 with cytotoxic chemotherapy agents enhanced cellular TF activity without increasing TF expression by increasing cellular PS exposure.86  Further, these cells demonstrated increased release of TF-positive MPs.86  Increased PS expression or release of PS-positive MPs has also been observed with cytotoxic chemotherapy treatment of other cell types, including endothelial cells, red blood cells, and acute promyelocytic leukemia cells.87-90 

Tesselaar and colleagues found that patients with cancer receiving chemotherapy do not have elevated plasma MP TF activity in comparison to untreated patients with cancer.91  We evaluated the effect of chemotherapy on MP TF activity in 26 patients with early-stage breast cancer.79  Plasma samples were analyzed at days 0, 2, and 8 after the start of chemotherapy, during the first 2 cycles of chemotherapy, but no increase in MP TF activity was observed.79  A recent study suggested that the activation of coagulation in patients with early-stage breast cancer undergoing chemotherapy may be mediated by the release of free DNA.92 

These findings suggest that mechanisms other than increased levels of circulating TF-positive MPs are responsible for thrombosis during cancer chemotherapy, such as release of nucleic acids and increased cellular PS exposure.

Retrospective studies

The earliest retrospective study that analyzed the link between TF-positive MPs and VTE in patients with cancer was performed by Tesselaar and colleagues in 2007 (Table 1).58  This study included 23 patients with nonresectable pancreatic adenocarcinoma, 27 patients with breast ductal adenocarcinoma (10 patients with early- and 17 patients with late-stage breast cancer), 7 patients with idiopathic VTE, and 37 healthy control participants. Mean MP TF activity was significantly higher in patients with pancreatic and metastatic breast cancer compared with either healthy control participants or patients with idiopathic VTE. Interestingly, patients with pancreatic cancer had higher MP TF activity than patients with breast cancer. Patients with cancer who also had VTE had higher plasma MP TF activity than patients with cancer without VTE.58  Approximately 50% of MPs in the patients with cancer expressed the tumor antigen mucin 1 (MUC-1) on their surface, which suggested they were derived from the tumor. Further, no MUC-1-positive TMPs were detected by light-scatter flow cytometry after breast tumor resection.58 

Table 1

Studies evaluating the association between TF-positive MPs and VTE in patients with cancer

StudyMethodPatientsTime of follow-upAssociation between MP TF and VTE?
Retrospective     
 Tesselaar, 200758  MP TF activity assay 7 late stage pancreatic and breast cancer patients with VTE n/a Yes 
33 late stage pancreatic and breast cancer patients without VTE 
 Zwicker, 200966  Impedance-based flow cytometry 30 cancer patients with VTE n/a Yes 
60 case-matched cancer patients without VTE 
 Tesselaar, 200991  MP TF activity assay 51 cancer patients with VTE n/a Yes 
49 case-matched cancer patients without VTE 
 Manly, 201093  MP TF activity 53 cancer patients with VTE n/a Yes 
13 cancer patients without VTE 
 Campello, 201163  Flow cytometry 30 cancer patients with VTE n/a Yes 
30 case-matched cancer patients without VTE 
Prospective     
 Khorana, 200856  MP TF activity 2 pancreatic cancer patients who develop VTE Every 4 wk for 20 wk Yes 
TF enzyme-linked immunosorbent assay 8 pancreatic cancer patients without VTE 
 Zwicker, 200966  Impedance-based flow cytometry 4 cancer patients who develop VTE 1 y Yes 
56 cancer patients without VTE 
 Bharthuar, 201097  MP TF activity 52 Pancreaticobiliary cancer patients who develop VTE 6 mo Yes 
65 Pancreaticobiliary cancer patients without VTE 
 Auwerda, 201194  MP TF activity 15 multiple myeloma patients undergoing chemotherapy with VTE Not specified No 
107 multiple myeloma patients undergoing chemotherapy without VTE 
 Van Doormaal, 201264  MP TF activityMP-dependent fibrin generation flow cytometryTF enzyme-linked immunosorbent assay 5 cancer patients who develop VTE (3 pancreatic)38 cancer patients without VTE (10 pancreatic) 6 mo Yes by functional assayNo by antigen assay 
 Thaler, 201260  MP TF activity 49 cancer patients who develop VTE (12 pancreatic) 2 y No 
299 cancer patients without VTE (48 pancreatic) 
 Sartori, 201381  Flow cytometry 11 glioblastoma patients who develop VTE 7 mo Yes 
50 glioblastoma patients without VTE 
StudyMethodPatientsTime of follow-upAssociation between MP TF and VTE?
Retrospective     
 Tesselaar, 200758  MP TF activity assay 7 late stage pancreatic and breast cancer patients with VTE n/a Yes 
33 late stage pancreatic and breast cancer patients without VTE 
 Zwicker, 200966  Impedance-based flow cytometry 30 cancer patients with VTE n/a Yes 
60 case-matched cancer patients without VTE 
 Tesselaar, 200991  MP TF activity assay 51 cancer patients with VTE n/a Yes 
49 case-matched cancer patients without VTE 
 Manly, 201093  MP TF activity 53 cancer patients with VTE n/a Yes 
13 cancer patients without VTE 
 Campello, 201163  Flow cytometry 30 cancer patients with VTE n/a Yes 
30 case-matched cancer patients without VTE 
Prospective     
 Khorana, 200856  MP TF activity 2 pancreatic cancer patients who develop VTE Every 4 wk for 20 wk Yes 
TF enzyme-linked immunosorbent assay 8 pancreatic cancer patients without VTE 
 Zwicker, 200966  Impedance-based flow cytometry 4 cancer patients who develop VTE 1 y Yes 
56 cancer patients without VTE 
 Bharthuar, 201097  MP TF activity 52 Pancreaticobiliary cancer patients who develop VTE 6 mo Yes 
65 Pancreaticobiliary cancer patients without VTE 
 Auwerda, 201194  MP TF activity 15 multiple myeloma patients undergoing chemotherapy with VTE Not specified No 
107 multiple myeloma patients undergoing chemotherapy without VTE 
 Van Doormaal, 201264  MP TF activityMP-dependent fibrin generation flow cytometryTF enzyme-linked immunosorbent assay 5 cancer patients who develop VTE (3 pancreatic)38 cancer patients without VTE (10 pancreatic) 6 mo Yes by functional assayNo by antigen assay 
 Thaler, 201260  MP TF activity 49 cancer patients who develop VTE (12 pancreatic) 2 y No 
299 cancer patients without VTE (48 pancreatic) 
 Sartori, 201381  Flow cytometry 11 glioblastoma patients who develop VTE 7 mo Yes 
50 glioblastoma patients without VTE 

