You are on page 1of 12

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/49745919

Venous and arterial thrombosis - Pathogenesis and the rationale for


anticoagulation

Article  in  Thrombosis and Haemostasis · April 2011


DOI: 10.1160/TH10-10-0683 · Source: PubMed

CITATIONS READS

56 5,576

2 authors, including:

Alexander G G Turpie
McMaster University
673 PUBLICATIONS   48,991 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Create new project "GARFIELD-AF" View project

Evaluation of D-dimer in the diagnosis of pulmonary embolism. View project

All content following this page was uploaded by Alexander G G Turpie on 08 January 2016.

The user has requested enhancement of the downloaded file.


586 © Schattauer 2011 Review Article

Venous and arterial thrombosis – pathogenesis and the rationale


for anticoagulation
Alexander G. G. Turpie1; Charles Esmon2
1Department of Medicine, McMaster University, Hamilton, Ontario, Canada; 2Oklahoma Medical Research Foundation, Howard Hughes Medical Institute, and Departments
of Pathology and Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA

Summary effective agents, but also for better clinical management of patients
Thromboembolic disorders are major causes of morbidity and mortality. who require anticoagulation therapy. In recent years, new oral agents
It is well-recognised that the pathogenesis is different for arterial and that target single enzymes of the coagulation cascade have been devel-
venous thrombosis; however, both involve coagulation activation. Anti- oped – some of those are in advanced stages of clinical development.
coagulants are used for the prevention and treatment of a wide variety Based on scientific rationale, both factor Xa and thrombin are viable
of thromboembolic and related conditions. Agents with anti- targets for effective anticoagulation.
inflammatory properties in addition to anticoagulation may be particu-
larly beneficial. Traditional anticoagulants, although effective, are as- Keywords
sociated with certain limitations. Understanding the pathological pro- Anticoagulation, arterial thrombosis, factor Xa, thrombin, venous
cesses associated with thrombosis and the rational target for anti- thrombosis
coagulation is essential, not only for the development of safer and more

Correspondence to: Received: October 28, 2010


Dr. Alexander G. G. Turpie, MD Accepted after minor revision: December 27, 2010
Department of Medicine, McMaster University Prepublished online: January 12, 2011
Hamilton Health Sciences-General Hospital doi:10.1160/TH10-10-0683
237 Barton Street East Thromb Haemost 2011; 105: 586–596
Hamilton, Ontario L8L 2X2, Canada
Tel: +1 416 363 6726, Fax: +1 905 628 9505
E-mail: turpiea@mcmaster.ca

Introduction itself causing thrombosis is crucial, not only for developing more
effective and safer antithrombotic agents, but also for better man-
Haemostasis is the normal physiological response that prevents agement of patients with thromboembolic disorders. Traditional
significant blood loss after vascular injury. Blood vessel injury anticoagulants are associated with limitations such as parenteral
triggers a sequence of responses: vessel constriction to reduce administration with the heparins and the requirement for routine
blood flow; platelet plug formation at the site of trauma involving coagulation monitoring and dose adjustment with the vitamin K
platelet adhesion, activation, and aggregation; and blood coagu- antagonists (5, 6). In recent years, small-molecule, oral agents that
lation involving a complex set of protease reactions. Although hae- target a single enzyme in the coagulation cascade have been devel-
mostasis is essential for survival, inappropriate coagulation can oped. Some of these new oral agents (such as direct factor Xa in-
lead to the formation of a thrombus. Thromboembolic disorders hibitors and direct thrombin inhibitors) have undergone extensive
are major causes of morbidity and mortality. Anticoagulants are evaluation for the prevention and treatment of thromboembolic
used to prevent or treat a wide variety of conditions that involve ar- disorders (7).
terial or venous thrombosis, including the prevention and treat- After a brief description of haemostasis and blood coagulation,
ment of venous thromboembolism (VTE), long-term prevention this article will provide an overview of the pathogenesis of venous
of ischaemic stroke in patients with atrial fibrillation (AF), and sec- and arterial thrombosis, and the rationale for using anticoagulants
ondary prevention of cardiac events in patients with acute coron- in those thromboembolic and related disease conditions. The im-
ary syndrome (ACS) (1–4). pact of the protein C pathway on coagulation and inflammation,
It is well-recognised that the pathogenesis of arterial thrombo- and the potential role of new oral anticoagulants and other anti-
sis differs from that of venous thrombosis, which is reflected by thrombotic agents in the management of thromboembolic dis-
their different management strategies. Understanding the patho- orders will also be discussed.
genic processes that occur in the blood vessel wall and in the blood

Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
Turpie, Esmon: Anticoagulation in thrombosis 587

