Abstract

Tissue factor (TF) is a transmembrane protein that, in association with its ligand factor VII (FVII)/activated factor VII (FVIIa), activates blood coagulation. TF is highly procoagulant and even very small amounts can activate blood coagulation. Levels of TF–positive extracellular vesicles (EVs) are increased in blood in diseases associated with thrombosis. However, it is challenging to accurately quantify the very low levels of TF in blood. Activity-based assays have higher sensitivity and specificity than antigen-based assays. Many anti-human TF antibodies have been generated but they differ in their affinity for TF and bind to different epitopes. They can be divided into 2 groups: those that compete with FVII/FVIIa binding to TF, and those that bind to both TF and the TF-FVII/VIIa complex. Commercial enzyme-linked immunosorbent assays are commonly used to measure TF antigen in plasma but have low sensitivity and specificity for the detection of TF antigen in plasma. Flow cytometry is used to measure TF antigen on EVs but also has low sensitivity and specificity. Functional TF activity assays should be performed in the presence and absence of an inhibitory anti-TF antibody to distinguish between TF-dependent and TF-independent FXa generation because FVIIa can activate FX in the absence of TF. TF pathway inhibitor inhibits the TF-FVIIa complex and reduces TF activity of isolated EVs. Two commercial assays are available for the measurement of TF activity of EVs isolated from human plasma. Measurement of TF activity of EVs isolated from plasma may be a useful biomarker of thrombotic risk in different diseases.

In this review, we describe the different assays used to measure tissue factor (TF) antigen and activity in human plasma, as well as in extracellular vesicles (EVs) isolated from human plasma. We discuss the strengths and weaknesses of the different assays and some of the challenges and pitfalls in measuring TF in plasma and EVs. Most studies use a single method to measure the level of TF and do not independently validate results using a different method.

A fundamental scientific principle is to use negative and positive controls in experiments. However, most investigators do not use these controls when measuring TF in plasma using different assays. Negative and positive controls can be easily made by preparing plasma of blood of healthy volunteers (negative control) or by preparing plasma from whole blood from healthy volunteers stimulated with lipopolysaccharide (LPS) for 5 hours, which induces TF expression in monocytes and the subsequent release of TF-positive EVs.1 In this review, we provide recommendations on the best methods that should be used to measure TF in EVs isolated from human plasma.

TF

TF is a transmembrane glycoprotein that binds plasma factor VII (FVII)/activated factor VII (FVIIa).2 It was designated CD142.3 FVII/FVIIa binds to TF with high affinity. TF localizes FVII/FVIIa to the membrane and functions as a cofactor for FVIIa by inducing an allosteric change in FVIIa that increases its catalytic activity and by providing a substrate binding site (exosite) for FX (Figure 1). TF initiates the coagulation protease cascade by first binding FVII, which is rapidly converted to the active protease FVIIa to form the extrinsic Xase complex (TF-FVIIa; Figure 1). FX then binds to the TF-FVIIa complex and is activated to FXa (Figure 1). The TF-FVIIa complex also activates FIX.2 Amplification of the coagulation protease cascade is mediated by the intrinsic Xase complex (FVIIIa-FIXa). The TF-FVIIa complex is rapidly inhibited by TF pathway inhibitor (TFPI) after the generation of FXa.4 FXa binds to Kunitz domain 2 of TFPI and then the TFPI-FXa complex binds to FVIIa in the TF-FVIIa-FXa complex via Kunitz domain 1 of TFPI to form a quaternary complex (Figure 2).

Figure 1.

Initiation of the coagulation protease cascade. The coagulation protease cascade is initiated by binding of FVII to TF. FVII is rapidly converted to FVIIa that then cleaves FX to FXa. FXa is the favored substrate for the TF-FVIIa complex but this complex can also activate FIX. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 1.

Initiation of the coagulation protease cascade. The coagulation protease cascade is initiated by binding of FVII to TF. FVII is rapidly converted to FVIIa that then cleaves FX to FXa. FXa is the favored substrate for the TF-FVIIa complex but this complex can also activate FIX. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal
Figure 2.

Inhibition of the coagulation protease cascade. TFPI binds to FXa via the Kunitz (K) 2 domain and then this bimolecular complex binds to FVIIa in the TF-FVIIa complex via the K1 domain to inhibit its activity. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 2.

Inhibition of the coagulation protease cascade. TFPI binds to FXa via the Kunitz (K) 2 domain and then this bimolecular complex binds to FVIIa in the TF-FVIIa complex via the K1 domain to inhibit its activity. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

There are 2 naturally occurring isoforms of TF: full-length TF (flTF) and alternatively spliced TF (asTF; Figure 3). Human flTF is a 263 amino acid polypeptide that contains 3 domains: an extracellular domain, a transmembrane domain, and a cytoplasmic domain (Figure 3). The extracellular domain is composed of 2 fibronectin type 3 domains (modules; Figure 3).5 The N-terminal and C-terminal modules bind FVII whereas the C-terminal module binds the substrates FX and FIX (Figure 1).6 asTF is generated by alternative splicing of the TF messenger RNA that results in expression of a protein in which part of the C-terminal module, the transmembrane domain, and the cytoplasmic domain are removed (Figure 3).7 asTF is soluble and has essentially no procoagulant activity because it has lost the substrate binding site in the C-terminal module.8,9 Several groups have expressed the extracellular domain in the absence of the transmembrane and cytoplasmic domains. This protein is referred to as soluble TF (sTF; Figure 3). One study showed that sTF has only 4% of the procoagulant activity of flTF.10 At present, there are no reports of sTF being generated in vivo.

Figure 3.

TF isoforms. flTF is a 263–amino acid polypeptide that is divided into 3 domains: an extracellular domain, a transmembrane domain, and a cytoplasmic domain. The extracellular domain is composed of 2 fibronectin type 2–like domains. There are 3 N-linked glycosylation sites (N) and 2 disulfide bonds (S-S) in the extracellular domain. asTF contains the first 167 amino acids of TF spliced to 38 different amino acids at the C-terminus. The extracellular domain without the transmembrane domain is a soluble protein (sTF). Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 3.

TF isoforms. flTF is a 263–amino acid polypeptide that is divided into 3 domains: an extracellular domain, a transmembrane domain, and a cytoplasmic domain. The extracellular domain is composed of 2 fibronectin type 2–like domains. There are 3 N-linked glycosylation sites (N) and 2 disulfide bonds (S-S) in the extracellular domain. asTF contains the first 167 amino acids of TF spliced to 38 different amino acids at the C-terminus. The extracellular domain without the transmembrane domain is a soluble protein (sTF). Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

TF expression

High levels of TF are expressed by cells surrounding blood vessels and at body surfaces, such as adventitial fibroblasts, pericytes, and keratinocytes, where it acts as a hemostatic envelope to activate the coagulation protease cascade after vessel injury.11 In 1999, it was reported that human native blood contains a small amount of TF that could drive thrombosis in ex vivo models.12 This TF was called “blood-borne” TF and may be derived, in part, from circulating monocytes, which were reported to express low levels of TF.13 However, a significant issue with measuring TF in blood is the quality of the blood draw and which tube is used for analysis. One study measured levels of fibrinopeptide A in sequential blood samples from healthy volunteers after discarding the first 2.5 mL.14 Levels of fibrinopeptide A serially decreased in 3 sequential samples from 5 healthy volunteers. We recommend discarding the first tube of blood because it is more likely to contain TF from the vessel wall compared with subsequent tubes. At present, it is unclear how much of the apparent TF reported in blood samples is from the blood itself vs TF from the vein wall.

TF-positive EVs

flTF is released from cells on EVs.15 In vitro studies show that TF-positive EVs are released from cancer cells, such as pancreatic cancer cells, and activated nonmalignant cells, such as LPS-stimulated monocytes (Figure 4). Importantly, increased levels of TF-positive EVs are present in the circulation of patients with diseases associated with activation of coagulation, disseminated intravascular coagulation, and thrombosis. These include patients with cancer, endotoxemia and sepsis, and viral infections, including severe acute respiratory syndrome coronavirus 2.15,16 Similar to human diseases, we and others have shown that endotoxemic mice and mice bearing pancreatic tumors have elevated levels of TF-positive EVs in the circulation.17-19 Most, if not all, active TF in plasma is present as flTF on EVs.

Figure 4.

TF-positive EVs. LPS-stimulated monocytes express TF and release TF-positive EVs. Tumor cells also constitutively release TF-positive EVs. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 4.

TF-positive EVs. LPS-stimulated monocytes express TF and release TF-positive EVs. Tumor cells also constitutively release TF-positive EVs. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Anti-TF antibodies

Antibodies are useful tools to detect proteins in different biological samples. The guidelines for the use of antibodies from the National Institutes of Health recommend providing the dilution, source, and how the antibodies were validated.20 There are many commercial and noncommercial polyclonal and monoclonal antibodies against human TF. It is important to note that these antibodies have different affinities for TF. In general, those generated by academic laboratories are more carefully characterized compared with those generated by commercial companies. Some monoclonal antibodies, such as HTF-1 and 10H10, were generated in academic laboratories but are sold by commercial companies.

Anti-human TF monoclonal antibodies can be divided into 2 groups based on where they bind to human TF (Figure 5). Antibodies in group 1 bind to the N-terminal module (epitopes amino acids 1-94) and compete with FVII/FVIIa binding to TF. This group includes IIID8, VD8, VIC12, 7G11, HTF-1, 9C3, 6B4, and SBTF-121-24 (F. Dignat-George, written communication, May 2024). Antibodies in group 2 bind to the C-terminal module (epitopes amino acids 136-214) and most compete with binding of the substrates FX and FIX to TF. This group includes IVC6, VIC7, VD10, D3, 5G6, 5G9, and 11D12.21-25 10H10 belongs to group 2 but does not inhibit TF activity.24 Antibodies in this group can bind to either TF or the TF-FVII/FVIIa complex. It was originally thought that antibodies in group 2, such as 5G9, D3, and cH36, could be used to prevent TF-dependent thrombosis because they do not have to compete with FVII/FVIIa for binding to TF.25-28 However, these antibodies were associated with increased bleeding.

Figure 5.

Epitopes of different anti-TF antibodies. FVII binds to the N- and C-modules of TF whereas FX binds to the C-module. Anti-TF antibodies can be divided into 2 groups: group 1 antibodies compete with FVII/FVIIa binding to TF via epitopes between amino acids 1 and 94; group 2 antibodies bind to TF (epitopes amino acids 136-214) as well as TF bound with FVII/FVIIa and all except 10H10 inhibit binding of FX and FIX. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 5.