n/a, not applicable.

Zwicker and colleagues observed elevated levels of TF-positive MPs in the plasma of patients with pancreatic, breast, colorectal, ovarian, and non-small cell lung cancer, using impedance-based flow cytometry.66  This study included 30 patients with cancer with VTE at study entry and 60 case-matched cancer patient controls without VTE. TF-positive MPs were detected in the plasma of 60% of the patients with cancer who had VTE compared with 27% of patients with cancer without VTE. Again, 50% of the circulating TF-positive MPs in 3 patients with pancreatic cancer expressed MUC-1. Similar to the study by Tesselaar and colleagues,58  plasma levels of TF and MUC-1 double-positive MPs in these 3 patients were reduced after surgical resection of the primary tumor.66 

A second study by Tesselaar and colleagues measured plasma MP TF activity levels in 51 unselected patients with cancer who presented with VTE in comparison with case-matched patients with cancer without VTE.91  The cancer types in this study included gastrointestinal tract (n = 27), genitourinary tract (n = 12), and a variety of other cancer types (n = 13). MP TF activity was higher in the patients with VTE compared with the non-VTE patients. Interestingly, patients with pancreatic cancer with VTE had the highest levels of MP TF activity. Increased MP TF activity was found to be associated with decreased survival of patients with cancer. Cancer patients with VTE also had increased plasma TAT levels, and there was a modest correlation between MP TF activity and TAT levels.91  Similar to Tesselaar and colleagues,91  we found increased levels of MP TF activity in patients with cancer who had VTE (n = 53) in comparison with non-VTE patients (n = 13), with the highest levels being in patients with pancreatic cancer.93  A more recent retrospective study also found increased levels of circulating TF-positive MPs identified by flow cytometry in patients with cancer who had VTE compared with patients with cancer without VTE.63 

Prospective studies

The earliest prospective study was performed by Khorana and colleagues in 2008.56  MP TF activity was measured in the plasma of 10 patients with pancreatic cancer receiving chemotherapy. Blood samples were drawn before starting chemotherapy and every 4 weeks during a 20-week period. MP TF activity was increased in 2 of the 10 patients, and these 2 patients developed VTE during the study. This is the first study to provide evidence to support the hypothesis that elevated levels of MP TF activity precede thrombosis and may be predictive of VTE in patients with pancreatic cancer.56 

Since this initial study, several other studies have evaluated the association between plasma levels of TF-positive MPs and development of VTE in patients with cancer. In the second portion of the study by Zwicker and colleagues, the researchers measured plasma levels of TF-positive MPs in patients with cancer and then followed these patients for the development of VTE.66  This study found a 1-year VTE rate estimate in patients with detectable TF-positive MPs of 34.8% (4 out of 16) compared with 0% (0 out of 44) in patients without detectable TF-positive MPs.66  Another study measured MP TF activity in 122 patients newly diagnosed with multiple myeloma who were eligible for high-dose chemotherapy.94  MP TF activity levels were elevated in patients with multiple myeloma but were not predictive of future VTE events.94  However, the rationale for this study is not clear, as neoplastic plasma cells have not been reported to express TF.95 