Haemostasis and blood coagulation Propagation phase

The endothelium of the blood vessel wall plays a crucial role in On the surface of activated platelets, factor XIa activates factor IX.
maintaining the integrity of the vasculature. When the vessel wall is This factor IXa, together with the factor IXa generated by the TF−
damaged or the endothelium is disrupted, collagen in the sub- factor VIIa complex, forms the tenase complex with factor VIIIa.
endothelial matrix becomes exposed to the flowing blood. Within This factor IXa−factor VIIIa complex activates factor X, which
seconds, platelets bind to the exposed collagen, which causes pla- forms the prothrombinase complex with factor Va and produces a
telet activation, degranulation, and aggregation, leading to the burst of thrombin generation.
formation of a primary platelet plug. In conjunction, coagulation
is activated at the site of injury by the exposed subendothelial tissue
factor (TF), resulting in fibrin formation, which stabilises the pla-
telet plug (8, 9). Clot formation
TF is an integral membrane protein that is constitutively ex-
pressed on fibroblasts and pericytes in the adventitia and medial The generated thrombin cleaves soluble fibrinogen to insoluble fi-
smooth muscle cells of the blood vessel wall (8). Under normal brin. Fibrin monomers polymerise to form the fibrin mesh that is
conditions, the endothelium acts as a barrier to separate TF from stabilised and cross-linked by thrombin-activated factor XIIIa
factor VII and factor VIIa in the circulating blood, which prevents (씰Fig. 1) (12, 14).
the initiation of coagulation in the absence of vessel injury. How- Recent evidence suggests that cellular injury can release mRNA
ever, TF is also expressed in many non-vascular cells (such as from the cells, which in turn can activate coagulation by triggering
monocytes) and microparticles in the circulation. This blood- factor XII activation. The physiological relevance of this pathway is
borne TF is thought to participate in the processes of blood coagu- demonstrated by the observation that vessel occlusion in the ferric
lation (8, 10). Coagulation reactions occur on specific cell surfaces chloride model of arterial thrombosis has been reduced by RNAase
such as activated platelets and TF-bearing cells. treatment (15). Cell injury can also release histones that are cytot-
For decades, the coagulation cascade was conceptualised as hav- oxic to cells and elicit fibrin and platelet deposition along with leu-
ing two distinct points of initiation – the extrinsic and intrinsic cocyte accumulation in the tissues. The trigger for these responses
pathways (11). However, it has become clear that these pathways do is probably the release of RNA from the cells and the exposure of
not function in the body as independent systems. An increasing TF (16). The relevance of these observations is supported by the
understanding of the role of different factors and cells involved in finding that the blockade of histone toxicity protects animals from
blood coagulation has led to a cell-based model of coagulation. the lethal responses to endotoxin.
Unlike the older cascade model, the cell-based model represents
more accurately the interaction between coagulation enzymes and
cellular activity that leads to thrombin generation and clot
formation (12). The role of endothelial cells in the control of
blood coagulation
The endothelium of the blood vessel wall plays a crucial role in
Initiation phase maintaining the integrity of the vasculature. By releasing sub-
stances such as nitric oxide, prostacyclin, and the ectonucleotidase
The initiation phase is localised to cells that express TF (8, 12). CD39, the endothelium keeps the platelets in their inactivated state
After binding to factor VIIa, the TF−factor VIIa complex activates and impairs their adhesion in the absence of vessel injury (8, 17).
small amounts of factor IX and factor X. Factor Xa then activates The endothelial cells also produce coagulation inhibitors, includ-
factor V on the TF-bearing cells and forms a complex with factor ing tissue factor pathway inhibitor (TFPI) and heparan sulphate.
Va, which then converts a small amount of prothrombin (factor II) TFPI inhibits the TF−factor VIIa/factor Xa complex (9), thus elim-
to thrombin (factor IIa; 씰Fig. 1) (12, 13). inating the generation of both factor Xa and factor IXa, and in-
hibiting the initiation of coagulation. Free thrombin and factor Xa
are inhibited by antithrombin (AT), and this inhibitory activity of
AT is markedly enhanced by heparan sulphates present on the sur-
Amplification phase face of endothelial cells (18). In addition, free thrombin in the cir-
culation binds to the endothelial surface-bound thrombomodulin
The small amount of thrombin generated on TF-bearing cells am- (TM) and activates the protein C system – a major negative control
plifies coagulation reactions. In addition to activating platelets, the mechanism of coagulation (18, 19).
generated thrombin activates factor V and factor VIII on the pla- The protein C anticoagulant system is initiated when thrombin
telet surface, leading to a burst of thrombin-generating potential. binds TM and forms the thrombin−TM complex, which decreases
Thrombin also activates platelet-bound factor XI (씰Fig. 1) (8, 12). free thrombin levels, thus preventing thrombin from cleaving fi-
brinogen or activating platelets. Furthermore, this complex masks

© Schattauer 2011 Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
588 Turpie, Esmon: Anticoagulation in thrombosis

Figure 1: An overview of blood


coagulation. TF, tissue factor; TFPI, tissue
factor pathway inhibitor; vWF, von Willebrand
factor.

Thrombosis and Haemostasis 105.4/2011 © Schattauer 2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
Turpie, Esmon: Anticoagulation in thrombosis 589

Figure 2: The anticoagulant effect of the


activated protein C system. Thrombin (IIa)
binds to thrombomodulin (TM) and activates
protein C (PC), which is reversibly bound to the
endothelial cell protein C receptor (EPCR). Acti-
vated protein C (APC) binds to protein S (PS)
and inactivates factor Va and factor VIIIa.

Figure 3: The cytoprotective effects of


activated protein C. IL, interleukin; MCP,
monocyte chemoattractant protein; TF, tissue
factor.

the binding sites of thrombin for fibrinogen and the platelet bearing microparticles derived from activated platelets and en-
thrombin receptor (20, 21). Protein C is a vitamin K-dependent dothelial cells (8, 21). Inflammation can trigger a coagulation re-
protein, and is activated after binding to the endothelial protein C sponse in a number of ways, such as inducing TF synthesis in in-
receptor and interacting with the thrombin−TM complex. Acti- travascular cells and stimulating blood-borne TF, increasing pla-
vated protein C (APC), together with its cofactor protein S, inacti- telet numbers and reactivity, causing the exposure of negatively
vates factor Va and factor VIIIa, thereby decreasing the formation charged phospholipids (22), and downregulating the natural anti-
of factor Xa−factor Va (the prothrombinase complex) and factor coagulant systems.
IXa−factor VIIIa (the intrinsic tenase complex), which results in The protein C pathway is particularly sensitive to downregu-
the inhibition of coagulation (9, 21) (씰Fig. 2). The APC system lation by inflammatory responses (22). For example, endotoxin,
also enhances fibrinolysis by inactivating plasminogen activator tumour necrosis factor (TNF)-α and interleukin (IL)-1β can
inhibitor-1 (which inhibits clot lysis or fibrinolysis) (20, 21). In ad- downregulate TM and endothelial protein C receptor – the APC
dition, thrombin within the thrombin−TM complex is more sensi- complex co-factors (23, 24). Acquired protein C deficiency is
tive to inhibition by circulating AT than free thrombin (20). Under prevalent in the majority of septic patients (>85%) and is associ-
normal conditions, natural anticoagulants and the fibrinolytic sys- ated with increased morbidity and mortality in patients with se-
tem ensure that thrombus formation is a controlled and balanced vere sepsis and septic shock (25). On the other hand, APC exhibits
process. Deficiency or any impairment of these systems may result anti-inflammatory activity by decreasing proinflammatory cyto-
in a hypercoagulable state. kines such as TNF-α, IL-1β, IL-6, IL-8, and MCP-1, reducing in-
flammatory mediator-induced TF expression on leucocytes, in-
hibiting neutrophil adhesion to the endothelium, and upregulat-
ing anti-inflammatory mediators (e.g. IL-10) (21, 24). In addition,
Relationship between coagulation and APC can also reduce endothelial cell apoptosis and help maintain
inflammation, and the role of activated endothelial cell barrier functions (21, 24). Accumulating evidence
protein C suggests that the APC system protectively modulates a variety of
disease processes, including diabetic nephropathy (26), stroke
Thrombosis and inflammation share some pathophysiological (27), tumour metastasis (28), multiple sclerosis (29), reperfusion-
processes that involve inflammatory mediators, the activated induced coronary (30), and renal injury (31). These effects are
endothelium, TF expression on monocytes, and circulating TF- thought to be attributed to its cytoprotective properties (씰Fig. 3).