Epitopes of different anti-TF antibodies. FVII binds to the N- and C-modules of TF whereas FX binds to the C-module. Anti-TF antibodies can be divided into 2 groups: group 1 antibodies compete with FVII/FVIIa binding to TF via epitopes between amino acids 1 and 94; group 2 antibodies bind to TF (epitopes amino acids 136-214) as well as TF bound with FVII/FVIIa and all except 10H10 inhibit binding of FX and FIX. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

There are very few monoclonal antibodies against mouse TF.29,30 The most commonly used antibody against mouse TF is a rat anti-mouse TF monoclonal antibody from Genentech, called 1H1.30 

One method to evaluate an antibody is to determine whether it detects its target on a western blot. However, this does not guarantee that it is suitable for other applications, such as flow cytometry (FCM) and immunohistochemistry, because the epitope may not be preserved in western blotting. In addition, an antibody may detect its target in a western blot but may have off-target effects in other assays. The need for validating antibodies in common research applications was highlighted by the International Working Group for Antibody Validation.31 Several recommendations were made for western blotting, including the elimination or reduction of the target protein and testing multiple antibodies. We found that 4 commercial anti-human TF antibodies, including 2 monoclonal antibodies (HTF-1 and 10H10) detected human TF in a western blot.32 All these antibodies were specific because they only detected TF in a cell lysate from a human pancreatic cancer cell line HPAF-II. Another study found that 5 different anti-human TF monoclonal antibodies (10H0, 9C3, 5G9, 6B4, and VD8) detected TF in a western blot.33 In contrast, only 2 of 5 commercial antibodies detected mouse TF in a western blot using a cell lysate from a mouse pancreatic cancer cell line.32 

There are several major challenges with the immunological detection of TF antigen in plasma. First, TF is present at very low levels compared with other proteins in plasma, such as cytokines. Only very small amounts of TF are needed to initiate the coagulation protease cascade because of amplification of the cascade. Significant amounts of TF are only present in the blood under pathological conditions, such as cancer, sepsis, or viral infections. Furthermore, the level of TF in plasma is often below the detection limit of most assays even under pathological conditions. Secondly, the majority of TF on EVs in plasma will be bound with FVII/FVIIa because of its high affinity and this will interfere with binding of anti-TF antibodies in group 1 (Figure 5). For instance, we find significant TF activity of EVs isolated from LPS-stimulated blood in the absence of exogenous FVIIa, which is consistent with the notion that FVII/FVIIa is already bound to TF (A. Sachetto and N. Mackman, unpublished data, October 2023). This is generally not considered by investigators when choosing antibodies for immunological detection of TF because these antibodies are evaluated in cell-based systems that do not contain FVII/FVIIa. Because FVII/FVIIa binding to TF is calcium dependent, addition of a calcium-chelating agent, such as EDTA, should disrupt binding of FVII/FVIIa to TF and increase binding of group 1 antibodies.34,35 Thirdly, plasma is complex and contains many proteins and multivalent substances, such as heterophilic antibodies, that can generate nonspecific signals independent of the target protein by bridging assay antibodies.36 It is possible that levels of these heterophilic antibodies increase in some disease states.

flTF is the major isoform of TF in plasma but small levels of asTF may also be present (Figure 6). In addition, it is also possible that sTF may be generated under certain disease conditions (Figure 6). In contrast to the multiple forms of TF that may be present in plasma, the only form of TF on isolated EVs is flTF (Figure 6). Therefore, the level of TF antigen in plasma may not correlate with the level of active TF because asTF and sTF have much lower procoagulant activity than flTF. This is important because many studies often use the level of TF antigen in plasma and EVs as a biomarker of thrombotic risk in different diseases.

Figure 6.

Isoforms of TF in plasma and EVs. Plasma contains flTF on EVs and asTF. It is possible that plasma may also contain sTF. In contrast, isolated EVs only contain flTF. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 6.

Isoforms of TF in plasma and EVs. Plasma contains flTF on EVs and asTF. It is possible that plasma may also contain sTF. In contrast, isolated EVs only contain flTF. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

There are several techniques that have been developed to the measure level of TF antigen in plasma and EVs isolated from plasma.

ELISAs

Enzyme-linked immunosorbent assays (ELISAs) are commonly used to measure proteins in plasma. Although several groups developed in-house ELISAs for human TF,37-40 most investigators use commercial ELISAs. We found 49 different human TF ELISAs sold by 20 different companies. IMUBIND Tissue Factor ELISA (BioMedica Diagnostics, catalog no. 845 [IMUBIND]) and Tissue Factor Quantikine ELISA (R&D Systems, catalog no. DCF300 [Quantikine]) are the most frequently used ELISAs in the literature.36 Concerns have been raised about the ability of commercial assays to accurately quantitate TF antigen levels in human plasma.38,41-43 One study used an in-house double anti-TF monoclonal antibody fluorescent-based immunoassay to detect TF in plasma from 72 healthy individuals.38 They found that 57 (79%) had TF antigen values of <94 pg/mL (the detection limit of the assay), 6 (7%) had values between 94 and 235 pg/mL, and 10 (14%) had a nonspecific signal that was higher than the specific signal.

Table 1 shows some of the recent studies that have used the IMUBIND and Quantikine ELISAs for the measurement of TF antigen in human plasma in healthy individuals and in different diseases. It is striking that IMUBIND gives high values and a large range of values for TF antigen in plasma from healthy individuals (62-215 pg/mL, n = 11; Table 1). The manufacturer of IMUBIND states that “a normal range for human plasma remains to be established” despite being sold for many years. In contrast, Quantikine gives lower values and a smaller range for TF antigen in plasma from healthy individuals (23-110, n = 9); Table 1). Quantikine states that the value for citrated plasma from healthy individuals is 33.3 ± 6.6 pg/mL. The different values reported in the different studies are likely due to differences in preanalytical variables, such as plasma preparation. Although these studies report statistically significant increases in TF antigen in the different diseases, it is a concern that in most of these studies values for the disease group overlap with values from the healthy controls. In addition, no other method was used to measure TF.

Table 1.

Studies measuring TF antigen in plasma using IMUBIND and Quantikine ELISAs

DiseaseKitHealthy group TF antigen (pg/mL)Disease group TF antigen (pg/mL)Data typeP valueReference
Leukemia IMUBIND 117 ± 19.2 273 ± 90 Mean ± SD <.01 44  
Coronary artery disease IMUBIND 187.3 ± 108.7 303.6 ± 134.1 Mean ± SD <.05 45  
Peripheral artery disease IMUBIND 158 ± 60 275 ± 122 Mean ± SD <.0001 46  
Ischemic heart disease IMUBIND 142 (18-262) 179.2 (51.2-380) (stable)
215 (56.8-834.3) (unstable) 
Median (IQR) <.0001
<.0001 
47  
Coronary artery disease IMUBIND 76 (32-108) 116 (92-150) (stable) Median (IQR) <.01 48  
   146 (104-188) (unstable)  <.001  
Glomerulonephritis IMUBIND 144 ± 73.9 283 ± 145 Mean ± SD .009 49  
Non–small cell lung cancer IMUBIND 124.1 ± 14.79 334.9 ± 95.44 Mean ± SD .02 50  
Obstructive sleep apnea IMUBIND 42.83 ± 14.18 66.78 ± 41.59 Mean ± SD .005 51  
Aneurysm IMUBIND 62 ± 20 134 ± 54 (AAA) Mean ± SD <.001 52  
   91 ± 30 (IAA/atherosclerotic lesion)  <.008  
Cancer IMUBIND 215.6 ± 55.1 265.2 ± 124.3 Mean ± SD .014 53  
Breast cancer IMUBIND 115.35 (92.65-166.81) 539.93 (400.94-710.40) Median (IQR) .0001 54  
 IMUBIND 110.9 ± 70.97  Mean ± SD  36  
Leukemia IMUBIND  172.2 ± 58.9 Mean ± SD  36  
Pancreatic adenocarcinoma Quantikine 52.4 (28.4-72.4) 63.8 (32.7-112) (stage I-III)
82.8 (45.6-141) (metastatic) 
Mean (range) NS
<.001 
55  
Pancreatic adenocarcinoma Quantikine 39.1 (16.8-87.3) 47.2 (0.0-224.8) (without CAT) Median (range) .009 33  
   56.0 (37.6-318.7) (with CAT)  .002  
Multiple sclerosis Quantikine 110.85 ± 56.31 94.06 ± 55.83 (remitting)
45.03 ± 37.66 (relapsing) 
Mean ± SD <.001 56  
 Quantikine 45.01 ± 12.10  Mean ± SD  36  
Leukemia Quantikine  99.30 ± 78.66 Mean ± SD  36  
DiseaseKitHealthy group TF antigen (pg/mL)Disease group TF antigen (pg/mL)Data typeP valueReference
Leukemia IMUBIND 117 ± 19.2 273 ± 90 Mean ± SD <.01 44  
Coronary artery disease IMUBIND 187.3 ± 108.7 303.6 ± 134.1 Mean ± SD <.05 45  
Peripheral artery disease IMUBIND 158 ± 60 275 ± 122 Mean ± SD <.0001 46  
Ischemic heart disease IMUBIND 142 (18-262) 179.2 (51.2-380) (stable)
215 (56.8-834.3) (unstable) 
Median (IQR) <.0001
<.0001 
47  
Coronary artery disease IMUBIND 76 (32-108) 116 (92-150) (stable) Median (IQR) <.01 48  
   146 (104-188) (unstable)  <.001  
Glomerulonephritis IMUBIND 144 ± 73.9 283 ± 145 Mean ± SD .009 49  
Non–small cell lung cancer IMUBIND 124.1 ± 14.79 334.9 ± 95.44 Mean ± SD .02 50  
Obstructive sleep apnea IMUBIND 42.83 ± 14.18 66.78 ± 41.59 Mean ± SD .005 51  
Aneurysm IMUBIND 62 ± 20 134 ± 54 (AAA) Mean ± SD <.001 52  
   91 ± 30 (IAA/atherosclerotic lesion)  <.008  
Cancer IMUBIND 215.6 ± 55.1 265.2 ± 124.3 Mean ± SD .014 53  
Breast cancer IMUBIND 115.35 (92.65-166.81) 539.93 (400.94-710.40) Median (IQR) .0001 54  
 IMUBIND 110.9 ± 70.97  Mean ± SD  36  
Leukemia IMUBIND  172.2 ± 58.9 Mean ± SD  36  
Pancreatic adenocarcinoma Quantikine 52.4 (28.4-72.4) 63.8 (32.7-112) (stage I-III)
82.8 (45.6-141) (metastatic) 
Mean (range) NS
<.001 
55  
Pancreatic adenocarcinoma Quantikine 39.1 (16.8-87.3) 47.2 (0.0-224.8) (without CAT) Median (range) .009 33  
   56.0 (37.6-318.7) (with CAT)  .002  
Multiple sclerosis Quantikine 110.85 ± 56.31 94.06 ± 55.83 (remitting)
45.03 ± 37.66 (relapsing) 
Mean ± SD <.001 56  
 Quantikine 45.01 ± 12.10  Mean ± SD  36  
Leukemia Quantikine  99.30 ± 78.66 Mean ± SD  36  

AAA, abdominal aortic aneurysm; CAT, cancer-associated thrombosis; IAA, inflammatory aortic aneurysm; IQR, interquartile range; NS, nonsignificant; SD, standard deviation.

A study evaluated the ability of 4 commercial ELISAs (IMUBIND, Quantikine, Zymutest [Hyphen BioMed], and Tissue Factor [CD142] Human ELISA [Abcam, catalog no. ab108903]) to detect TF expressed by a variety of different human cancer cell lines.33 The level of TF detected by all the ELISAs except CD142 correlated with the signal from western blotting. We evaluated the ability of 4 commercial ELISAs (IMUBIND, Quantikine, CD142, and Human Tissue Factor SimpleStep ELISA [Abcam, catalog no. ab220653]) to detect recombinant human TF (12.5-100 pg/mL), recombinant human TF spiked into plasma (12.5-100 pg/mL), low levels of endogenous TF in LPS-stimulated human whole blood, and high levels of endogenous TF in plasma from patients with acute leukemia.36 The level of TF was measured using an EV TF activity assay. All 4 ELISAs stated that they could be used to measure TF in plasma with a limit of detection ranging from 0.69 to 3.6 pg/mL. Consistent with the aforementioned study,33 the CD142 ELISA failed to detect recombinant TF whereas the SimpleStep ELISA detected recombinant TF but failed to detect recombinant TF spiked into plasma. The Quantikine and IMUBIND ELISAs detected recombinant TF spiked into plasma but did not detect low levels of TF in plasma from LPS-treated blood. However, Quantikine but not IMUBIND detected TF in plasma from patients with acute leukemia. We also found that both ELISAs detected TF in EVs isolated from the plasma of patients with leukemia with high levels of EV TF activity, but only Quantikine detected TF in EV-depleted plasma. At present, the source of the TF signal in EV-depleted plasma is unclear.

In conclusion, commercial ELISAs can be used to measure high levels of TF in cell lysates and, in rare cases, of high levels of TF in plasma from patients, such as patients with acute leukemia. However, in general, they were not able to detect low levels of TF antigen in plasma. We do not recommend using commercial ELISAs for measuring TF antigen in human plasma.