Van Doormaal and colleagues followed 43 patients with cancer (13 with pancreatic cancer) for a period of 6 months for the development of VTE.64  Five patients developed VTE during the study, 3 of whom were patients with pancreatic cancer. Levels of TF antigen were measured by flow cytometry and enzyme-linked immunosorbent assay, and TF and TF-factor VIIa activity of MPs were measured using the MP TF activity assay and a fibrin generation test with or without an anti-factor VIIa blocking antibody, respectively.64  In addition, levels of PS-positive MPs were measured using flow cytometry and a functional assay. This study found that there was an association between both MP TF activity and MP TF-factor VIIa-dependent fibrin generation and the development of VTE. In contrast, VTE did not correlate with either of the TF antigen-based assays or the level of PS-positive MPs.64  These results indicate that it is important to measure levels of MP TF activity, and not simply levels of PS-positive MPs.96 

The most extensive study to date measured MP TF activity in plasma samples from the Vienna cancer and thrombosis study.60  Four different types of patients with cancer (60 pancreatic, 43 gastric, 126 colorectal, and 119 brain) were chosen because these cancers have a relatively high rate of VTE. These patients were followed for up to 2 years for the development of VTE. For many samples, plasma MP TF activity was measured using both the end-point57  and kinetic58  assays. Patients with pancreatic and gastric cancer expressed higher levels of MP TF activity in comparison to patients with brain and colorectal cancer. MP TF activity was associated with decreased survival in patients with pancreatic and gastric cancer. Patients with pancreatic cancer demonstrated borderline significance for an association between MP TF activity and VTE in the end-point assay.60 

It is somewhat surprising that the level of circulating MP TF activity was not increased in patients with brain cancer in the above study.60  It is possible that the blood–brain barrier may limit the release of TMPs into the circulation in these patients. Sartori and colleagues found that procoagulant activity of MPs and TF-positive MPs measured by flow cytometry were increased in 61 patients with preoperative glioblastoma multiforme.80,81  Levels of TF-positive MPs were also increased over baseline at 7 days and at 1 month after tumor resection.81  This increase is most likely a result of disruption of the blood–brain barrier during surgery.81  Furthermore, the 11 patients who developed VTE had significantly higher levels of non-tumor-derived TF-positive MPs at baseline than the non-VTE patients.81  However, no increases in MP TF activity were detected in the same samples (R. L. Bradford and N. S. Key, unpublished data).

We have also analyzed MP TF activity in 117 patients with newly diagnosed pancreaticobiliary cancer at all stages. These patients are part of the Roswell Park Cancer Institute Data Bank and Biorepository.97  Blood was collected from each patient after cancer diagnosis. In this study, elevated plasma MP TF activity was associated with the future development of VTE and decreased survival.97 

A general issue with prospective studies is that the collection of blood samples may occur many months before the thrombotic event. Most studies rely on a single sample to predict future events. We found that in 2 patients with pancreatic cancer, MP TF activity serially increased in the months before the development of thrombosis.56  Further, MP TF activity was found to correlate with VTE in a study with a 6-month follow-up,64  but only weakly in a study with a 2-year follow-up.60  Clearly, it would be better to collect multiple blood samples from each patient, which would allow for the monitoring of levels of TF-positive MPs over time. We have an ongoing study in which we collect 8 blood samples from patients with either advanced pancreatic cancer or advanced colorectal cancer before and during 4 cycles of chemotherapy and monitor for the development of symptomatic and asymptomatic VTE. Using this study design, we will further examine the hypothesis that elevated levels of MP TF activity are predictive of symptomatic and asymptomatic VTE in these patients.

In summary, TF is expressed by tumor cells, and expression is increased in advanced cancer. Tumor cells spontaneously release high levels of TF-positive MPs that are associated with a prothrombotic state and enhanced thrombosis in animal models. In human cancer patient studies, elevated levels of MP TF activity are predictive of VTE in the majority of studies, suggesting that MP TF likely contributes to thrombosis in patients with cancer, particularly in those with pancreatic cancer. However, the current laboratory-based assays need to be improved before they can be used for clinical diagnosis. The clinical utility for the use of plasma MP-TF in the guiding of thromboprophylaxis in patients with cancer also remains to be determined. The first study using plasma MP-TF to guide this type of treatment decision has produced promising results.67  The MicroTEC study found that use of the low–molecular weight heparin enoxaparin for thromboprophylaxis in patients with cancer with high plasma MP-TF reduced VTE and improved patient survival.67 

Most studies of patients with cancer focus on measurement of MP TF activity rather than TF activity in other cells. However, levels of MP TF activity may represent the tip of the iceberg in terms of the MP-associated procoagulant activity present in the blood of patients with cancer, as TMPs may be binding to other vascular cell types. Indeed, a recent study analyzed circulating TF activity in the blood of patients undergoing total knee arthroplasty and found that the majority of circulating TF was associated with monocytes, with only ∼5% of the TF activity present in the free MP fraction.98  It is possible that measurement of TF activity in vascular cell populations will be more predictive of thrombosis than MP TF activity. More studies are needed to determine the distribution of TF-positive TMPs in the circulation in animal models and patients with cancer and to characterize the tumor markers and adhesion proteins that are present on the surface of the TMPs (Figure 1). One study found that P-selectin was required for the TMP-induced enhancement of microvascular thrombosis.69  However, it seems likely that other receptors contribute to the delivery of TMPs to thrombosis sites and that these receptors may represent good targets for the development of cancer-specific antithrombotic drugs.