© Schattauer 2011 Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
590 Turpie, Esmon: Anticoagulation in thrombosis

Venous thrombosis found in cancer-associated thrombosis (8). Although surgery or


trauma can directly cause vascular wall injury, and coagulation is
VTE comprises deep-vein thrombosis (DVT) and pulmonary em- activated via the exposed subendothelial TF, deep vessel wall injury
bolism. DVT occurs most often in the large veins of the legs. When is not a common feature in non-surgical patients with DVT.
part of the thrombus breaks away, it can travel to the lungs and dis- Factors that may allow a small, silent thrombus to enlarge, or a
rupt or block the blood flow in a pulmonary artery. Over 150 years new thrombus to develop, include the prolonged impairment of
ago, Rudolph Virchow proposed a triad of causes for venous venous function (37), sustained hypercoagulability (38), and im-
thrombosis: venous stasis, vascular endothelial injury, and hyper- pairment of the endogenous anticoagulant or fibrinolytic systems
coagulability of blood. This triad still applies, with one or more (39, 40). Venous thrombi consist of mostly red blood cells and large
factors participating in the development of DVT (씰Fig. 4). amounts of fibrin. Activation of the coagulation system is the pri-
Venous stasis (or reduced blood flow in the veins) can be caused mary cause of venous thrombosis, and precedes platelet activation
by many medical conditions, including immobility (e.g. hospital and aggregation (17). This explains why anticoagulation therapy
or nursing home confinement), major orthopaedic surgery (e.g. has become the primary management strategy for VTE, and evi-
during and after surgery), or increased venous pressure (e.g. in dence-based guidelines strongly recommend the use of anticoagu-
heart failure). The majority of venous thrombi form in regions lants for the prevention and treatment of VTE (3, 4, 41).
with a slow blood flow (32) and reducing venous stasis in the legs
has been found to reduce the risk of VTE (33). Venous stasis pro-
motes thrombus formation by failing to clear activated coagu-
lation factors quickly from the site of vascular injury. In addition, Arterial thrombosis
local hypoxia and distension of the vessel wall as a result of venous
stasis can activate the endothelium, causing the expression of ad- The primary trigger for arterial thrombosis is the rupture of an
hesion molecule selectins on its surface (17). Accumulating evi- atherosclerotic plaque, causing a complete or partial vessel occlu-
dence suggests that circulating TF-bearing microparticles may play sion. When plaque rupture occurs, its lipid core is exposed to the
important roles in venous thrombosis (8, 17) – similar to that of circulating blood in the arterial lumen. The core area of the plaque
platelets in arterial thrombosis (34). These microparticles attach to contains TF and fragments of collagen, which are highly thrombo-
the activated endothelial cells and transfer TF to them, initiating genic (42, 43). Circulating TF is also increased in patients with car-
coagulation reactions and clot formation (17). An increase in en- diovascular disease and might contribute to thrombosis after
dothelial microparticles and leucocyte activation has been found plaque rupture (44). In the initial stage of plaque rupture, platelets
in patients with VTE (35). are rapidly recruited to the site, followed by aggregation and the re-
It has also been suggested that TF-bearing microparticles may sulting rapid growth of the thrombus. The coagulation cascade is
also contribute to the hypercoagulable state associated with disease also activated at this stage with the formation of thrombin, which
conditions with an increased risk of DVT (such as cancer, inflam- activates platelets. Activated platelets promote further platelet re-
mation, and congestive heart failure) (8, 17, 36). Elevated numbers cruitment, adhesion, aggregation, and activation (44). Thus, an ar-
of tumour-derived TF-bearing microparticles in plasma have been terial thrombus is platelet-rich and exposed to fast flowing blood.

Figure 4: Venous thrombosis. Pathogenic


processes that can trigger venous thrombosis
include: abnormal blood flow (e.g. venous
stasis), vessel wall damage or alterations in the
endothelium, and hypercoagulability of blood.

Thrombosis and Haemostasis 105.4/2011 © Schattauer 2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
Turpie, Esmon: Anticoagulation in thrombosis 591

Figure 5: Arterial thrombosis. An athero-


sclerotic plaque develops through the accumu-
lation of lipid deposits and foam cells. The rup-
ture of an atherosclerotic plaque is the primary
trigger of arterial thrombosis. The core area of
the plaque contains tissue factor and collagen
(highly thrombogenic), which are released into
the lumen of the blood vessel upon rupture.