We found 37 different mouse TF ELISAs sold by 15 different companies. Mouse Coagulation Factor III/Tissue Factor DuoSet ELISA (R&D Systems, catalog no. Dy3178-05) is the most frequently used ELISA, 5 papers published between 2018 and 2023. We evaluated the ability of 4 commercial mouse TF ELISAs to detect recombinant TF (16-1000 pg/mL), recombinant TF spiked into plasma (16-1000 pg/mL), TF in a cell lysate of a TF-expressing cell line, and endogenous TF-positive EVs in plasma from endotoxemic mice.57,58 The Mouse Tissue Factor ELISA (MyBioSource, catalog no. MBS2512143) and Mouse Tissue Factor Competitive ELISA (MyBioSource, catalog no. MBS722558) failed to detect recombinant mouse TF. The Mouse Tissue Factor SimpleStep ELISA (Abcam, catalog no. ab214091) and the Mouse Coagulation Factor III/Tissue Factor DuoSet ELISA (R&D Systems, catalog no. Dy3178-05) detected recombinant mouse TF and TF in cell lysates but not TF in plasma from endotoxemic mice. These results indicate that, similar to human TF ELISAs, commercial mouse TF ELISAs can detect high levels of TF in cell lysate but cannot detect low levels of TF in plasma.

PLA

The proximity ligation assay (PLA) is a sensitive method to measure levels of proteins in plasma. It uses 2 antibodies against the target protein with attached complementary oligonucleotides that anneal when in close proximity to form double-stranded DNA, which can be amplified by quantitative polymerase chain reaction. One study used a nonconventional PLA strategy that used a single polyclonal anti-human TF antibody that was separately labeled with the complementary oligonucleotides to measure TF in plasma.59 Interestingly, the assay detected recombinant TF spiked into plasma but failed to detect TF-positive EVs in plasma from LPS-stimulated whole blood. OLINK Proteomics (Uppsala, Sweden) uses PLA to measure levels of proteins in plasma, including TF. The advantages of the assay are that it only requires a small amount of plasma and can measure many different proteins in the same sample. However, the disadvantages of the assay are that there is no information on the anti-TF antibodies used in the assay and the results are presented as normalized protein expression rather than absolute values. This means that the results cannot be compared with other antigen-based assays.

Some studies have used OLINK to identify plasma proteins, including TF, that are biomarkers of cardiovascular disease.60,61 Other studies have used OLINK to show that levels of TF in plasma are increased in patients with coronavirus disease 2019 (COVID-19).62-64 

FCM

FCM is typically used to analyze cells in solution. TF expression on cells is detected using anti-TF antibodies conjugated to different fluorophores, which include phycoerythrin (PE; yellow-orange), fluorescein isothiocyanate (FITC; green), and allophycocyanin (far-red). However, conjugation of antibodies with fluorophores can decrease their avidity. There are several factors to consider when choosing an anti-TF antibody to detect TF using FCM. It is preferable to know the affinity, the binding epitope on TF, and the specificity of the antibody. One method to validate an anti-TF antibody is to determine whether it binds to LPS-stimulated monocytes. One study analyzed the ability of different anti-TF monoclonal antibodies to bind to unstimulated and LPS-stimulated monocytes.65 As expected, 3 antibodies (HTF-1, VD8, and 10H10) bound to TF on LPS-stimulated monocytes but not to unstimulated monocytes. However, 2 antibodies (VIC7 and CLB/TF-5) bound nonspecifically to unstimulated monocytes. In contrast, another study found that VIC7 bound to LPS-stimulated monocytes but not unstimulated monocytes.3 Recently, we showed that 3 anti-TF monoclonal antibodies (5G9, VIC12, and IIID8) bound to TF on LPS-stimulated monocytes.66 

There are 2 major challenges with measuring the level of TF on EVs in plasma or isolated from plasma. First, EVs are very small (∼10 times smaller than cells) and need specialized flow cytometers for detection. Secondly, EVs bind ∼100 times less antibodies compared with a cell. Thirdly, compared with other receptors, such as CD41 that has an abundance of 200 molecules per platelet-derived EV,67 the number of TF molecules per EV is very low, making it difficult to detect. Table 2 shows some of the studies that have used FCM to measure the level of TF-positive EVs in plasma from patients with different diseases. A variety of commercial labeled anti-TF antibodies were used in these studies. Of 17 studies, 53% used HTF-1, 24% used VD8, 6% used VIC7, 6% used IID8, and 11% used other antibodies. VIC7 is the only group 2 antibody used in these studies that binds to both TF and the TF-FVII/FVIIa complex.

Table 2.

Studies measuring TF using FCM

Disease/treatmentSample typeTF antibodyResultsReference
Cancer Plasma CD142-PE (HTF-1), BD Higher levels of TF-positive EVs in samples from patients with cancer compared with controls. Higher levels of TF-positive EVs in samples from patients with cancer with VTE compared with without VTE 68  
Atorvastatin treatment Plasma CD142-PE (HTF-1), BD Higher levels of TF-positive EVs during active treatment compared with nontreatment 69  
Deep venous thrombosis EV CD142-FITC (VIC7), BioMedica Diagnostics Higher levels of TF-positive EVs in samples from patients with VTE compared with controls 70  
Statin therapy EV CD142-FITC (VD8), BioMedica Diagnostics Statin therapy decreased the levels of TF-positive EVs 71  
Lung cancer Plasma CD142-FITC, BioMedica Diagnostics Levels of TF-positive EVs predicted distant metastasis 72  
Cardiovascular risk EV CD142-FITC (VD8), BioMedica Diagnostics Levels of TF-positive EVs correlated with lipid-rich atherosclerotic plaques 73  
Cardiovascular risk EV CD142-FITC (VD8), BioMedica Diagnostics Levels of TF-positive EVs were higher in patients with high cardiovascular risk than patients without risk 74  
Type 2 diabetes EV CD142-PE (HTF-1), BD Levels of TF-positive EVs were higher in patients with poor glycemic control than patients with good glycemic control 75  
COVID-19 EV CD142-BV421 (HTF-1), BD Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 76  
HIV EV CD142-APC (NY2), BioLegend No difference in levels of TF-positive EVs between patients infected with HIV and uninfected controls 77  
COVID-19 Plasma CD142-PE (HTF-1), BD No difference in levels of TF-positive EVs between nonpregnant woman, healthy pregnant woman, and pregnant woman with COVID-19 78  
COVID-19 Plasma CD142-FITC, BioMedica Diagnostics Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 79  
COVID-19 EV CD142-BV421 (HTF-1), BD Levels of TF-positive EVs are higher in small EVs than in large EVs 80  
COVID-19 EV CD142-FITC (VD8), BioMedica Diagnostics and CD142-PE (HTF-1), BD Comparison of 2 anti-TF antibodies and fluorochrome-to-protein ratio in the detection of TF on EVs by FCM 81  
COVID-19 Plasma CD142-PE (HTF-1), eBioscience Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 82  
Cirrhosis and bacterial infection Plasma CD142 (IIID8), BioMedica Diagnostics and Alexa Fluor 750 (Sigma-Aldrich) Patients with cirrhosis and bacterial infection had higher levels of TF-positive EVs than patients with cirrhosis without infection 83  
Colorectal cancer Plasma CD142-PE (HTF-1), eBioscience Levels of TF-positive EVs higher in patients with cancer than in controls 84  
Disease/treatmentSample typeTF antibodyResultsReference
Cancer Plasma CD142-PE (HTF-1), BD Higher levels of TF-positive EVs in samples from patients with cancer compared with controls. Higher levels of TF-positive EVs in samples from patients with cancer with VTE compared with without VTE 68  
Atorvastatin treatment Plasma CD142-PE (HTF-1), BD Higher levels of TF-positive EVs during active treatment compared with nontreatment 69  
Deep venous thrombosis EV CD142-FITC (VIC7), BioMedica Diagnostics Higher levels of TF-positive EVs in samples from patients with VTE compared with controls 70  
Statin therapy EV CD142-FITC (VD8), BioMedica Diagnostics Statin therapy decreased the levels of TF-positive EVs 71  
Lung cancer Plasma CD142-FITC, BioMedica Diagnostics Levels of TF-positive EVs predicted distant metastasis 72  
Cardiovascular risk EV CD142-FITC (VD8), BioMedica Diagnostics Levels of TF-positive EVs correlated with lipid-rich atherosclerotic plaques 73  
Cardiovascular risk EV CD142-FITC (VD8), BioMedica Diagnostics Levels of TF-positive EVs were higher in patients with high cardiovascular risk than patients without risk 74  
Type 2 diabetes EV CD142-PE (HTF-1), BD Levels of TF-positive EVs were higher in patients with poor glycemic control than patients with good glycemic control 75  
COVID-19 EV CD142-BV421 (HTF-1), BD Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 76  
HIV EV CD142-APC (NY2), BioLegend No difference in levels of TF-positive EVs between patients infected with HIV and uninfected controls 77  
COVID-19 Plasma CD142-PE (HTF-1), BD No difference in levels of TF-positive EVs between nonpregnant woman, healthy pregnant woman, and pregnant woman with COVID-19 78  
COVID-19 Plasma CD142-FITC, BioMedica Diagnostics Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 79  
COVID-19 EV CD142-BV421 (HTF-1), BD Levels of TF-positive EVs are higher in small EVs than in large EVs 80  
COVID-19 EV CD142-FITC (VD8), BioMedica Diagnostics and CD142-PE (HTF-1), BD Comparison of 2 anti-TF antibodies and fluorochrome-to-protein ratio in the detection of TF on EVs by FCM 81  
COVID-19 Plasma CD142-PE (HTF-1), eBioscience Levels of TF-positive EVs higher in patients with COVID-19 than in healthy controls 82  
Cirrhosis and bacterial infection Plasma CD142 (IIID8), BioMedica Diagnostics and Alexa Fluor 750 (Sigma-Aldrich) Patients with cirrhosis and bacterial infection had higher levels of TF-positive EVs than patients with cirrhosis without infection 83  
Colorectal cancer Plasma CD142-PE (HTF-1), eBioscience Levels of TF-positive EVs higher in patients with cancer than in controls 84  

APC, allophycocyanin; VTE, venous thromboembolism.

A recent study evaluated the ability of 2 anti-TF antibodies (VD8-FITC and HTF-1-PE) to detect TF expression on LPS-stimulated monocytes and TF-positive EVs released from LPS-stimulated monocytes, mesenchymal stems cells, and in plasma from patients with COVID-19.81 As expected, both antibodies detected TF expression on LPS-stimulated monocytes. However, the study found that VD8-FITC but not HTF-1-PE detected TF on EVs. VD8 and HTF-1 belong to group 1 anti-TF antibodies and compete with FVII/FVIIa for binding to TF. However, the specific epitopes are different (the epitope for VD8 is amino acids 1-25 whereas the epitope for HTF-1 is amino acid 94 and 76) and VD8 appears to have a higher affinity than HTF-1. Furthermore, the fluorochrome-to-protein ratio for the anti-TF–fluorochrome conjugates affected the ability of the antibody to detect TF on EVs but not TF on LPS-stimulated monocytes. A higher ratio was associated with a higher percentage detection of TF-positive EVs (7.6% vs 1.1% with ratios of 7.7:1 and 5.2:1, respectively, for detection of TF-positive EVs from LPS-stimulated monocytes). These results indicate that the choice of anti-TF antibody and the fluorochrome-to-protein ratio affects the ability of anti-TF antibodies to detect TF on EVs.

The MACSPlex Exosome Kit (Miltenyi Biotec) quantifies EVs by capturing them onto different fluorescently labeled beads using specific antibodies. This system has been used by 1 group to measure levels of TF-positive EVs in plasma from patients with COVID-19.85,86 In contrast, we found that the MACSPlex Exosome Kit failed to detect TF-positive EVs isolated from plasma from LPS-stimulated whole blood.87 We also noted that the TF signal was below the threshold of detection of the assay and >100-times lower than the signal for platelet-derived CD41+ EVs.

Exoview

The ExoView R100 system (Unchained Labs, Pleasanton, CA) is a chip-based system that uses printed antibodies against cell surface proteins to capture EVs.88 We evaluated the ability of ExoView to capture TF-positive EVs from various sources.66 We found that the anti-TF monoclonal antibody 5G9 (group 2) captured more TF-positive EVs from the culture supernatant of the human pancreatic cancer cell line BxPC-3 than HTF-1 (group 1), which suggests that it has a higher affinity for TF because there is no FVII/FVIIa present. Similarly, 5G9 captured more TF-positive EVs present in plasma from LPS-stimulated blood compared with HTF-1, which may also be because of the ability of 5G9 to bind to both TF and the TF-FVII/FVIIa complex. The TF-positive EVs were detected using the anti-TF antibody IIID8 labeled with Alexa Fluor-647. However, the ExoView system was not sensitive enough to detect TF-positive EVs in plasma from patients with ovarian cancer and patients with trauma who had increased levels of EV TF activity.