Figure 1

TMP surface proteins and their functions. TMPs are constitutively released from tumors into the circulation. The procoagulant activity of TMPs is mediated by the expression of TF and the exposure of PS on the MP surface. Tumor markers such as MUC-1 can allow for the identification of TMPs in the circulation. Adhesion proteins including P-selectin ligand CD24 and E-selectin ligand CD43 have been proposed to be involved in the binding of TMPs to endothelium and thrombosis sites.2,69  Delivery of TMP TF to the site of thrombosis can then initiate thrombosis. This diagram is an example of proteins that can be expressed on the surface of TMPs. Protein expression on the surface of TMPs varies with each tumor. FVIIa, factor VIIa.

Figure 1

TMP surface proteins and their functions. TMPs are constitutively released from tumors into the circulation. The procoagulant activity of TMPs is mediated by the expression of TF and the exposure of PS on the MP surface. Tumor markers such as MUC-1 can allow for the identification of TMPs in the circulation. Adhesion proteins including P-selectin ligand CD24 and E-selectin ligand CD43 have been proposed to be involved in the binding of TMPs to endothelium and thrombosis sites.2,69  Delivery of TMP TF to the site of thrombosis can then initiate thrombosis. This diagram is an example of proteins that can be expressed on the surface of TMPs. Protein expression on the surface of TMPs varies with each tumor. FVIIa, factor VIIa.

Close modal

This work was supported by grants from the National Heart, Lung, and Blood Institute at the National Institutes of Health, grant numbers R01HL095096-04 and R01HL095096-04S (to N.M. and N. S. Key).

Contribution: J.G. and N.M wrote this review.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Nigel Mackman, Division of Hematology/Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599; e-mail: nmackman@med.unc.edu.

1
Varki
 
A
Trousseau’s syndrome: multiple definitions and multiple mechanisms.
Blood
2007
, vol. 
110
 
6
(pg. 
1723
-
1729
)
2
Mackman
 
N
New insights into the mechanisms of venous thrombosis.
J Clin Invest
2012
, vol. 
122
 
7
(pg. 
2331
-
2336
)
3
Menapace
 
LA
Peterson
 
DR
Berry
 
A
Sousou
 
T
Khorana
 
AA
Symptomatic and incidental thromboembolism are both associated with mortality in pancreatic cancer.
Thromb Haemost
2011
, vol. 
106
 
2
(pg. 
371
-
378
)
4
Khorana
 
AA
Francis
 
CW
Culakova
 
E
Kuderer
 
NM
Lyman
 
GH
Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy.
J Thromb Haemost
2007
, vol. 
5
 
3
(pg. 
632
-
634
)
5
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
, vol. 
166
 
4
(pg. 
458
-
464
)
6
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
, vol. 
4
 
3
(pg. 
529
-
535
)
7
Khorana
 
AA
Connolly
 
GC
Assessing risk of venous thromboembolism in the patient with cancer.
J Clin Oncol
2009
, vol. 
27
 
29
(pg. 
4839
-
4847
)
8
Falanga
 
A
Marchetti
 
M
Russo
 
L
Venous thromboembolism in the hematologic malignancies.
Curr Opin Oncol
2012
, vol. 
24
 
6
(pg. 
702
-
710
)
9
Khorana
 
AA
Kuderer
 
NM
Culakova
 
E
Lyman
 
GH
Francis
 
CW
Development and validation of a predictive model for chemotherapy-associated thrombosis.
Blood
2008
, vol. 
111
 
10
(pg. 
4902
-
4907
)
10
Cannegieter
 
S
Epidemiology of cancer-associated thrombosis.
Blood
 
In press
11
Lee
 
A
Treatment of cancer-related thrombosis.
Blood
 
In press
12
Mackman
 
N
Tilley
 
RE
Key
 
NS
Role of the extrinsic pathway of blood coagulation in hemostasis and thrombosis.
Arterioscler Thromb Vasc Biol
2007
, vol. 
27
 
8
(pg. 
1687
-
1693
)
13
Owens
 
AP
Mackman
 
N
Tissue factor and thrombosis: The clot starts here.
Thromb Haemost
2010
, vol. 
104
 
3
(pg. 
432
-
439
)
14
Vadivel
 
K
Bajaj
 
SP
Structural biology of factor VIIa/tissue factor initiated coagulation.
Front Biosci
2012
, vol. 
17
 (pg. 
2476
-
2494
)
15
Mackman
 
N
The many faces of tissue factor.
J Thromb Haemost
2009
, vol. 
7
 
Suppl 1
(pg. 
136
-
139
)
16
Osterud
 
B
Bjorklid
 
E
Tissue factor in blood cells and endothelial cells.
Front Biosci (Elite Ed)
2012
, vol. 
4
 (pg. 
289
-
299
[Elite Ed]
17
Butenas
 
S
Orfeo
 
T
Mann
 
KG
Tissue factor in coagulation: Which? Where? When?
Arterioscler Thromb Vasc Biol
2009
, vol. 
29
 