The fibrin component of the thrombus increases as it extends into likely that LDL lowering may be a primary driver for the reduction
the arterial lumen, although the surface area of the thrombus that in inflammation that contributes to lower cardiovascular risk (50).
is exposed to the blood in the lumen will be covered by activated Cyclooxygenase (COX)-2 is the inducible form of cyclooxyge-
platelets. A loose network of fibrin with large numbers of trapped nase and has been shown to be involved in the proinflammatory re-
red blood cells also form part of the thrombus in the final stage of sponse of vascular tissue, participating in both atherosclerosis and
thrombosis (43) (씰Fig. 5). thrombosis (51). Increased inflammatory activity that involves
ACS is usually the outcome of atherosclerosis and thrombosis COX-2 may predispose patients to cardiovascular events. Recent
in the coronary arteries. Most acute myocardial infarctions are evidence also suggests that altered histone acetylation levels in in-
caused by thrombosis developed on a culprit coronary athero- flammatory processes are associated with atherosclerosis and res-
sclerotic plaque. Thrombosis is also the major initiating factor in tenosis, and histone hyperacetylation may have a protective mech-
unstable angina. Studies have suggested that a new thrombotic cor- anism in vascular injury and remodelling, possibly by reducing the
onary event accounts for 50–70% of sudden deaths caused by is- expression of factors involved in these pathological processes (52).
chaemic heart disease (43). An arterial thrombus that is rich in fi- In acute arterial thrombotic events, drugs that reduce the
brin is often fully occlusive and results in ST-elevated myocardial growth of a thrombus should be administered, with platelets as the
infarction, whereas a platelet-rich arterial thrombus is often par- main target. Although antiplatelet agents are most commonly used
tially occlusive, resulting in unstable angina and non-ST-elevated to prevent the incidence of arterial thrombosis and recurrent is-
myocardial infarction. chaemic events, coagulation is clearly activated after plaque rup-
It is now well recognised that inflammation plays a key role in ture, which provides a mechanistic rationale for anticoagulation
the pathogenesis of coronary artery disease and other manifes- therapy. Clinical evidence has shown that the combination of anti-
tations of atherosclerosis. Blood-borne inflammatory and im- coagulant and antiplatelet therapy is more effective than either
mune cells constitute an important part of an atheroma. Many of treatment alone (53). In addition, based on the role of inflam-
these cells are activated, producing several types of molecules (in- mation in the pathogenesis of ACS, anticoagulants with anti-
cluding inflammatory cytokines and proteases), which can des- inflammatory properties may be beneficial. It has been shown that
tabilise the plaque, resulting in plaque rapture and ACS (45). The the combination of vitamin K antagonists with acetylsalicylic acid
levels of C-reactive protein and IL-6, as well as other inflammatory causes higher bleeding rates (42). New oral anticoagulants have
markers, are elevated in patients with unstable angina and myo- demonstrated effective anticoagulation in some clinical studies.
cardial infarction (24, 45, 46). Interestingly, it has been suggested However, adequate dose-findings studies for the new oral agents to
that some beneficial effects of statins may be partly attributed to be co-administered with antiplatelet drugs in this setting are
their anti-inflammatory properties (47–49). However, results from necessary to ensure that the potential benefit of the combination
a meta-analysis have indicated that most of the anti-inflammatory therapy is not offset by any increase in bleeding complications.
effect of statins is related to the magnitude of low-density
lipoprotein (LDL) reduction, and the potential non-LDL effects of
statins on inflammation are relatively small (50). Nevertheless, it is

© Schattauer 2011 Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
592 Turpie, Esmon: Anticoagulation in thrombosis

Figure 6: Targets for anticoagulation.


Schematic representation of the coagulation
cascade demonstrating the inhibiting effects
of traditional and new anticoagulants on
coagulation factors. AT, antithrombin.

Thrombus formation in atrial fibrillation Targets for anticoagulation


AF is the most common significant cardiac arrhythmia and it in- The coagulation pathway provides many targets for potential anti-
creases the risk of stroke four- to five-fold across all age groups coagulants (씰Fig. 6). Drugs that target the TF−factor VIIa com-
(54). Left atrial thrombus formation in patients with AF fulfils the plex inhibit the initiation phase of coagulation, whereas those that
Virchow’s triad for thrombogenesis, with abnormal blood flow, target factor IXa or factor Xa, or their co-factors (factor VIIIa and
endothelial dysfunction, and a hypercoagulable state (55, 56). factor Va), inhibit the propagation of coagulation. Thrombin in-
Thrombi associated with AF occur most often in the left atrial ap- hibitors inhibit thrombin activities. Direct factor Xa inhibitors and
pendage, and abnormal blood flow is evident in the left atrium. AF direct thrombin inhibitors are in the most advanced stage of clini-
is also associated with markers of coagulation and platelet acti- cal development. These include rivaroxaban, apixaban, edoxaban,
vation that may reflect a systemic hypercoagulable or prothrom- betrixaban (oral, direct factor Xa inhibitors), and dabigatran etex-
botic state (56). For example, systemic fibrinogen and fibrin ilate (an oral, direct thrombin inhibitor). Some of these agents
D-dimer levels are elevated in patients with persistent and paroxy- have already shown promise in providing safe, equivalent, or more
smal AF, indicating the presence of active intravascular thrombo- effective anticoagulation compared with traditional anticoagu-
genesis (55). Both von Willebrand factor and TF are overexpressed lants, without the need for routine coagulation monitoring (7, 57,
in the atrial endothelium in patients with AF who have a history of 58).
cardiogenic thromboembolism (56). In addition, recent evidence
suggests that inflammation and various growth factors may also
play a potential role in driving the prothrombotic state in AF (56).
Based on the existing evidence, the pathogenesis of throm- Direct factor Xa inhibition
boembolism in AF seems to fulfil the Virchow’s triad for thrombo-
genesis. Thus, the thrombus may be a 'venous-type clot'. There- Factor X is a key component of coagulation. As part of the pro-
fore, anticoagulation therapy is an important part of patient man- thrombinase complex, factor Xa catalyses the conversion of pro-
agement strategies and is strongly recommended in evidence- thrombin to thrombin – one molecule of factor Xa results in the
based guidelines (41). Numerous clinical studies have demon- generation of approximately 1,000 thrombin molecules (59, 60).
strated that anticoagulants can effectively reduce the incidence of Whenever there is vessel damage, exposing subendothelial TF, fac-
ischaemic stroke in patients with AF. tor Xa is generated after the formation of the TF−factor VIIa com-
plex. Thus, factor Xa inhibition would be desirable in both arterial
and venous thrombosis. Moreover, inhibiting factor Xa may also be