Activity-based assays are more sensitive than antigen-based assays for the detection of TF. This is important because there are very low levels of TF in plasma. Various methods have been used to detect TF activity in plasma and EVs but the most common method is to measure FXa generation by the TF-FVIIa complex. In this 2-stage assay, FVII or FVIIa and FX are added to the sample containing TF and incubated at 37°C for a period of time to allow the generation of FXa, and then the amount of FXa is determined using a chromogenic substrate.1 However, FVIIa can cleave FX to FXa independently of TF. One study showed that the concentration of FVIIa directly correlates with the TF-independent FXa generation.89 Therefore, we strongly recommend that all TF activity assays are performed with and without an inhibitory anti-TF antibody to distinguish between TF-dependent and TF-independent FXa generation.

Plasma-based TF activity assays

There are 3 commercial assays that claim to measure TF activity in plasma (Actichrome Tissue Factor, catalog no. 846, Sekisui Diagnostics, LLC, Lexington, MA; tissue factor activity assay kit, catalog no. ab109006, Abcam Inc, Boston, MA; and AssaySense human tissue factor chromogenic assay kit, catalog no. CT1002b, AssayPro, St. Charles, MO). These kits use either FVII or FVIIa. There are several concerns with these assays. First, plasma contains endogenous FVII/FVIIa and FX that may interfere with the assay (Figure 7). Second, plasma contains TFPI that will inhibit the TF-FVIIa complex (Figure 7). Third, no anti-TF antibody is used to distinguish between TF-dependent and TF-independent FXa generation.90 Finally, the color of the plasma may interfere with detection of the yellow chromophore. Despite these concerns there are several publications that have used these kits to measure TF activity in plasma.53,91-100 One study measured the level of TF activity in plasma in hospitalized patients with COVID-19 with elevated D-dimer before and after treatment with the TF-FVIIa inhibitor recombinant nematode anticoagulant protein c2.100 Interestingly, there was a 20.3% reduction in TF activity in patients with COVID-19 treated with recombinant nematode anticoagulant protein c2 whereas no decrease was observed in patients treated with heparin. One study measured TF activity in plasma in the presence of an inhibitory anti-TFPI antibody.101 We do not recommend using plasma-based assays to measure the level of TF activity in plasma.

Figure 7.

Measurement of TF activity in plasma and isolated EVs. In plasma-based activity assays, exogenous FVIIa and FX are added to plasma that already contains FVII and FX. FVII or FVIIa and FX are added to isolated EVs to determine TF activity. Plasma contains TFPI that can inhibit the TF-FVIIa complex and reduce the amount of FXa generated. Isolated EVs have a small amount of TFPI that is carried over from plasma during the isolation of the EVs. FXa cleavage of a chromogenic peptide substrate leads to the release of a chromophore. Professional illustration by Patrick Lane, ScEYEnce Studios.

Figure 7.

Measurement of TF activity in plasma and isolated EVs. In plasma-based activity assays, exogenous FVIIa and FX are added to plasma that already contains FVII and FX. FVII or FVIIa and FX are added to isolated EVs to determine TF activity. Plasma contains TFPI that can inhibit the TF-FVIIa complex and reduce the amount of FXa generated. Isolated EVs have a small amount of TFPI that is carried over from plasma during the isolation of the EVs. FXa cleavage of a chromogenic peptide substrate leads to the release of a chromophore. Professional illustration by Patrick Lane, ScEYEnce Studios.

Close modal

Measurement of TF activity of isolated EVs using FXa generation

The TF activity of isolated EVs can be measured using different readouts, such as FXa generation, thrombin generation, fibrin generation, and clot formation.39,41,102-106 We, and another group, described EV TF activity assays in the late 2000s that used centrifugation to isolate EVs from plasma.39,102 These assays have been used to show that the level of EV TF activity is increased in a variety of thrombotic diseases, including sepsis, cancer, and viral infections.107 The Chapel Hill assay has been improved by (1) increasing the time and speed of the centrifugation to isolate the EVs, (2) using an inhibitory antibody SBTF-1 that is more potent than HTF-1, and (3) using FVII instead of FVIIa.108 The epitope for the binding of SBTF-1 to TF is the same as VD8 (amino acids 1-25; Dignat-George, personal communication). Finally, a recent study used immunomagnetic separation (IMS) beads that were conjugated with anti-CD29 and anti-CD59 antibodies to capture EVs from plasma instead of isolating them by centrifugation.109 TF activity of the isolated EVs was measured using a 2-stage FXa generation assay. The IMS-based assay had superior reproducibility with no loss of sensitivity or specificity compared with the isolation of EVs using centrifugation. Table 3 shows some of the recent studies that have used a FXa generation assay with an anti-TF antibody to measure the level of TF activity of EVs isolated from plasma. It is notable that elevated levels of EV TF activity were shown to be associated with thrombosis and mortality in patients with COVID-19.16,110 

Table 3.

Studies measuring TF activity of EVs using a FXa generation assay

Disease/treatmentSampleResultsReference
Pancreatic adenocarcinoma PPP Levels of TF activity was higher in patients with cancer with VTE than those without VTE, and were associated with metastatic disease and short survival 111  
Ovarian cancer PPP No association between levels of EV TF activity and VTE in patients with cancer 112  
Cancer PPP Levels of EV TF activity were increased in patients with cancer compared with patients without cancer who were severely ill and healthy controls. Levels of EV TF activity were associated with mortality 113  
Lung cancer PPP No difference of levels of EV TF activity between patients with cancer with or without low–molecular weight heparin treatment 114  
Lupus PPP No difference in levels of EV TF activity between patients with lupus anticoagulants with history of thrombosis and healthy controls 115  
Hantavirus cardiopulmonary syndrome Plasma Levels of EV TF activity were increased in patients with HCPS compared with healthy controls 116  
Multiple myeloma PFP Levels of EV TF activity were increased in patients with multiple myeloma compared with controls 117  
Pancreatic and colorectal cancer PPP Two of 13 patients with pancreatic cancer had elevated levels of EV TF activity. No patient with colorectal cancer had elevated levels of EV TF activity 118  
Hemorrhagic fever with renal syndrome Plasma Levels of EV TF activity were transiently increased during HFRS and patients with DIC showed higher levels of EV TF activity than patients without DIC 119  
HIV  Levels of EV TF activity were associated with carotid artery plaque in patients with HIV 120  
Cancer Plasma No association between the levels of EV TF activity and VTE 121  
Cancer PPP Levels of EV TF activity were increased in patients with cancer compared with healthy controls 122  
COVID-19 PPP Levels of EV TF activity were increased in patients with COVID-19 compared with controls, and were associated with disease severity and mortality 110  
COVID-19 PFP Levels of EV TF activity were increased in patients with severe COVID-19 and were associated with an increased thrombotic risk 16  
COVID-19 and sepsis PPP Levels of EV TF activity were higher in ICU patients with COVID-19 than in patients with sepsis 123  
Sepsis PPP Levels of EV TF activity were higher in patients with septic DIC than in healthy controls 124  
Leukemia PFP Levels of EV TF activity were increased in patients with leukemia compared with controls 125  
Prostate cancer PPP Levels of EV TF activity were increased in patients with cancer with DIC compared with patients with cancer without DIC 126  
Lung cancer PFP Levels of EV TF activity were increased in patients with cancer compared with healthy controls. 127  
COVID-19 PFP Levels of EV TF activity were higher in patients with COVID-19 than in healthy controls 128  
COVID-19 PFP Levels of EV TF activity were higher in small EVs than in large EVs from patients with COVID-19 80  
Sepsis, pancreatic cancer, and COVID-19 PFP and PPP Levels of EV TF activity of small or large EVs were different depending on the disease 129  
Sepsis PPP Levels of EV TF activity were increased in patients with sepsis with DIC compared with patients with sepsis without DIC. Levels of EV TF activity were associated with inflammatory markers and DIC score 130  
Leukemia PPP Levels of EV TF activity were increased in patients with leukemia compared with controls, and were associated with risk of bleeding 131  
Disease/treatmentSampleResultsReference
Pancreatic adenocarcinoma PPP Levels of TF activity was higher in patients with cancer with VTE than those without VTE, and were associated with metastatic disease and short survival 111  
Ovarian cancer PPP No association between levels of EV TF activity and VTE in patients with cancer 112  
Cancer PPP Levels of EV TF activity were increased in patients with cancer compared with patients without cancer who were severely ill and healthy controls. Levels of EV TF activity were associated with mortality 113  
Lung cancer PPP No difference of levels of EV TF activity between patients with cancer with or without low–molecular weight heparin treatment 114  
Lupus PPP No difference in levels of EV TF activity between patients with lupus anticoagulants with history of thrombosis and healthy controls 115  
Hantavirus cardiopulmonary syndrome Plasma Levels of EV TF activity were increased in patients with HCPS compared with healthy controls 116  
Multiple myeloma PFP Levels of EV TF activity were increased in patients with multiple myeloma compared with controls 117  
Pancreatic and colorectal cancer PPP Two of 13 patients with pancreatic cancer had elevated levels of EV TF activity. No patient with colorectal cancer had elevated levels of EV TF activity 118  
Hemorrhagic fever with renal syndrome Plasma Levels of EV TF activity were transiently increased during HFRS and patients with DIC showed higher levels of EV TF activity than patients without DIC 119  
HIV  Levels of EV TF activity were associated with carotid artery plaque in patients with HIV 120  
Cancer Plasma No association between the levels of EV TF activity and VTE 121  
Cancer PPP Levels of EV TF activity were increased in patients with cancer compared with healthy controls 122  
COVID-19 PPP Levels of EV TF activity were increased in patients with COVID-19 compared with controls, and were associated with disease severity and mortality 110  
COVID-19 PFP Levels of EV TF activity were increased in patients with severe COVID-19 and were associated with an increased thrombotic risk 16  
COVID-19 and sepsis PPP Levels of EV TF activity were higher in ICU patients with COVID-19 than in patients with sepsis 123  
Sepsis PPP Levels of EV TF activity were higher in patients with septic DIC than in healthy controls 124  
Leukemia PFP Levels of EV TF activity were increased in patients with leukemia compared with controls 125  
Prostate cancer PPP Levels of EV TF activity were increased in patients with cancer with DIC compared with patients with cancer without DIC 126  
Lung cancer PFP Levels of EV TF activity were increased in patients with cancer compared with healthy controls. 127  
COVID-19 PFP Levels of EV TF activity were higher in patients with COVID-19 than in healthy controls 128  
COVID-19 PFP Levels of EV TF activity were higher in small EVs than in large EVs from patients with COVID-19 80  
Sepsis, pancreatic cancer, and COVID-19 PFP and PPP Levels of EV TF activity of small or large EVs were different depending on the disease 129  
Sepsis PPP Levels of EV TF activity were increased in patients with sepsis with DIC compared with patients with sepsis without DIC. Levels of EV TF activity were associated with inflammatory markers and DIC score 130  
Leukemia PPP Levels of EV TF activity were increased in patients with leukemia compared with controls, and were associated with risk of bleeding 131  

DIC, disseminated intravascular coagulation; HCPS, hantavirus cardiopulmonary syndrome; HFRS, hemorrhagic fever with renal syndrome; ICU, intensive care unit; PFP, platelet-free plasma; PPP, platelet-poor plasma; VTE, venous thromboembolism.