12
(pg. 
1989
-
1996
)
18
Callander
 
NS
Varki
 
N
Rao
 
LV
Immunohistochemical identification of tissue factor in solid tumors.
Cancer
1992
, vol. 
70
 
5
(pg. 
1194
-
1201
)
19
Kakkar
 
AK
Lemoine
 
NR
Scully
 
MF
Tebbutt
 
S
Williamson
 
RC
Tissue factor expression correlates with histological grade in human pancreatic cancer.
Br J Surg
1995
, vol. 
82
 
8
(pg. 
1101
-
1104
)
20
Khorana
 
AA
Ahrendt
 
SA
Ryan
 
CK
, et al. 
Tissue factor expression, angiogenesis, and thrombosis in pancreatic cancer.
Clin Cancer Res
2007
, vol. 
13
 
10
(pg. 
2870
-
2875
)
21
Davila
 
M
Amirkhosravi
 
A
Coll
 
E
, et al. 
Tissue factor-bearing microparticles derived from tumor cells: impact on coagulation activation.
J Thromb Haemost
2008
, vol. 
6
 
9
(pg. 
1517
-
1524
)
22
Al-Nedawi
 
K
Meehan
 
B
Micallef
 
J
Lhotak
 
V
May
 
L
Guha
 
A
Rak
 
J
Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells.
Nat Cell Biol
2008
, vol. 
10
 
5
(pg. 
619
-
624
)
23
Dvorak
 
HF
Quay
 
SC
Orenstein
 
NS
Dvorak
 
AM
Hahn
 
P
Bitzer
 
AM
Carvalho
 
AC
Tumor shedding and coagulation.
Science
1981
, vol. 
212
 
4497
(pg. 
923
-
924
)
24
Dvorak
 
HF
Van DeWater
 
L
Bitzer
 
AM
, et al. 
Procoagulant activity associated with plasma membrane vesicles shed by cultured tumor cells.
Cancer Res
1983
, vol. 
43
 
9
(pg. 
4434
-
4442
)
25
Mackman
 
N
Triggers, targets and treatments for thrombosis.
Nature
2008
, vol. 
451
 
7181
(pg. 
914
-
918
)
26
Sevitt
 
S
The structure and growth of valve-pocket thrombi in femoral veins.
J Clin Pathol
1974
, vol. 
27
 
7
(pg. 
517
-
528
)
27
Bovill
 
EG
van der Vliet
 
A
Venous valvular stasis-associated hypoxia and thrombosis: what is the link?
Annu Rev Physiol
2011
, vol. 
73
 (pg. 
527
-
545
)
28
Kumar
 
DR
Hanlin
 
E
Glurich
 
I
Mazza
 
JJ
Yale
 
SH
Virchow’s contribution to the understanding of thrombosis and cellular biology.
Clin Med Res
2010
, vol. 
8
 
3-4
(pg. 
168
-
172
)
29
Moll
 
S
Mackman
 
N
Venous thromboembolism: a need for more public awareness and research into mechanisms.
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 
3
(pg. 
367
-
369
)
30
Reitsma
 
PH
Versteeg
 
HH
Middeldorp
 
S
Mechanistic view of risk factors for venous thromboembolism.
Arterioscler Thromb Vasc Biol
2012
, vol. 
32
 
3
(pg. 
563
-
568
)
31
Manly
 
DA
Boles
 
J
Mackman
 
N
Role of tissue factor in venous thrombosis.
Annu Rev Physiol
2011
, vol. 
73
 (pg. 
515
-
525
)
32
Brooks
 
EG
Trotman
 
W
Wadsworth
 
MP
, et al. 
Valves of the deep venous system: an overlooked risk factor.
Blood
2009
, vol. 
114
 
6
(pg. 
1276
-
1279
)
33
Diaz
 
JA
Obi
 
AT
Myers
 
DD
, et al. 
Critical review of mouse models of venous thrombosis.
Arterioscler Thromb Vasc Biol
2012
, vol. 
32
 
3
(pg. 
556
-
562
)
34
Hampton
 
AL
Diaz
 
JA
Hawley
 
AE
, et al. 
Myeloid cell tissue factor does not contribute to venous thrombogenesis in an electrolytic injury model.
Thromb Res
2012
, vol. 
130
 
4
(pg. 
640
-
645
)
35
von Bruhl
 
ML
Stark
 
K
Steinhart
 
A
, et al. 
 
Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. 2012;209(4):819-835
36
Chou
 
J
Mackman
 
N
Merrill-Skoloff
 
G
Pedersen
 
B
Furie
 
BC
Furie
 
B
Hematopoietic cell-derived microparticle tissue factor contributes to fibrin formation during thrombus propagation.
Blood
2004
, vol. 
104
 
10
(pg. 
3190
-
3197
)
37
Darbousset
 
R
Thomas
 
GM
Mezouar
 
S
, et al. 
Tissue factor-positive neutrophils bind to injured endothelial wall and initiate thrombus formation.
Blood
2012
, vol. 
120
 
10
(pg. 
2133
-
2143
)
38
Day
 
SM
Reeve
 
JL
Pedersen
 
B
, et al. 
Macrovascular thrombosis is driven by tissue factor derived primarily from the blood vessel wall.
Blood
2005
, vol. 
105
 
1
(pg. 
192
-
198
)
39
György
 
B
Szabó
 
TG
Pásztói
 
M
, et al. 
Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles.
Cell Mol Life Sci
2011
, vol. 
68
 
16
(pg. 
2667
-
2688
)
40
D’Souza-Schorey
 
C
Clancy
 
JW
Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers.
Genes Dev
2012
, vol. 
26
 
12
(pg. 
1287
-
1299
)
41
Garnier
 
D
Magnus
 
N
Lee
 
TH
, et al. 
Cancer cells induced to express mesenchymal phenotype release exosome-like extracellular vesicles carrying tissue factor.
J Biol Chem
2012
, vol. 
287
 
52
(pg. 
43565
-
43572
)
42
Burnier
 
L
Fontana
 
P
Kwak
 
BR
Angelillo-Scherrer
 
A
Cell-derived microparticles in haemostasis and vascular medicine.
Thromb Haemost
2009
, vol. 
101
 
3
(pg. 
439
-
451
)
43
Owens
 
AP
Mackman
 
N
Microparticles in hemostasis and thrombosis.
Circ Res
2011
, vol. 
108
 
10
(pg. 
1284
-
1297
)
44
Wolf
 
P
The nature and significance of platelet products in human plasma.
Br J Haematol
1967
, vol. 
13
 
3
(pg. 
269
-
288
)
45
Del Conde
 
I
Bharwani
 
LD
Dietzen
 
DJ
Pendurthi
 
U
Thiagarajan
 
P
López
 
JA
Microvesicle-associated tissue factor and Trousseau’s syndrome.
J Thromb Haemost
2007
, vol. 
5
 
1
(pg. 
70
-
74
)
46
Mause
 
SF
Weber
 
C
Microparticles: protagonists of a novel communication network for intercellular information exchange.
Circ Res
2010
, vol. 
107
 
9
(pg. 
1047
-
1057
)
47
Ramacciotti
 
E
Hawley
 
AE
Wrobleski
 
SK
, et al. 
Proteomics of microparticles after deep venous thrombosis.
Thromb Res
2010
, vol. 
125
 
6
(pg. 
e269
-
e274
)
48
Aleman
 
MM
Gardiner
 
C
Harrison
 
P
Wolberg
 
AS
Differential contributions of monocyte- and platelet-derived microparticles towards thrombin generation and fibrin formation and stability.
J Thromb Haemost
2011
, vol. 
9
 
11
(pg. 
2251
-
2261
)
49
Morrissey
 
JH
Tajkhorshid
 
E
Rienstra
 
CM
Nanoscale studies of protein-membrane interactions in blood clotting.
J Thromb Haemost
2011
, vol. 
9
 
Suppl 1
(pg. 
162
-
167
)
50
Key
 
NS
Mackman
 
N
Tissue factor and its measurement in whole blood, plasma, and microparticles.
Semin Thromb Hemost
2010
, vol. 
36
 
8
(pg. 
865
-
875
)
51
Thaler
 
J
Ay
 
C
Pabinger
 
I
Clinical significance of circulating microparticles for venous thromboembolism in cancer patients.
Hamostaseologie
2012
, vol. 
32
 
2
(pg. 
127
-
131
)
52
Parhami-Seren
 
B
Butenas
 
S
Krudysz-Amblo
 
J
Mann
 
KG
Immunologic quantitation of tissue factors.
J Thromb Haemost
2006
, vol. 
4
 
8
(pg. 
1747
-
1755
)
53
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
54
Basavaraj
 
MG
Olsen
 
JO
Østerud
 
B
Hansen
 
JB
Differential ability of tissue factor antibody clones on detection of tissue factor in blood cells and microparticles.
Thromb Res
2012
, vol. 
130
 
3
(pg. 
538
-
546
)
55
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
, vol. 
101
 
6
(pg. 
1147
-
1155
)
56
Khorana
 
AA
Francis
 
CW
Menzies
 
KE
, et al. 
Plasma tissue factor may be predictive of venous thromboembolism in pancreatic cancer.
J Thromb Haemost
2008
, vol. 
6
 
11
(pg. 
1983
-
1985
)
57
Wang
 
JG
Manly
 
D
Kirchhofer
 
D
Pawlinski
 
R
Mackman
 
N
 
Levels of microparticle tissue factor activity correlate with coagulation activation in endotoxemic mice. J Thromb Haemost. 2009;7(7):1092-1098
58
Tesselaar
 
ME
Romijn
 
FP
Van Der Linden
 
IK
Prins
 
FA
Bertina
 
RM
Osanto
 
S
Microparticle-associated tissue factor activity: a link between cancer and thrombosis?
J Thromb Haemost
2007
, vol. 
5
 