Thrombosis and Haemostasis 105.4/2011 © Schattauer 2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
Turpie, Esmon: Anticoagulation in thrombosis 593

important in preventing platelet activation, deposition and re- Direct thrombin inhibition
cruitment at the sites of vessel injury by preventing thrombin gen-
eration. In addition, direct factor Xa inhibition does not affect the Thrombin has multiple effects on coagulation, including the con-
activity of preformed thrombin, which may allow the function of version of fibrinogen to fibrin and the activation of factor XIII and
the existing thrombin to continue (such as activating protein C) platelet-bound factor XI. Thrombin is the most potent activator of
(61–63). platelets, and also induces the synthesis and/or secretion of adeno-
Inhibition of factor Xa with new, oral, direct inhibitors may sine diphosphate (ADP), endothelial platelet-activating factor, se-
have potential advantages over indirect factor Xa inhibitors. These rotonin, and thromboxane A2 (20). In addition, thrombin am-
include their ability to inhibit factor Xa within the prothrombinase plifies its own generation by activating factor VIII and factor V –
complex, which the AT-dependent, indirect factor Xa inhibitors key co-factors for the tenase complex and the prothrombinase
(such as unfractionated heparin and low-molecular-weight hepa- complex, respectively (80). Moreover, thrombin mediates a
rin – which have anti-factor Xa activity – and fondaparinux, which number of processes that promote anticoagulation and fibrinoly-
is an indirect factor Xa inhibitor) are unable to do. Direct factor Xa sis, via the thrombin−TM complex and APC system.
inhibitors inhibit free, prothrombinase-bound, and clot- Fibrin-bound thrombin is an important potentiator of throm-
associated factor Xa (61, 64, 65). Clot-associated factor Xa has been bus growth. Unlike indirect thrombin inhibitors, direct thrombin
shown to be enzymatically active in vitro and is able to activate pro- inhibitors inactivate fibrin-bound thrombin and free thrombin
thrombin to thrombin (66) thus contributing to clot-associated (81). In addition, because direct thrombin inhibitors do not bind
procoagulant activity (67).Therefore, direct inhibition of clot- to platelet factor 4, their anticoagulant activity is not affected by the
associated factor Xa could be an effective and localised approach to large quantities of this chemokine released from activated platelets
prevent thrombus growth. Recent evidence suggests that rivar- in platelet-rich thrombi (7).
oxaban, a direct factor Xa inhibitor, also affects clot structure by in- Thrombin is also involved in many other physiological and pa-
creasing the permeability and degradability of the clot, thus pro- thophysiological processes such as inflammation and wound heal-
moting clot lysis (68). ing (20). Vascular endothelial growth factor (VEGF) is a specific
In addition to coagulation, factor Xa also exhibits proinflam- endothelial mitogen that initiates angiogenesis. It mediates en-
matory and proliferative activities. Factor Xa induces macrophage dothelial cell proliferation, cellular migration, and vascular tube
migration inhibitory factor in endothelial cells and the expression formation. Thrombin potentiates the effects of VEGF; clot-bound
of IL-6, IL-8, and monocyte chemotactic protein, thus contribu- thrombin, which is resistant to inhibition by AT, contributes to an-
ting to the inflammatory processes (69–71). Recent studies have giogenesis and tissue repair at the sites of injury. The effect of
also indicated a potential role for factor Xa in the progression of thrombin on vascular permeability contributes to oedema and
tissue fibrosis and wound healing (72). The binding of factor Xa to swelling associated with inflammation (20). Furthermore, throm-
vascular endothelial cells induces the release of platelet-derived bin mediates inflammatory processes via increasing monocyte ad-
growth factor, which is involved in cell proliferation (73, 74). The hesion to selectins (19) and the stimulation of proinflammatory
migration and proliferation of vascular smooth muscle cells in re- cytokine (e.g. IL-6 and IL-8) and chemokine production (20, 82,
sponse to endothelial injury contribute to the development of res- 83). These properties also present thrombin as a target for other
tenosis and atherosclerosis. Preclinical evidence suggests that fac- pharmacological interventions beyond coagulation. However,
tor Xa inhibition could block some of the proinflammatory and thrombin also has anti-inflammatory properties via the thrombin−
proliferative processes, such as the expression of proinflammatory TM complex and the APC system, and the APC system exhibits
cytokines (75), and the proliferation of vascular smooth muscle other cytoprotective functions (such as anti-apoptosis and
cells (76) and, thus, limit restenosis after balloon angioplasty (77, endothelial barrier protection) (21). While inhibiting thrombin
78). activity may have anti-inflammatory potential by blocking throm-
It has been shown recently that the oral, direct factor Xa in- bin-mediated proinflammatory activities, it is not known whether
hibitor rivaroxaban concentration-dependently inhibited the pro- long-term inhibition of the APC system (e.g. by thrombin
coagulant activity of activated monocytes and macrophages, inhibition) would have any adverse consequences.
whereas the indirect factor Xa inhibitor fondaparinux did not dis-
play this effect (79). This property could be attributed to the abil-
ity of rivaroxaban to inhibit factor Xa bound to monocytes. Inter-
estingly, both rivaroxaban and fondaparinux exhibited anti- Inhibition of platelets
inflammatory activity by inhibiting the secretion of inflammatory
chemokines – an effect that was thought to be due to the inhibition Platelets play a vital role in haemostasis and are key participants in
of thrombin generation (79); it is not yet known to what extent this the pathogenesis of arterial thrombosis (84). When vascular injury
is attributed to the direct inhibition of factor Xa-mediated occurs (such as the rupture of an atherosclerotic plaque), platelets
proinflammatory activities. are rapidly recruited to the site, followed by activation and aggre-
gation. Activated platelets then release the contents of granules,
which further promote platelet recruitment, adhesion, aggre-
gation, and activation. This results in the rapid growth of the