Interestingly, incubating IMS-captured EVs from the human pancreatic cancer cell line BxPC-3 with normal plasma reduced TF activity.109 This inhibitory activity was lost with TFPI-deficient plasma or with normal plasma in the presence of an anti-TFPI antibody, indicating that it was due to TFPI. The level of TFPI in EVs isolated from plasma is much lower than the level in plasma (Figure 7). However, we found that addition of an anti-TFPI antibody increased the TF activity of EVs isolated from plasma from LPS-stimulated whole blood. This demonstrated that TFPI present in EVs isolated from plasma by centrifugation inhibited TF-FVIIa–dependent FXa generation.132 

There are 2 commercial assays that can be used to measure TF activity of EVs isolated from plasma (ZYMUPHEN MP-TF assay, catalog no. 521196, Aniara Diagnostica, West Chester Township, OH; and CY-QUANT MV-TF, catalog no. 01514, Diagnostica Stago, Parsippany, NJ). The ZYMUPHEN MP-TF assay captures TF-positive EVs onto microtiter plates using the noninhibitory anti-TF antibody 10H10 that binds to both TF and the TF-FVIIa complex. TF activity is then measured using a standard 2-stage FXa generation assay. A strength of the assay is that it uses a plate-based format, which is more convenient than isolating EVs by centrifugation. Table 4 shows the recent studies that have used the ZYMUPHEN MP-TF assay to measure TF activity of EVs isolated from plasma.

Table 4.

Studies measuring TF in EV in plasma using the ZYMUPHEN MP-TF assay

Disease/treatmentSampleResultsReference
Deep infiltrating endometriosis PFP There was no difference between groups of patients with endometriosis with or without deep infiltration endometriosis 133  
Acute leukemia PFP 4 of 6 patients who had increased EV TF levels had a thrombotic event 134  
Acute ischemic stroke PFP Levels of EV TF activity were higher in patients with stroke than in healthy controls 135  
Cancer PPP Patients with cancer had higher levels of EV TF activity than healthy controls 122  
Immune thrombocytopenia PPP No difference in EV TF activity between patients with immune thrombocytopenia and controls 136  
Cancer and chemotherapy PFP Levels of EV TF activity did not change during chemotherapy but increased after the end of the treatment 137  
Sepsis PFP Levels of EV TF activity correlated with levels of interleukin-6 in plasma 138  
COVID-19 PFP Patients with COVID-19 had higher levels of EV TF activity than healthy controls 139  
Malaria PFP Patients with Plasmodium falciparum infection had increased levels of EV TF activity compared with controls 140  
Disease/treatmentSampleResultsReference
Deep infiltrating endometriosis PFP There was no difference between groups of patients with endometriosis with or without deep infiltration endometriosis 133  
Acute leukemia PFP 4 of 6 patients who had increased EV TF levels had a thrombotic event 134  
Acute ischemic stroke PFP Levels of EV TF activity were higher in patients with stroke than in healthy controls 135  
Cancer PPP Patients with cancer had higher levels of EV TF activity than healthy controls 122  
Immune thrombocytopenia PPP No difference in EV TF activity between patients with immune thrombocytopenia and controls 136  
Cancer and chemotherapy PFP Levels of EV TF activity did not change during chemotherapy but increased after the end of the treatment 137  
Sepsis PFP Levels of EV TF activity correlated with levels of interleukin-6 in plasma 138  
COVID-19 PFP Patients with COVID-19 had higher levels of EV TF activity than healthy controls 139  
Malaria PFP Patients with Plasmodium falciparum infection had increased levels of EV TF activity compared with controls 140  

PFP, platelet-free plasma; PPP, platelet-poor plasma.

We found that the ZYMUPHEN MP-TF assay was less sensitive, less specific, and had a higher background compared with our in-house assay.141 We believe that the ZYMUPHEN MP-TF assay is less efficient because it does not capture all the TF-positive EVs in the sample. The lower specificity and higher background are likely because of the use of a higher concentration of FVIIa compared with our in-house assay (12 nM vs 2.5 nM). CY-QUANT MV-TF was recently introduced by Diagnostica Stago, and isolates EVs from plasma using centrifugation. This has been used in 1 study to measure levels of TF activity in EVs isolated from plasma.142 

The International Society for Thrombosis and Hemostasis Scientific Standardization Subcommittee on Vascular Biology recently organized a multicenter study to compare the specificity, sensitivity, and reproducibility of assays that measure TF antigen and activity of EVs in plasma.142 Twenty-one laboratories participated in the study. The study included 8 groups that used conventional FCM assays and 1 group that used the MACSPlex Exosome Kit. Three groups used HTF-1-PE (Miltenyi Biotec/Becton Dickinson), 2 groups used VD8-FITC BioMedica Diagnostics/American Diagnostic [now Sekisui Diagnostics]), 1 group used H9-FITC (Santa Cruz Biotechnology), and 1 group used unconjugated IIID8 (Sekisui Diagnostics) to detect TF on EVs. HTF-1, VD8, and IIID8 are group 1 antibodies and compete with FVII/FVIIa binding to TF. The study also included 18 groups that ran 16 different functional assays (2 laboratories ran the Chapel Hill assay, and 2 laboratories ran the ZYMUPHEN MP-TF assay). Fourteen of the functional assays used FXa generation (9 with an anti-TF antibody and 5 without), 3 used thrombin generation (2 with an anti-TF antibody and 1 without), and 1 used a fibrin generation assay with an anti-TF antibody (Table 5).

Table 5.

Characteristics of functional assays

LaboratoryReadoutAnti-TF antibody or FVIIaiIsolated EVs
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
10 FXa No Yes 
11 FXa No Yes 
12 FXa No Yes 
13 FXa No Yes 
14 FXa No No 
15 Thrombin Yes No 
16 Thrombin Yes Yes 
17 Thrombin No No 
18 Fibrin Yes No 
LaboratoryReadoutAnti-TF antibody or FVIIaiIsolated EVs
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
FXa Yes Yes 
10 FXa No Yes 
11 FXa No Yes 
12 FXa No Yes 
13 FXa No Yes 
14 FXa No No 
15 Thrombin Yes No 
16 Thrombin Yes Yes 
17 Thrombin No No 
18 Fibrin Yes No 

Three core laboratories (Lacroix, Nieuwland, and Mackman) prepared 4 paired samples: (1) high and low levels of TF-positive and TF-negative EVs from a cancer cell line spiked into EV-depleted plasma, (2) high and low levels of TF-positive EVs from a cancer cell line spiked into EV-depleted plasma, (3) high and low levels of TF-positive EVs from milk spiked into EV-depleted plasma, and (4) plasma prepared from whole blood with and without LPS stimulation. For reference, high and low TF-positive EVs from the cancer line had 2.6 and 0.48 pg/mL of TF activity, respectively, whereas TF-negative EVs had no TF activity. The samples not stimulated and stimulated with LPS had 0 and 1.40 pg/mL TF activity, respectively. Recombinant TF was used as a common standard for the function assays.

To assess the specificity of the assays, the ability of the assays to discriminate between TF-positive EVs and TF-negative EVs was determined. The median ratio for high TF-positive EVs vs high TF-negative EVs for the 8 FCM assays was 1 compared with 10.1 for the 13 functional assays with a denominator of >0 (Table 6). This indicates that activity assays have a higher specificity than an assay that detects TF protein. In addition, the FXa assays that used an anti-TF antibody to distinguish between TF-dependent and TF-independent FXa generation had a higher ratio (23.3) than FXa generation assays that did not use an anti-TF antibody (2.4; Table 6).

Table 6.

Comparison of TF antigen and activity assays for measuring of TF in EVs in plasma samples


Sample
Antigen assayFunctional assays
FCMTotalWith anti-TF antibodyWithout anti-TF antibody
High TF-positive EVs/high TF-negative EV (specificity) 1 (0.5-1.5) n = 8 10.1 (0.9-39) n = 13 23.3 (6.7-39) n = 5 2.4 (1.3-8.9) n = 4 
High TF-positive EVs/low TF-positive EVs (sensitivity) 1.6 (0.7-2.9) n = 8 2.4 (0.8-5.9) n = 16 2.9 (1.9-5.9) n = 9 1.1 (0.9-4.9) n = 5 
With LPS stimulation/without LPS stimulation (sensitivity) 2.3 (1-4.1) n = 8 6.2 (0.9-36.6) n = 4 6.4 (4-26.1) n = 7 2.6 (1.6-32.9) n = 4 

Sample
Antigen assayFunctional assays
FCMTotalWith anti-TF antibodyWithout anti-TF antibody
High TF-positive EVs/high TF-negative EV (specificity) 1 (0.5-1.5) n = 8 10.1 (0.9-39) n = 13 23.3 (6.7-39) n = 5 2.4 (1.3-8.9) n = 4 
High TF-positive EVs/low TF-positive EVs (sensitivity) 1.6 (0.7-2.9) n = 8 2.4 (0.8-5.9) n = 16 2.9 (1.9-5.9) n = 9 1.1 (0.9-4.9) n = 5 
With LPS stimulation/without LPS stimulation (sensitivity) 2.3 (1-4.1) n = 8 6.2 (0.9-36.6) n = 4 6.4 (4-26.1) n = 7 2.6 (1.6-32.9) n = 4 

Results expressed as median (range); only includes data from assays with a denominator of >0; data are from Bonifay et al.142 

To assess the sensitivity of the assays, the ability of the assays to discriminate between samples with high TF-positive EVs and low TF-positive EVs was determined. The ratio for the high TF-positive EVs vs low TF-positive EVs was higher for the functional assays (2.3) than the FCM assays (1.6; Table 6). Similarly, the ratio for LPS-stimulated samples vs unstimulated samples was higher for the functional assays (6.2) than for the FCM assays (2.3; Table 6). This again indicates that activity assays are more sensitive compared with an antigen assay. Inclusion of an anti-TF antibody in the FXa generation assays also increased the sensitivity of the function assay (6.4 vs 2.6; Table 6). Similar results were observed with the thrombin generation assay.142 

Anti-TF antibodies have different abilities to detect TF on EVs by FCM that may depend on the affinity of the antibody and the fluorochrome-to-protein ratio. Activity-based assays are more sensitive and specific compared with antigen-based FCM for the measurement of TF on EVs isolated from plasma. Functional assays should use an anti-TF antibody to distinguish TF-dependent and TF-independent activity.

The authors thank Yohei Hisada and Sierra Archibald for reading the manuscript and Mac Monroe for helpful suggestions.

This work was supported by National Institutes of Health National Heart, Lung, and Blood Institute grant R35HL155657 (N.M.) and the John C. Parker professorship (N.M.).

Contribution: N.M. wrote the manuscript; and N.M. and A.T.A.S. edited and revised the manuscript.

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

Correspondence: Nigel Mackman, Department of Medicine, University of North Carolina at Chapel Hill, 116 Manning Dr, 8004B Mary Ellen Jones Bldg, Chapel Hill, NC 27599; email: nmackman@med.unc.edu.