3
(pg. 
520
-
527
)
59
Lee
 
RD
Barcel
 
DA
Williams
 
JC
, et al. 
Pre-analytical and analytical variables affecting the measurement of plasma-derived microparticle tissue factor activity.
Thromb Res
2012
, vol. 
129
 
1
(pg. 
80
-
85
)
60
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
, vol. 
10
 
7
(pg. 
1363
-
1370
)
61
Lacroix
 
R
Robert
 
S
Poncelet
 
P
Dignat-George
 
F
Overcoming limitations of microparticle measurement by flow cytometry.
Semin Thromb Hemost
2010
, vol. 
36
 
8
(pg. 
807
-
818
)
62
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
, vol. 
97
 
1
(pg. 
119
-
123
)
63
Campello
 
E
Spiezia
 
L
Radu
 
CM
, et al. 
Endothelial, platelet, and tissue factor-bearing microparticles in cancer patients with and without venous thromboembolism.
Thromb Res
2011
, vol. 
127
 
5
(pg. 
473
-
477
)
64
van Doormaal
 
F
Kleinjan
 
A
Berckmans
 
RJ
, et al. 
Coagulation activation and microparticle-associated coagulant activity in cancer patients. An exploratory prospective study.
Thromb Haemost
2012
, vol. 
108
 
1
(pg. 
160
-
165
)
65
Zwicker
 
JI
Impedance-based flow cytometry for the measurement of microparticles.
Semin Thromb Hemost
2010
, vol. 
36
 
8
(pg. 
819
-
823
)
66
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
, vol. 
15
 
22
(pg. 
6830
-
6840
)
67
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
, vol. 
160
 
4
(pg. 
530
-
537
)
68
Yu
 
JL
May
 
L
Lhotak
 
V
, et al. 
 
Oncogenic events regulate tissue factor expression in colorectal cancer cells: implications for tumor progression and angiogenesis. Blood. 2005;105(4):1734-1741
69
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
, vol. 
206
 
9
(pg. 
1913
-
1927
)
70
Brill
 
A
Fuchs
 
TA
Chauhan
 
AK
, et al. 
von Willebrand factor-mediated platelet adhesion is critical for deep vein thrombosis in mouse models.
Blood
2011
, vol. 
117
 
4
(pg. 
1400
-
1407
)
71
Brill
 
A
Fuchs
 
TA
Savchenko
 
AS
, et al. 
Neutrophil extracellular traps promote deep vein thrombosis in mice.
J Thromb Haemost
2012
, vol. 
10
 
1
(pg. 
136
-
144
)
72
Brill
 
A
Yesilaltay
 
A
De Meyer
 
SF
, et al. 
Extrahepatic high-density lipoprotein receptor SR-BI and apoA-I protect against deep vein thrombosis in mice.
Arterioscler Thromb Vasc Biol
2012
, vol. 
32
 
8
(pg. 
1841
-
1847
)
73
Thomas
 
GM
Brill
 
A
Wagner
 
DD
Cancer-Associated Thrombosis: Cancer Cell-Derived Microparticles as a Trigger of Venous Thrombosis in a Mouse Model of Flow Restriction. Blood (ASH Annual Meeting Abstracts)
2011
 
118:Abstract 34
74
Yu
 
JL
Rak
 
JW
Shedding of tissue factor (TF)-containing microparticles rather than alternatively spliced TF is the main source of TF activity released from human cancer cells.
J Thromb Haemost
2004
, vol. 
2
 
11
(pg. 
2065
-
2067
)
75
Thaler
 
J
Preusser
 
M
Ay
 
C
, et al. 
Intratumoral tissue factor expression and risk of venous thromboembolism in brain tumor patients.
Thromb Res
2013
, vol. 
131
 
2
(pg. 
162
-
165
)
76
Kasthuri
 
RS
Taubman
 
MB
Mackman
 
N
Role of tissue factor in cancer.
J Clin Oncol
2009
, vol. 
27
 
29
(pg. 
4834
-
4838
)
77
van den Berg
 
YW
Osanto
 
S
Reitsma
 
PH
Versteeg
 
HH
The relationship between tissue factor and cancer progression: insights from bench and bedside.
Blood
2012
, vol. 
119
 
4
(pg. 
924
-
932
)
78
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
, vol. 
43
 
3
(pg. 
277
-
285
)
79
Mukherjee
 
SD
Swystun
 
LL
Mackman
 
N
Wang
 
JG
Pond
 
G
Levine
 
MN
Liaw
 
PC
Impact of chemotherapy on thrombin generation and on the protein C pathway in breast cancer patients.
Pathophysiol Haemost Thromb
2010
, vol. 
37
 
2-4
(pg. 
88
-
97
)
80
Sartori
 
MT
Della Puppa
 
A
Ballin
 
A
, et al. 
Prothrombotic state in glioblastoma multiforme: an evaluation of the procoagulant activity of circulating microparticles.
J Neurooncol
2011
, vol. 
104
 
1
(pg. 
225
-
231
)
81
Sartori
 
MT
Della Puppa
 
A
Ballin
 
A
, et al. 
Circulating microparticles of glial origin and tissue factor bearing in high-grade glioma: a potential prothrombotic role.
Thromb Haemost
 