© Schattauer 2011 Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
594 Turpie, Esmon: Anticoagulation in thrombosis

thrombus (44). Antiplatelet agents are used for the prevention and blocking thrombin generation or activity, the new oral anticoagu-
acute treatment of arterial thrombosis. The primary targets are lants might have potential anti-inflammatory effects, which could
molecules involved in platelet activation and aggregation. The contribute to their antithrombotic efficacy and the overall clinical
commonly used agents include acetylsalicylic acid (a COX-1 in- benefits. However, it is not yet known whether long-term suppres-
hibitor) and the thienopyridines (ADP receptor antagonists) (84). sion of the APC pathway (e.g. through inhibiting thrombin activ-
Although the established efficacy has validated platelet COX-1 and ity) could cause adverse consequences. Further studies examining
ADP receptors as clinically viable targets for antiplatelet therapy, the influence of direct factor Xa inhibitors and direct thrombin in-
there are limitations with the use of these agents, such as the in- hibitors on the APC system, as well as inflammatory markers, may
complete inhibition of platelet aggregation and/or ongoing provide further insight.
thromboxane A2 production with the use of acetylsalicylic acid and
the delayed onset and offset of action with the thienopyridines (7). Acknowledgements
Recent attention has focused on the protease activated receptor-1 The authors would like to acknowledge Yong-Ling Liu who pro-
inhibitors and novel ADP receptor antagonists (7). vided editorial support with funding from Bayer Schering Pharma
AG and Johnson & Johnson Pharmaceutical Research & Develop-
ment, L.L.C.
Other pharmacological agents

In addition to anticoagulant and antiplatelet therapies, there are


also other agents that exhibit antithrombotic activity. For example, References
statins have been shown to significantly reduce the risk of VTE in 1. Albers GW, Amarenco P, Easton JD, et al. Antithrombotic and thrombolytic ther-
recent clinical studies (85–87). The potential mechanisms that me- apy for ischemic stroke: American College of Chest Physicians evidence-based
clinical practice guidelines (8th Edition). Chest 2008; 133: 630S–669S.
diate the antithrombotic effect of statins include the reduction of 2. Harrington RA, Becker RC, Cannon CP, et al. Antithrombotic therapy for non-
TF expression and thrombin generation, and thus the attenuation ST-segment elevation acute coronary syndromes: American College of Chest
of fibrinogen cleavage (88). In addition, statins have been shown to Physicians evidence-based clinical practice guidelines (8th Edition). Chest 2008;
promote TM expression on endothelial cells, thereby enhancing 133: 670S–707S.
3. Kearon C. Natural history of venous thromboembolism. Circulation 2003; 107:
the APC anticoagulant pathway (89, 90). Thus, the benefits of sta- I22–I30.
tins may be attributed not only to their effects on lipid levels and 4. Geerts WH, Bergqvist D, Pineo GF, et al. Prevention of venous thromboembolism:
anti-inflammatory properties, but also their potential influence on American College of Chest Physicians evidence-based clinical practice guidelines
(8th Edition). Chest 2008; 133: 381S–453S.
coagulation (88, 91).
5. Ansell J, Hirsh J, Hylek E, et al. Pharmacology and management of the vitamin K
antagonists: American College of Chest Physicians evidence-based clinical prac-
tice guidelines (8th Edition). Chest 2008; 133: 160S–198S.
6. Hirsh J, Guyatt G, Albers GW, et al. Antithrombotic and thrombolytic therapy:
American College of Chest Physicians evidence-based clinical practice guidelines
Conclusions (8th Edition). Chest 2008; 133: 110S–112S.
7. Weitz JI, Hirsh J, Samama MM. New antithrombotic drugs: American College of
The aetiology and pathological mechanisms are different for ve- Chest Physicians evidence-based clinical practice guidelines (8th Edition). Chest
nous and arterial thrombosis, which are reflected in their different 2008; 133: 234S–256S.
8. Furie B, Furie BC. Mechanisms of thrombus formation. N Engl J Med 2008; 359:
treatment strategies. Both oral, direct factor Xa inhibitors and oral, 938–949.
direct thrombin inhibitors have demonstrated potential in clinical 9. Seré KM, Hackeng TM. Basic mechanisms of hemostasis. Semin Vasc Med 2003;
studies to date (92–98), providing further evidence for both 3: 3–12.
factor Xa and thrombin being viable targets of anticoagulant ther- 10. Chou J, Mackman N, Merrill-Skoloff G, et al. Hematopoietic cell-derived micro-
particle tissue factor contributes to fibrin formation during thrombus propa-
apy. However, each drug or drug class may behave differently when gation. Blood 2004; 104: 3190–3197.
used in different clinical indications, likely due to the different 11. Mann KG. The biochemistry of coagulation. Clin Lab Med 1984; 4: 207–220.
mechanisms of action of the drugs and differences in the patho- 12. Monroe DM, Hoffman M. What does it take to make the perfect clot? Arterioscler
genesis of disease conditions. Thromb Vasc Biol 2006; 26: 41–48.
13. Weitz JI, Bates SM. New anticoagulants. J Thromb Haemost 2005; 3: 1843–1853.
Accumulating evidence suggests a strong link between coagu- 14. Bates SM, Weitz JI. Coagulation assays. Circulation 2005; 112: e53–e60.
lation and inflammation. Inflammatory mediators trigger coagu- 15. Kannemeier C, Shibamiya A, Nakazawa F, et al. Extracellular RNA constitutes a
lation responses, and coagulation factors mediate inflammatory natural procoagulant cofactor in blood coagulation. Proc Natl Acad Sci USA 2007;
104: 6388–6393.
processes and cell proliferation. The APC system, which has
16. Xu J, Zhang X, Pelayo R, et al. Extracellular histones are major mediators of death
multiple cytoprotective roles, is downregulated in inflammation. in sepsis. Nat Med 2009; 15: 1318–1321.
The biological and pathophysiological roles of factor Xa and 17. Lopez JA, Kearon C, Lee AY. Deep venous thrombosis. Hematology Am Soc He-
thrombin are far beyond coagulation. The anti-inflammatory and matol Educ Program 2004; 439–456.
18. Tanaka KA, Key NS, Levy JH. Blood coagulation: hemostasis and thrombin regu-
metastatic inhibitory effects of heparin, for example, are believed lation. Anesth Analg 2009; 108: 1433–1446.
to be independent of their anticoagulation property (99). Al- 19. Esmon CT. Crosstalk between inflammation and thrombosis. Maturitas 2008; 61:
though there are no clinical data currently available, by directly 122–131.