1.
Hisada
Y
,
Mackman
N
.
Measurement of tissue factor activity in extracellular vesicles from human plasma samples
.
Res Pract Thromb Haemost
.
2019
;
3
(
1
):
44
-
48
.
2.
Grover
SP
,
Mackman
N
.
Tissue factor: an essential mediator of hemostasis and trigger of thrombosis
.
Arterioscler Thromb Vasc Biol
.
2018
;
38
(
4
):
709
-
725
.
3.
Morrissey
J
,
Agis
H
,
Albrecht
S
, et al
. CD142 (Tissue Factor) Workshop Panel report.
Leucocyte Typing VI: White Cell Differentiation Antigens, Kishimoto et al
.
Garland Publishing
;
1997
:
742
-
746
.
4.
Wood
JP
,
Ellery
PER
,
Maroney
SA
,
Mast
AE
.
Biology of tissue factor pathway inhibitor
.
Blood
.
2014
;
123
(
19
):
2934
-
2943
.
5.
Bazan
JF
.
Structural design and molecular evolution of a cytokine receptor superfamily
.
Proc Natl Acad Sci U S A
.
1990
;
87
(
18
):
6934
-
6938
.
6.
Mann
KG
,
Krudysz-Amblo
J
,
Butenas
S
.
Tissue factor controversies
.
Thromb Res
.
2012
;
129
(
suppl 2
):
S5
-
S7
.
7.
Srinivasan
R
,
Bogdanov
VY
.
Alternatively spliced tissue factor: discovery, insights, clinical implications
.
Front Biosci
.
2011
;
16
(
8
):
3061
-
3071
.
8.
Censarek
P
,
Bobbe
A
,
Grandoch
M
,
Schrör
K
,
Weber
AA
.
Alternatively spliced human tissue factor (asHTF) is not pro-coagulant
.
Thromb Haemost
.
2007
;
97
(
1
):
11
-
14
.
9.
Mackman
N
.
Alternatively spliced tissue factor – one cut too many?
.
Thromb Haemost
.
2007
;
97
(
1
):
5
-
8
.
10.
Waxman
E
,
Ross
JB
,
Laue
TM
, et al
.
Tissue factor and its extracellular soluble domain: the relationship between intermolecular association with factor VIIa and enzymatic activity of the complex
.
Biochemistry
.
1992
;
31
(
16
):
3998
-
4003
.
11.
Drake
TA
,
Morrissey
JH
,
Edgington
TS
.
Selective cellular expression of tissue factor in human tissues. Implications for disorders of hemostasis and thrombosis
.
Am J Pathol
.
1989
;
134
(
5
):
1087
-
1097
.
12.
Giesen
PLA
,
Rauch
U
,
Bohrmann
B
, et al
.
Blood-borne tissue factor: another view of thrombosis
.
Proc Natl Acad Sci U S A
.
1999
;
96
(
5
):
2311
-
2315
.
13.
Osterud
B
,
Breimo
ES
,
Olsen
JO
.
Blood borne tissue factor revisited
.
Thromb Res
.
2008
;
122
(
3
):
432
-
434
.
14.
Woodhams
BJ
,
Kernoff
PB
.
Rapid radioimmunoassay for fibrinopeptide A in human plasma
.
Thromb Res
.
1981
;
22
(
4
):
407
-
416
.
15.
Hisada
Y
,
Sachetto
ATA
,
Mackman
N
.
Circulating tissue factor-positive extracellular vesicles and their association with thrombosis in different diseases
.
Immunol Rev
.
2022
;
312
(
1
):
61
-
75
.
16.
Guervilly
C
,
Bonifay
A
,
Burtey
S
, et al
.
Dissemination of extreme levels of extracellular vesicles: tissue factor activity in patients with severe COVID-19
.
Blood Adv
.
2021
;
5
(
3
):
628
-
634
.
17.
Wang
J-G
,
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
.
18.
Wang
J-G
,
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
.
19.
Davila
M
,
Amirkhosravi
A
,
Coll
E
, et al
.
Tissue factor-bearing microparticles derived from tumor cells: impact on coagulation activation
.
J Thromb Haemost
.
2008
;
6
(
9
):
1517
-
1524
.
20.
National Institutes of Health
.
Principles and Guidelines for Reporting Preclinical Research
. Accessed 3 June 2024. https://grants.nih.gov/policy/reproducibility/principles-guidelines-reporting-preclinical-research.htm.
21.
Magdolen
V
,
Albrecht
S
,
Kotzsch
M
, et al
.
Immunological and functional analyses of the extracellular domain of human tissue factor
.
Biol Chem
.
1998
;
379
(
2
):
157
-
165
.
22.
Kirchhofer
D
,
Moran
P
,
Chiang
N
, et al
.
Epitope location on tissue factor determines the anticoagulant potency of monoclonal anti-tissue factor antibodies
.
Thromb Haemost
.
2000
;
84
(
6
):
1072
-
1081
.
23.
Fiore
MM
,
Neuenschwander
PF
,
Morrissey
JH
.
An unusual antibody that blocks tissue factor/factor VIIa function by inhibiting cleavage only of macromolecular substrates
.
Blood
.
1992
;
80
(
12
):
3127
-
3134
.
24.
Morrissey
JH
,
Fair
DS
,
Edgington
TS
.
Monoclonal antibody analysis of purified and cell-associated tissue factor
.
Thromb Res
.
1988
;
52
(
3
):
247
-
261
.
25.
Huang
M
,
Syed
R
,
Stura
EA
, et al
.
The mechanism of an inhibitory antibody on TF-initiated blood coagulation revealed by the crystal structures of human tissue factor, Fab 5G9 and TF.G9 complex
.
J Mol Biol
.
1998
;
275
(
5
):
873
-
894
.
26.
Presta
L
,
Sims
P
,
Meng
YG
, et al
.
Generation of a humanized, high affinity anti-tissue factor antibody for use as a novel antithrombotic therapeutic
.
Thromb Haemost
.
2001
;
85
(
3
):
379
-
389
.
27.
Taylor
FB
,
Chang
A
,
Ruf
W
, et al
.
Lethal E. coli septic shock is prevented by blocking tissue factor with monoclonal antibody
.
Circ Shock
.
1991
;
33
(
3
):
127
-
134
.
28.
Morrow
DA
,
Murphy
SA
,
Mccabe
CH
,
Mackman
N
,
Wong
HC
,
Antman
EM
.
Potent inhibition of thrombin with a monoclonal antibody against tissue factor (Sunol-cH36): results of the PROXIMATE-TIMI 27 trial
.
Eur Heart J
.
2005
;
26
(
7
):
682
-
688
.
29.
Furlan-Freguia
C
,
Marchese
P
,
Gruber
A
,
Ruggeri
ZM
,
Ruf
W
.
P2X7 receptor signaling contributes to tissue factor–dependent thrombosis in mice
.
J Clin Invest
.
2011
;
121
(
7
):
2932
-
2944
.
30.
Kirchhofer
D
,
Moran
P
,
Bullens
S
,
Peale
F
,
Bunting
S
.
A monoclonal antibody that inhibits mouse tissue factor function
.
J Thromb Haemost
.
2005
;
3
(
5
):
1098
-
1099
.
31.
Uhlen
M
,
Bandrowski
A
,
Carr
S
, et al
.
A proposal for validation of antibodies
.
Nat Methods
.
2016
;
13
(
10
):
823
-
827
.
32.
Rosell
A
,
Moser
B
,
Hisada
Y
, et al
.
Evaluation of different commercial antibodies for their ability to detect human and mouse tissue factor by western blotting
.
Res Pract Thromb Haemost
.
2020
;
4
(
6
):
1013
-
1023
.
33.
Kobayashi
S
,
Koizume
S
,
Takahashi
T
, et al
.
Tissue factor and its procoagulant activity on cancer-associated thromboembolism in pancreatic cancer
.
Cancer Sci
.
2021
;
112
(
11
):
4679
-
4691
.
34.
Ploplis
VA
,
Edgington
TS
,
Fair
DS
.
Initiation of the extrinsic pathway of coagulation. Association of factor VIIa with a cell line expressing tissue factor
.
J Biol Chem
.
1987
;
262
(
20
):
9503
-
9508
.
35.
Carlsson
K
,
Osterlund
M
,
Persson
E
,
Freskgård
PO
,
Carlsson
U
,
Svensson
M
.
Site-directed fluorescence probing to dissect the calcium-dependent association between soluble tissue factor and factor VIIa domains
.
Biochim Biophys Acta
.
2003
;
1648
(
1-2
):
12
-
16
.
36.
Sachetto
ATA
,
Archibald
SJ
,
Bhatia
R
,
Monroe
D
,
Hisada
Y
,
Mackman
N
.
Evaluation of four commercial ELISAs to measure tissue factor in human plasma
.
Res Pract Thromb Haemost
.
2023
;
7
(
3
):
100133
.
37.
Koyama
T
,
Nishida
K
,
Ohdama
S
, et al
.
Determination of plasma tissue factor antigen and its clinical significance
.
Br J Haematol
.
1994
;
87
(
2
):
343
-
347
.
38.
Parhami-Seren
B
,
Butenas
S
,
Krudysz-Amblo
J
,
Mann
KG
.
Immunologic quantitation of tissue factors
.
J Thromb Haemost
.
2006
;
4
(
8
):
1747
-
1755
.
39.
Khorana
AA
,
Francis
CW
,
Menzies
KE
, et al
.
Plasma tissue factor may be predictive of venous thromboembolism in pancreatic cancer
.
J Thromb Haemost
.
2008
;
6
(
11
):
1983
-
1985
.
40.
Albrecht
S
,
Luther
T
,
Grossmann
H
,
Flössel
C
,
Kotzsch
M
,
Müller
M
.
An ELISA for tissue factor using monoclonal antibodies
.
Blood Coagul Fibrinolysis
.
1992
;
3
(
3
):
263
-
270
.
41.
Claussen
C
,
Rausch
A-V
,
Lezius
S
, et al
.
Microvesicle-associated tissue factor procoagulant activity for the preoperative diagnosis of ovarian cancer
.
Thromb Res
.
2016
;
141
:
39
-
48
.
42.
Ay
C
,
Mackman
N
.
Tissue factor: catch me if you can
.
J Clin Oncol
.
2017
;
35
(
10
):
1128
-
1130
.
43.
Mackman
N
,
Sachetto
ATA
,
Hisada
Y
.
Measurement of tissue factor-positive extracellular vesicles in plasma: strengths and weaknesses of current methods
.
Curr Opin Hematol
.
2022
;
29
(
5
):
266
-
274
.
44.
Shimura
M
,
Wada
H
,
Wakita
Y
, et al
.
Plasma tissue factor and tissue factor pathway inhibitor levels in patients with disseminated intravascular coagulation
.
Am J Hematol
.
1997
;
55
(
4
):
169
-
174
.
45.
Kim
HK
,
Song
KS
,
Park
YS
,
Yun
Y-S
,
Shim
WH
.
Changes of plasma tissue factor and tissue factor pathway inhibitor antigen levels and induction of tissue factor expression on the monocytes in coronary artery disease
.
Cardiology
.
2000
;
93
(
1-2
):
31
-
36
.
46.
Blann
AD
,
Amiral
J
,
McCollum
CN
,
Lip
GY
.
Differences in free and total tissue factor pathway inhibitor, and tissue factor in peripheral artery disease compared to healthy controls
.
Atherosclerosis
.
2000
;
152
(
1
):
29
-
34
.
47.
Marcucci
R
,
Prisco
D
,
Brunelli
T
, et al
.
Tissue factor and homocysteine levels in ischemic heart disease are associated with angiographically documented clinical recurrences after coronary angioplasty
.
Thromb Haemost
.
2000
;
83
(
6
):
826
-
832
.
48.
Lindmark
E
,
Wallentin
L
,
Siegbahn
A
.
Blood cell activation, coagulation, and inflammation in men and women with coronary artery disease
.
Thromb Res
.
2001
;
103
(
3
):
249
-
259
.
49.
Naumnik
B
,
Borawski
J
,
Chyczewski
L
,
Pawlak
K
,
Mysliwiec
M
.
Tissue factor and its inhibitor in human non-crescentic glomerulonephritis--immunostaining vs plasma and urinary levels
.
Nephrol Dial Transplant
.
2006
;
21
(
12
):
3450
-
3457
.
50.
Goldin-Lang
P
,
Tran
Q-V
,
Fichtner
I
, et al
.
Tissue factor expression pattern in human non-small cell lung cancer tissues indicate increased blood thrombogenicity and tumor metastasis
.
Oncol Rep
.
2008
;
20
(
1
):
123
-
128
.
51.
El Solh
AA
,
Akinnusi
ME
,
Berim
IG
,
Peter
AM
,
Paasch
LL
,
Szarpa
KR
.
Hemostatic implications of endothelial cell apoptosis in obstructive sleep apnea
.
Sleep Breath
.
2008
;
12
(
4
):
331
-
337
.
52.
Skóra
J
,
Dawiskiba
T
,
Zaleska
P
, et al
.