2013;110:2
82
Del Conde
 
I
Shrimpton
 
CN
Thiagarajan
 
P
López
 
JA
Tissue-factor-bearing microvesicles arise from lipid rafts and fuse with activated platelets to initiate coagulation.
Blood
2005
, vol. 
106
 
5
(pg. 
1604
-
1611
)
83
Rauch
 
U
Bonderman
 
D
Bohrmann
 
B
, et al. 
Transfer of tissue factor from leukocytes to platelets is mediated by CD15 and tissue factor.
Blood
2000
, vol. 
96
 
1
(pg. 
170
-
175
)
84
Sovershaev
 
MA
Egorina
 
EM
Osterud
 
B
Hansen
 
JB
Evidence for direct transfer of tissue factor from monocytes to platelets in whole blood.
Blood Coagul Fibrinolysis
2012
, vol. 
23
 
4
(pg. 
345
-
350
)
85
Tilley
 
RE
Holscher
 
T
Belani
 
R
Nieva
 
J
Mackman
 
N
Tissue factor activity is increased in a combined platelet and microparticle sample from cancer patients.
Thromb Res
2008
, vol. 
122
 
5
(pg. 
604
-
609
)
86
Boles
 
JC
Williams
 
JC
Hollingsworth
 
RM
, et al. 
Anthracycline treatment of the human monocytic leukemia cell line THP-1 increases phosphatidylserine exposure and tissue factor activity.
Thromb Res
2012
, vol. 
129
 
2
(pg. 
197
-
203
)
87
Lechner
 
D
Kollars
 
M
Gleiss
 
A
Kyrle
 
PA
Weltermann
 
A
Chemotherapy-induced thrombin generation via procoagulant endothelial microparticles is independent of tissue factor activity.
J Thromb Haemost
2007
, vol. 
5
 
12
(pg. 
2445
-
2452
)
88
Fu
 
Y
Zhou
 
J
Li
 
H
, et al. 
Daunorubicin induces procoagulant activity of cultured endothelial cells through phosphatidylserine exposure and microparticles release.
Thromb Haemost
2010
, vol. 
104
 
6
(pg. 
1235
-
1241
)
89
Zhou
 
J
Zheng
 
Y
Shi
 
J
, et al. 
Daunorubicin induces procoagulant response through phosphatidylserine exposure in red blood cells.
Thromb Res
2010
, vol. 
125
 
2
(pg. 
178
-
183
)
90
Zhou
 
J
Shi
 
J
Hou
 
J
, et al. 
Phosphatidylserine exposure and procoagulant activity in acute promyelocytic leukemia.
J Thromb Haemost
2010
, vol. 
8
 
4
(pg. 
773
-
782
)
91
Tesselaar
 
ME
Romijn
 
FP
van der Linden
 
IK
Bertina
 
RM
Osanto
 
S
Microparticle-associated tissue factor activity in cancer patients with and without thrombosis.
J Thromb Haemost
2009
, vol. 
7
 
8
(pg. 
1421
-
1423
)
92
Swystun
 
LL
Mukherjee
 
S
Liaw
 
PC
Breast cancer chemotherapy induces the release of cell-free DNA, a novel procoagulant stimulus.
J Thromb Haemost
2011
, vol. 
9
 
11
(pg. 
2313
-
2321
)
93
Manly
 
DA
Wang
 
J
Glover
 
SL
Kasthuri
 
R
Liebman
 
HA
Key
 
NS
Mackman
 
N
Increased microparticle tissue factor activity in cancer patients with Venous Thromboembolism.
Thromb Res
2010
, vol. 
125
 
6
(pg. 
511
-
512
)
94
Auwerda
 
JJ
Yuana
 
Y
Osanto
 
S
de Maat
 
MP
Sonneveld
 
P
Bertina
 
RM
Leebeek
 
FW
Microparticle-associated tissue factor activity and venous thrombosis in multiple myeloma.
Thromb Haemost
2011
, vol. 
105
 
1
(pg. 
14
-
20
)
95
Cesarman-Maus
 
G
Braggio
 
E
Maldonado
 
H
Fonseca
 
R
Absence of tissue factor expression by neoplastic plasma cells in multiple myeloma.
Leukemia
2012
, vol. 
26
 
7
(pg. 
1671
-
1674
)
96
Rautou
 
PE
Mackman
 
N
Microvesicles as risk markers for venous thrombosis.
Expert Rev Hematol
2013
, vol. 
6
 
1
(pg. 
91
-
101
)
97
Bharthuar
 
A
Khorana
 
AA
Hutson
 
A
, et al. 
 
Circulating microparticle tssue factor, thromboembolism and survival in pancreaticobiliary cancers. Thromb Res. Accepted
98
Johnson
 
GJ
Leis
 
LA
Bach
 
RR
Tissue factor activity of blood mononuclear cells is increased after total knee arthroplasty.
Thromb Haemost
2009
, vol. 
102
 
4
(pg. 
728
-
734
)
Sign in via your Institution