Thrombosis and Haemostasis 105.4/2011 © Schattauer 2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
Turpie, Esmon: Anticoagulation in thrombosis 595

20. Licari LG, Kovacic JP. Thrombin physiology and pathophysiology. J Vet Emerg 47. Crisby M, Nordin-Fredriksson G, Shah PK, et al. Pravastatin treatment increases
Crit Care (San Antonio) 2009; 19: 11–22. collagen content and decreases lipid content, inflammation, metalloproteinases,
21. Sarangi PP, Lee HW, Kim M. Activated protein C action in inflammation. Br J and cell death in human carotid plaques: implications for plaque stabilization.
Haematol 2010; 148: 817–833. Circulation 2001; 103: 926–933.
22. Esmon CT. Coagulation inhibitors in inflammation. Biochem Soc Trans 2005; 33: 48. Lefer DJ. Statins as potent antiinflammatory drugs. Circulation 2002; 106:
401–405. 2041–2042.
23. Conway EM, Rosenberg RD. Tumor necrosis factor suppresses transcription of 49. Stewart RA. Predicting benefit from statins by C-reactive protein, LDL-choleste-
the thrombomodulin gene in endothelial cells. Mol Cell Biol 1988; 8: 5588–5592. rol or absolute cardiovascular risk. Future Cardiol 2009; 5: 231–236.
24. Esmon CT. Inflammation and the activated protein C anticoagulant pathway. 50. Kinlay S. Low-density lipoprotein-dependent and -independent effects of choles-
Semin Thromb Hemost 2006; 32 (Suppl. 1): 49–60. terol-lowering therapies on C-reactive protein: a meta-analysis. J Am Coll Cardiol
25. Fisher CJ, Jr., Yan SB. Protein C levels as a prognostic indicator of outcome in sep- 2007; 49: 2003–2009.
sis and related diseases. Crit Care Med 2000; 28: S49–S56. 51. Wang K, Tarakji K, Zhou Z, et al. Celecoxib, a selective cyclooxygenase-2 inhibitor,
26. Isermann B, Vinnikov IA, Madhusudhan T, et al. Activated protein C protects decreases monocyte chemoattractant protein-1 expression and neointimal hy-
against diabetic nephropathy by inhibiting endothelial and podocyte apoptosis. perplasia in the rabbit atherosclerotic balloon injury model. J Cardiovasc Phar-
Nat Med 2007; 13: 1349–1358. macol 2005; 45: 61–67.
27. Cheng T, Liu D, Griffin JH, et al. Activated protein C blocks p53-mediated apop- 52. Chen SS, Jenkins AJ, Majewski H. Elevated plasma prostaglandins and acetylated
tosis in ischemic human brain endothelium and is neuroprotective. Nat Med histone in monocytes in Type 1 diabetes patients. Diabet Med 2009; 26: 182–186.
2003; 9: 338–342. 53. Goodman SG, Menon V, Cannon CP, et al. Acute ST-segment elevation myo-
28. Van Sluis GL, Niers TM, Esmon CT, et al. Endogenous activated protein C limits cardial infarction: American College of Chest Physicians evidence-based clinical
cancer cell extravasation through sphingosine-1-phosphate receptor 1-mediated practice guidelines (8th Edition). Chest 2008; 133: 708S–775S.
vascular endothelial barrier enhancement. Blood 2009; 114: 1968–1973. 54. Lip GYH, Boos CJ. Antithrombotic treatment in atrial fibrillation. Heart 2006; 92:
29. Han MH, Hwang SI, Roy DB, et al. Proteomic analysis of active multiple sclerosis 155–161.
lesions reveals therapeutic targets. Nature 2008; 451: 1076–1081. 55. Fuster V, Rydén LE, Asinger RW, et al. ACC/AHA/ESC guidelines for the manage-
30. Loubele ST, Spek CA, Leenders P, et al. Activated protein C protects against myo- ment of patients with atrial fibrillation. A report of the American College of Car-
cardial ischemia/reperfusion injury via inhibition of apoptosis and inflam- diology/American Heart Association Task Force on Practice Guidelines and the
mation. Arterioscler Thromb Vasc Biol 2009; 29: 1087–1092. European Society of Cardiology Committee for Practice Guidelines and Policy
31. Mizutani A, Okajima K, Uchiba M, et al. Activated protein C reduces ischemia/ Conferences (Committee to develop guidelines for the management of patients
reperfusion-induced renal injury in rats by inhibiting leukocyte activation. Blood with atrial fibrillation) developed in collaboration with the North American So-
2000; 95: 3781–3787. ciety of Pacing and Electrophysiology. Eur Heart J 2001; 22: 1852–1923.
32. Kroegel C, Reissig A. Principle mechanisms underlying venous thromboembol- 56. Watson T, Shantsila E, Lip GY. Mechanisms of thrombogenesis in atrial fibril-
ism: epidemiology, risk factors, pathophysiology and pathogenesis. Respiration lation: Virchow's triad revisited. Lancet 2009; 373: 155–166.
2003; 70: 7–30. 57. Olsson SB, Rasmussen LH, Tveit A, et al. Safety and tolerability of an immediate-
33. Lowe GD. Virchow's triad revisited: abnormal flow. Pathophysiol Haemost release formulation of the oral direct thrombin inhibitor AZD0837 in the preven-