Prognostic value of tissue factor in patients with abdominal aortic and iliac arterial aneurysms – preliminary study
.
Arch Med Sci
.
2013
;
9(6)
:
1071
-
1077
.
53.
Hernández
C
,
Orbe
J
,
Roncal
C
, et al
.
Tissue factor expressed by microparticles is associated with mortality but not with thrombosis in cancer patients
.
Thromb Haemost
.
2013
;
110
(
3
):
598
-
608
.
54.
Zarychta
E
,
Rhone
P
,
Bielawski
K
, et al
.
Elevated plasma levels of tissue factor as a valuable diagnostic biomarker with relevant efficacy for prediction of breast cancer morbidity
.
J Physiol Pharmacol
.
2018
;
69
(
6
):
921
-
931
.
55.
Talar-Wojnarowska
R
,
Woźniak
M
,
Borkowska
A
,
Cypryk
K
,
Olakowski
M
,
Małecka-Panas
E
.
Procoagulant disorders in patients with newly diagnosed pancreatic adenocarcinoma
.
Medicina
.
2020
;
56
(
12
):
677
.
56.
Koudriavtseva
T
,
Lorenzano
S
,
Cellerino
M
, et al
.
Tissue factor as a potential coagulative/vascular marker in relapsing-remitting multiple sclerosis
.
Front Immunol
.
2023
;
14
:
1226616
.
57.
Sachetto
ATA
,
Mackman
N
.
Evaluation of the ability of commercial enzyme-linked immunosorbent assays to measure mouse tissue factor
.
Res Pract Thromb Haemost
.
2024
;
8
(
1
):
102325
.
58.
Senis
YA
.
Mouse tissue factor enzyme-linked immunosorbent assays: a sensitive issue
.
Res Pract Thromb Haemost
.
2024
;
8
(
3
):
102386
.
59.
Thulin
Å
,
Yan
J
,
Åberg
M
,
Christersson
C
,
Kamali-Moghaddam
M
,
Siegbahn
A
.
Sensitive and specific detection of platelet-derived and tissue factor–positive extracellular vesicles in plasma using solid-phase proximity ligation assay
.
TH Open
.
2018
;
2
(
3
):
e250
-
e260
.
60.
Folkersen
L
,
Fauman
E
,
Sabater-Lleal
M
, et al
.
Mapping of 79 loci for 83 plasma protein biomarkers in cardiovascular disease
.
PLoS Genet
.
2017
;
13
(
4
):
e1006706
.
61.
Folkersen
L
,
Gustafsson
S
,
Wang
Q
, et al
.
Genomic and drug target evaluation of 90 cardiovascular proteins in 30,931 individuals
.
Nat Metab
.
2020
;
2
(
10
):
1135
-
1148
.
62.
Krishnamachary
B
,
Cook
C
,
Kumar
A
,
Spikes
L
,
Chalise
P
,
Dhillon
NK
.
Extracellular vesicle-mediated endothelial apoptosis and EV-associated proteins correlate with COVID-19 disease severity
.
J Extracell Vesicles
.
2021
;
10
(
9
):
e12117
.
63.
Patel
H
,
Ashton
NJ
,
Dobson
RJB
, et al
.
Proteomic blood profiling in mild, severe and critical COVID-19 patients
.
Sci Rep
.
2021
;
11
(
1
):
6357
.
64.
Goonewardena
SN
,
Chen
Q
,
Tate
AM
, et al
.
Monocyte-mediated thrombosis linked to circulating tissue factor and immune paralysis in COVID-19
.
Arterioscler Thromb Vasc Biol
.
2024
;
44
(
5
):
1124
-
1134
.
65.
Basavaraj
MG
,
Olsen
JO
,
Østerud
B
,
Hansen
J-B
.
Differential ability of tissue factor antibody clones on detection of tissue factor in blood cells and microparticles
.
Thromb Res
.
2012
;
130
(
3
):
538
-
546
.
66.
Price
JMJ
,
Hisada
Y
,
Hazeldine
J
, et al
.
Detection of tissue factor–positive extracellular vesicles using the ExoView R100 system
.
Res Pract Thromb Haemost
.
2023
;
7
(
4
):
100177
.
67.
Nolan
JP
,
Jones
JC
.
Detection of platelet vesicles by flow cytometry
.
Platelets
.
2017
;
28
(
3
):
256
-
262
.
68.
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
;
127
(
5
):
473
-
477
.
69.
Mobarrez
F
,
Egberg
N
,
Antovic
J
,
Bröijersen
A
,
Jörneskog
G
,
Wallén
H
.
Release of endothelial microparticles in vivo during atorvastatin treatment; a randomized double-blind placebo-controlled study
.
Thromb Res
.
2012
;
129
(
1
):
95
-
97
.
70.
Ye
R
,
Ye
C
,
Huang
Y
,
Liu
L
,
Wang
S
.
Circulating tissue factor positive microparticles in patients with acute recurrent deep venous thrombosis
.
Thromb Res
.
2012
;
130
(
2
):
253
-
258
.
71.
Suades
R
,
Padró
T
,
Alonso
R
,
Mata
P
,
Badimon
L
.
Lipid-lowering therapy with statins reduces microparticle shedding from endothelium, platelets and inflammatory cells
.
Thromb Haemost
.
2013
;
110
(
2
):
366
-
377
.
72.
Tseng
J-C
,
Chang
L-C
,
Jiang
B-Y
, et al
.
Elevated circulating levels of tissue factor-positive microvesicles are associated with distant metastasis in lung cancer
.
J Cancer Res Clin Oncol
.
2014
;
140
(
1
):
61
-
67
.
73.
Suades
R
,
Padró
T
,
Alonso
R
,
Mata
P
,
Badimon
L
.
High levels of TSP1+/CD142+ platelet-derived microparticles characterise young patients with high cardiovascular risk and subclinical atherosclerosis
.
Thromb Haemost
.
2015
;
114
(
6
):
1310
-
1321
.
74.
Chiva-Blanch
G
,
Crespo
J
,
Suades
R
, et al
.
CD142+/CD61+, CD146+ and CD45+ microparticles predict cardiovascular events in high risk patients following a Mediterranean diet supplemented with nuts
.
Thromb Haemost
.
2016
;
116
(
1
):
103
-
114
.
75.
Giannella
A
,
Ceolotto
G
,
Radu
CM
, et al
.
PAR-4/Ca2+-calpain pathway activation stimulates platelet-derived microparticles in hyperglycemic type 2 diabetes
.
Cardiovasc Diabetol
.
2021
;
20
(
1
):
77
.
76.
Canzano
P
,
Brambilla
M
,
Porro
B
, et al
.
Platelet and endothelial activation as potential mechanisms behind the thrombotic complications of COVID-19 patients
.
JACC Basic Transl Sci
.
2021
;
6
(
3
):
202
-
218
.
77.
Marques de Menezes
EG
,
Deng
X
,
Liu
J
, et al
.
Plasma CD16+ extracellular vesicles associate with carotid artery intima-media thickness in HIV+ adults on combination antiretroviral therapy
.
mBio
.
2022
;
13
(
3
):
e0300521
.
78.
Dangot
A
,
Zavaro
M
,
Bar-Lev
TH
, et al
.
Characterization of extracellular vesicles in COVID-19 infection during pregnancy
.
Front Cell Dev Biol
.
2023
;
11
:
1135821
.
79.
Eichhorn
T
,
Weiss
R
,
Huber
S
, et al
.
Expression of tissue factor and platelet/leukocyte markers on extracellular vesicles reflect platelet–leukocyte interaction in severe COVID-19
.
Int J Mol Sci
.
2023
;
24
(
23
):
16886
.
80.
Brambilla
M
,
Frigerio
R
,
Becchetti
A
, et al
.
Head-to-head comparison of tissue factor-dependent procoagulant potential of small and large extracellular vesicles in healthy subjects and in patients with SARS-CoV-2 infection
.
Biology
.
2023
;
12
(
9
):
1233
.
81.
Weiss
R
,
Mostageer
M
,
Eichhorn
T
, et al
.
The fluorochrome-to-protein ratio is crucial for the flow cytometric detection of tissue factor on extracellular vesicles
.
Sci Rep
.
2024
;
14
(
1
):
6419
.
82.
Barbosa
MS
,
De Lima
F
,
Peachazepi Moraes
CR
, et al
.
Angiopoietin2 is associated with coagulation activation and tissue factor expression in extracellular vesicles in COVID-19
.
Front Med
.
2024
;
11
:
1367544
.
83.
Campello
E
,
Zanetto
A
,
Radu
CM
, et al
.
Profiling plasma alterations of extracellular vesicles in patients with acutely decompensated cirrhosis and bacterial infection
.
Liver Int
.
2024
;
44
(
7
):
1610
-
1623
.
84.
Kriebardis
AG
,
Chardalias
L
,
Damaskos
C
, et al
.
Precision oncology: circulating microvesicles as new biomarkers in a very early stage of colorectal cancer
.
Cancers
.
2024
;
16
(
10
):
1943
.
85.
Balbi
C
,
Burrello
J
,
Bolis
S
, et al
.
Circulating extracellular vesicles are endowed with enhanced procoagulant activity in SARS-CoV-2 infection
.
EBioMedicine
.
2021
;
67
:
103369
.
86.
Burrello
J
,
Caporali
E
,
Gauthier
LG
, et al
.
Risk stratification of patients with SARS-CoV-2 by tissue factor expression in circulating extracellular vesicles
.
Vascul Pharmacol
.
2022
;
145
:
106999
.
87.
Archibald
SJ
,
Hisada
Y
,
Bae-Jump
VL
,
Mackman
N
.
Evaluation of a new bead-based assay to measure levels of human tissue factor antigen in extracellular vesicles in plasma
.
Res Pract Thromb Haemost
.
2022
;
6
(
2
):
e12677
.
88.
Daaboul
GG
,
Gagni
P
,
Benussi
L
, et al
.
Digital detection of exosomes by interferometric imaging
.
Sci Rep
.
2016
;
6
(
1
):
37246
.
89.
Bom
VJJ
,
Bertina
RM
.
The contributions of Ca2+, phospholipids and tissue-factor apoprotein to the activation of human blood-coagulation factor X by activated factor VII
.
Biochem J
.
1990
;
265
(
2
):
327
-
336
.
90.
Bogdanov
VY
,
Cimmino
G
,
Tardos
JG
,
Tunstead
JR
,
Badimon
JJ
.
Assessment of plasma tissue factor activity in patients presenting with coronary artery disease: limitations of a commercial assay
.
J Thromb Haemost
.
2009
;
7
(
5
):
894
-
897
.
91.
Chiva-Blanch
G
,
Laake
K
,
Myhre
P
, et al
.
Platelet-monocyte-derived and tissue factor-carrying circulating microparticles are related to acute myocardial infarction severity
.
PLoS One
.
2017
;
12
(
2
):
e0172558
.
92.
Wang
J
,
Ciaraldi
TP
,
Samad
F
.
Tissue factor expression in obese type 2 diabetic subjects and its regulation by antidiabetic agents
.
J Obes
.
2015
;
2015
:
291209
. 8.
93.
Funderburg
NT
,
Mayne
E
,
Sieg
SF
, et al
.
Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation
.
Blood
.
2010
;
115
(
2
):
161
-
167
.
94.
White
TA
,
Pan
S
,
Witt
TA
,
Simari
RD
.
Murine strain differences in hemostasis and thrombosis and tissue factor pathway inhibitor
.
Thromb Res
.
2010
;
125
(
1
):
84
-
89
.
95.
Campbell
MD
,
Walker
M
,
Ajjan
RA
,
Birch
KM
,
Gonzalez
JT
,
West
DJ
.
An additional bolus of rapid-acting insulin to normalise postprandial cardiovascular risk factors following a high-carbohydrate high-fat meal in patients with type 1 diabetes: a randomised controlled trial
.
Diab Vasc Dis Res
.
2017
;
14
(
4
):
336
-
344
.
96.
Sachetto
ATA
,
Rosa
JG
,
Santoro
ML
.
Rutin (quercetin-3-rutinoside) modulates the hemostatic disturbances and redox imbalance induced by Bothrops jararaca snake venom in mice
.
PLoS Negl Trop Dis
.
2018
;
12
(
10
):
e0006774
.
97.
Yang
M
,
Deng
C
,
Wu
D
, et al
.
The role of mononuclear cell tissue factor and inflammatory cytokines in patients with chronic thromboembolic pulmonary hypertension
.
J Thromb Thrombolysis
.
2016
;
42
(
1
):
38
-
45
.
98.
Puhm
F
,
Allaeys
I
,
Lacasse
E
, et al
.
Platelet activation by SARS-CoV-2 implicates the release of active tissue factor by infected cells
.
Blood Adv
.
2022
;
6
(
12
):
3593
-
3605
.
99.
Yang
X
,
Cheng
X
,
Tang
Y
, et al
.
Bacterial endotoxin activates the coagulation cascade through gasdermin D-dependent phosphatidylserine exposure
.
Immunity
.
2019
;
51
(
6
):
983
-
996.e6
.
100.
Hess
CN
,
Hsia
J
,
Carroll
IA
, et al
.
Novel tissue factor inhibition for thromboprophylaxis in COVID-19: primary results of the ASPEN-COVID-19 trial
.
Arterioscler Thromb Vasc Biol
.
2023
;
43
(
8
):
1572
-
1582
.
101.
Doubre
H
,
Monnet
I
,
Azarian
R
, et al
.
Plasma tissue factor activity in lung cancer patients predicts venous thromboembolism and poor overall survival
.
Res Pract Thromb Haemost
.
2024
;
8
(
2
):
102359
.
102.
Tesselaar
MET
,
Romijn
FPHTM
,
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
;
5
(
3
):
520
-
527
.
103.
Østerud
B
,
Latysheva
N
,
Schoergenhofer
C
,
Jilma
B
,
Hansen
JB
,
Snir
O
.
A rapid, sensitive, and specific assay to measure TF activity based on chromogenic determination of thrombin generation
.
J Thromb Haemost
.
2022
;
20
(
4
):
866
-
876
.
104.
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
;
108
(
1
):
160
-
165
.
105.
Kristensen
SR
,
Nybo
J
.
A sensitive tissue factor activity assay determined by an optimized thrombin generation method
.
PLoS One
.
2023
;
18
(
7
):
e0288918
.
106.
Aras
O
,
Shet
A
,
Bach
RR
, et al
.
Induction of microparticle- and cell-associated intravascular tissue factor in human endotoxemia
.
Blood
.
2004
;
103
(
12
):
4545
-
4553
.
107.
Hisada
Y
,
Alexander
W
,
Kasthuri
R
, et al
.
Measurement of microparticle tissue factor activity in clinical samples: a summary of two tissue factor-dependent FXa generation assays
.
Thromb Res
.
2016
;
139
:
90
-
97
.
108.
Vallier
L
,
Bouriche
T
,
Bonifay
A
, et al
.
Increasing the sensitivity of the human microvesicle tissue factor activity assay
.
Thromb Res
.
2019
;
182
:
64
-
74
.
109.
Franco
C
,
Lacroix
R
,
Vallier
L
, et al
.
A new hybrid immunocapture bioassay with improved reproducibility to measure tissue factor-dependent procoagulant activity of microvesicles from body fluids
.
Thromb Res
.
2020
;
196
:
414
-
424
.
110.
Rosell
A
,
Havervall
S
,
Von Meijenfeldt
F
, et al
.
Patients with COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality—brief report
.
Arterioscler Thromb Vasc Biol
.
2021
;
41
(
2
):
878
-
882
.
111.
Woei-A-Jin
FJSH
,
Tesselaar
MET
,
Garcia Rodriguez
P
,
Romijn
FPHTM
,
Bertina
RM
,
Osanto
S
.
Tissue factor-bearing microparticles and CA19.9: two players in pancreatic cancer-associated thrombosis?
.
Br J Cancer
.
2016
;
115
(
3
):
332
-
338
.
112.
Cohen
JG
,
Prendergast
E
,
Geddings
JE
, et al
.
Evaluation of venous thrombosis and tissue factor in epithelial ovarian cancer
.
Gynecol Oncol
.
2017
;
146
(
1
):
146
-
152
.
113.
Hisada
Y
,
Thålin
C
,
Lundström
S
,
Wallén
H
,
Mackman
N
.
Comparison of microvesicle tissue factor activity in non-cancer severely ill patients and cancer patients
.
Thromb Res
.
2018
;
165
:
1
-
5
.
114.
Gezelius
E
,
Flou Kristensen
A
,
Bendahl
PO
, et al
.
Coagulation biomarkers and prediction of venous thromboembolism and survival in small cell lung cancer: a sub-study of RASTEN - a randomized trial with low molecular weight heparin
.
PLoS One
.
2018
;
13
(
11
):
e0207387
.
115.
Hell
L
,
Ay
C
,
Posch
F
, et al
.
Low extracellular vesicle–associated tissue factor activity in patients with persistent lupus anticoagulant and a history of thrombosis
.
Ann Hematol
.
2019
;
98
(
2
):
313
-
319
.
116.
Tatsumi
K
,
Hisada
Y
,
Connolly
A-MF
,
Buranda
T
,
Mackman
N
.
Patients with severe orthohantavirus cardiopulmonary syndrome due to Sin Nombre Virus infection have increased circulating extracellular vesicle tissue factor and an activated coagulation system
.
Thromb Res
.
2019
;
179
:
31
-
33
.
117.
Nielsen
T
,
Kristensen
SR
,
Gregersen
H
,
Teodorescu
EM
,
Christiansen
G
,
Pedersen
S
.
Extracellular vesicle-associated procoagulant phospholipid and tissue factor activity in multiple myeloma
.
PLoS One
.
2019
;
14
(
1
):
e0210835
.
118.
Kasthuri
RS
,
Hisada
Y
,
Ilich
A
,
Key
NS
,
Mackman
N
.
Effect of chemotherapy and longitudinal analysis of circulating extracellular vesicle tissue factor activity in patients with pancreatic and colorectal cancer
.
Res Pract Thromb Haemost
.
2020
;
4
(
4
):
636
-
643
.
119.
Schmedes
CM
,
Grover
SP
,
Hisada
YM
, et al
.
Circulating extracellular vesicle tissue factor activity during orthohantavirus infection is associated with intravascular coagulation
.
J Infect Dis
.
2020
;
222
(
8
):
1392
-
1399
.
120.
Lin
J
,
Xue
X
,
Anastos
K
, et al
.
Elevated microparticle tissue factor activity is associated with carotid artery plaque in HIV-infected women
.
J Acquir Immune Defic Syndr
.
2019
;
81
(
1
):
36
-
43
.
121.
Reitter
E-M
,
Kaider
A
,
Prager
G
,
Ay
C
,
Pabinger
I
,
Thaler
J
.
Longitudinal analysis of extracellular vesicle-associated tissue factor activity in cancer patients
.
Thromb Res
.
2020
;
195
:
215
-
218
.
122.
Ender
F
,
Freund
A
,
Quecke
T
, et al
.
Tissue factor activity on microvesicles from cancer patients
.
J Cancer Res Clin Oncol
.
2020
;
146
(
2
):
467
-
475
.
123.
Campbell
RA
,
Hisada
Y
,
Denorme
F
, et al
.
Comparison of the coagulopathies associated with COVID-19 and sepsis
.
Res Pract Thromb Haemost
.
2021
;
5
(
4
):
e12525
.
124.
Meng
S
,
Kang
K
,
Fei
D
, et al
.
Preliminary study of microparticle coagulation properties in septic patients with disseminated intravascular coagulation
.
J Int Med Res
.
2021
;
49
(
5
):
3000605211014094
.
125.
Zhao
H
,
Sun
J
,
Yan
L
, et al
.
Tissue factor–bearing microparticles are a link between acute promyelocytic leukemia cells and coagulation activation: a human subject study
.
Ann Hematol
.
2021
;
100
(
6
):
1473
-
1483
.
126.
Hell
L
,
Däullary
T
,
Burghart
V
, et al
.
Extracellular vesicle-associated tissue factor activity in prostate cancer patients with disseminated intravascular coagulation
.
Cancers
.
2021
;
13
(
7
):
1487
.
127.
Pedersen
S
,
Kristensen
AF
,
Falkmer
U
,
Christiansen
G
,
Kristensen
SR
.
Increased activity of procoagulant factors in patients with small cell lung cancer
.
PLoS One
.
2021
;
16
(
7
):
e0253613
.
128.
Henderson
MW
,
Lima
F
,
Moraes
CRP
, et al
.
Contact and intrinsic coagulation pathways are activated and associated with adverse clinical outcomes in COVID-19
.
Blood Adv
.
2022
;
6
(
11
):
3367
-
3377
.
129.
Sachetto
ATA
,
Archibald
SJ
,
Hisada
Y
, et al
.
Tissue factor activity of small and large extracellular vesicles in different diseases
.
Res Pract Thromb Haemost
.
2023
;
7
(
3
):
100124
.
130.
Meng
S
,
Xu
B
,
Yang
W
,
Zhao
M
.
Microparticle-associated tissue factor activity correlates with the inflammatory response in septic disseminated intravascular coagulation patients
.
PeerJ
.
2024
;
12
:
e16636
.
131.
Hisada
Y
,
Archibald
SJ
,
Bansal
K
, et al
.
Biomarkers of bleeding and venous thromboembolism in patients with acute leukemia
.
J Thromb Haemost
.
2024
;
22
(
7
):
1984
-
1996
.
132.
Tanratana
P
,
Sachetto
ATA
,
Mast
AE
,
Mackman
N
.
An anti-tissue factor pathway inhibitor antibody increases tissue factor activity in extracellular vesicles isolated from human plasma
.
Res Pract Thromb Haemost
.
2024
;
8
(
1
):
102275
.
133.
Munrós
J
,
Martínez-Zamora
MA
,
Tàssies
D
, et al
.
Total circulating microparticle levels are increased in patients with deep infiltrating endometriosis
.
Hum Reprod
.
2017
;
32
(
2
):
325
-
331
.
134.
Gheldof
D
,
Haguet
H
,
Dogné
J-M
, et al
.
Procoagulant activity of extracellular vesicles as a potential biomarker for risk of thrombosis and DIC in patients with acute leukaemia
.
J Thromb Thrombolysis
.
2017
;
43
(
2
):
224
-
232
.
135.
Słomka
A
,
Świtońska
M
,
Sinkiewicz
W
,
Żekanowska
E
.
Assessing circulating factor VIIa–antithrombin complexes in acute ischemic stroke: a pilot study
.
Clin Appl Thromb Hemost
.
2017
;
23
(
4
):
351
-
359
.
136.
Garabet
L
,
Ghanima
W
,
Hellum
M
, et al
.
Increased microvesicle-associated thrombin generation in patients with immune thrombocytopenia after initiation of thrombopoietin receptor agonists
.
Platelets
.
2020
;
31
(
3
):
322
-
328
.
137.
Beleva
EA
,
Deneva
TI
,
Stoencheva
SS
,
Grudeva-Popova
ZG
.
Longitudinal dynamics of coagulation and angiogenesis markers in cancer patients during and after chemotherapy
.
Clin Appl Thromb Hemost
.
2021
;
27
:
10760296211056637
.
138.
Brandtner
AK
,
Lehner
GF
,
Pircher
A
,
Feistritzer
C
,
Joannidis
M
.
Differential procoagulatory response of microvascular, arterial and venous endothelial cells upon inflammation in vitro
.
Thromb Res
.
2021
;
205
:
70
-
80
.
139.
Hamali
HA
,
Saboor
M
,
Dobie
G
, et al
.
Procoagulant microvesicles in COVID-19 patients: possible modulators of inflammation and prothrombotic tendency
.
Infect Drug Resist
.
2022
;
15
:
2359
-
2368
.
140.
Madkhali
AM
,
Mobarki
AA
,
Ghzwani
AH
, et al
.
Elevated levels of procoagulant microvesicles and tissue-factor bearing microvesicles in malaria patients
.
Int J Gen Med
.
2023
;
16
:
1205
-
1215
.
141.
Tatsumi
K
,
Antoniak
S
,
Monroe
DM
,
Khorana
AA
,
Mackman
N
;
Subcommittee on Hemostasis and Malignancy of the Scientific and Standardization Committee of the International Society on Thrombosis and Hemostasis
.
Evaluation of a new commercial assay to measure microparticle tissue factor activity in plasma: communication from the SSC of the ISTH
.
J Thromb Haemost
.
2014
;
12
(
11
):
1932
-
1934
.
142.
Bonifay
A
,
Mackman
N
,
Hisada
Y
, et al
.
Comparison of assays measuring extracellular vesicle tissue factor in plasma samples: communication from the ISTH SSC Subcommittee on Vascular Biology
.
J Thromb Haemost
.
Published online 25 June 2024
.

Author notes

Data are available on request from the corresponding author, Nigel Mackman (nmackman@med.unc.edu).