Thromb 2003; 33: 455–457. tion of stroke and systemic embolism in patients with atrial fibrillation. Thromb
34. Sevitt S. The structure and growth of valve-pocket thrombi in femoral veins. J Clin Haemost 2010; 103: 604–612.
Pathol 1974; 27: 517–528. 58. Perzborn E. Factor Xa inhibitors – New anticoagulants for secondary haemosta-
35. Chirinos JA, Heresi GA, Velasquez H, et al. Elevation of endothelial micropar- sis. Hamostaseologie 2009; 29: 260–267.
ticles, platelets, and leukocyte activation in patients with venous thromboembol- 59. Mann KG, Brummel K, Butenas S. What is all that thrombin for? J Thromb Hae-
ism. J Am Coll Cardiol 2005; 45: 1467–1471. most 2003; 1: 1504–1514.
36. Esmon CT. Basic mechanisms and pathogenesis of venous thrombosis. Blood Rev 60. Ansell J. Factor Xa or thrombin: is factor Xa a better target? J Thromb Haemost
2009; 23: 225–229. 2007; 5 (Suppl. 1): 60–64.
37. McNally MA, Mollan RA. Total hip replacement, lower limb blood flow and ve- 61. Perzborn E, Strassburger J, Wilmen A, et al. In vitro and in vivo studies of the novel
nous thrombogenesis. J Bone Joint Surg Br 1993; 75: 640–644. antithrombotic agent BAY 59-7939 − an oral, direct Factor Xa inhibitor. J Thromb
38. Dahl OE, Aspelin T, Arnesen H, et al. Increased activation of coagulation and Haemost 2005; 3: 514–521.
formation of late deep venous thrombosis following discontinuation of thrombo- 62. Leadley RJ, Jr. Coagulation factor Xa inhibition: biological background and
prophylaxis after hip replacement surgery. Thromb Res 1995; 80: 299–306. rationale. Curr Top Med Chem 2001; 1: 151–159.
39. Lindahl TL, Lundahl TH, Nilsson L, et al. APC-resistance is a risk factor for post- 63. Orvim U, Barstad RM, Vlasuk GP, et al. Effect of selective factor Xa inhibition on
operative thromboembolism in elective replacement of the hip or knee – a pros- arterial thrombus formation triggered by tissue factor/factor VIIa or collagen in
pective study. Thromb Haemost 1999; 81: 18–21. an ex vivo model of shear-dependent human thrombogenesis. Arterioscler
40. Meltzer ME, Doggen CJ, de Groot PG, et al. The impact of the fibrinolytic system Thromb Vasc Biol 1995; 15: 2188–2194.
on the risk of venous and arterial thrombosis. Semin Thromb Hemost 2009; 35: 64. Depasse F, Busson J, Mnich J, et al. Effect of BAY 59-7939 − a novel, oral, direct
468–477. Factor Xa inhibitor − on clot-bound Factor Xa activity in vitro. J Thromb Hae-
41. Singer DE, Albers GW, Dalen JE, et al. Antithrombotic therapy in atrial fibril- most 2005; 3 (Suppl. 1): Abstract P1104.
lation: American College of Chest Physicians evidence-based clinical practice 65. Jiang X, Crain EJ, Luettgen JM, et al. Apixaban, an oral direct factor Xa inhibitor,
guidelines (8th Edition). Chest 2008; 133: 546S–592S. inhibits human clot-bound factor Xa activity in vitro. Thromb Haemost 2009;
42. Bassand JP, Hamm CW, Ardissino D, et al. Guidelines for the diagnosis and treat- 101: 780–782.
ment of non-ST-segment elevation acute coronary syndromes. Eur Heart J 2007; 66. Eisenberg PR, Siegel JE, Abendschein DR, et al. Importance of factor Xa in deter-
28: 1598–1660. mining the procoagulant activity of whole-blood clots. J Clin Invest 1993; 91:
43. Davies MJ. The pathophysiology of acute coronary syndromes. Heart 2000; 83: 1877–1883.
361–366. 67. Prager NA, Abendschein DR, McKenzie CR, et al. Role of thrombin compared
44. Mackman N. Triggers, targets and treatments for thrombosis. Nature 2008; 451: with factor Xa in the procoagulant activity of whole blood clots. Circulation 1995;
914–918. 92: 962–967.
45. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl 68. Varin R, Mirshahi S, Mirshahi P, et al. Effect of rivaroxaban, an oral direct Factor
J Med 2005; 352: 1685–1695. Xa inhibitor, on whole blood clot permeation and thrombolysis: critical role of
46. Ballantyne CM, Hoogeveen RC, Bang H, et al. Lipoprotein-associated phospholi- red blood cells. Blood (ASH Annual Meeting Abstracts) 2009; 114: Abstract 1064.
pase A2, high-sensitivity C-reactive protein, and risk for incident coronary heart 69. Bachli EB, Pech CM, Johnson KM, et al. Factor Xa and thrombin, but not factor
disease in middle-aged men and women in the Atherosclerosis Risk in Commu- VIIa, elicit specific cellular responses in dermal fibroblasts. J Thromb Haemost
nities (ARIC) study. Circulation 2004; 109: 837–842. 2003; 1: 1935–1944.

© Schattauer 2011 Thrombosis and Haemostasis 105.4/2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
596 Turpie, Esmon: Anticoagulation in thrombosis

70. Shimizu T, Nishihira J, Watanabe H, et al. Macrophage migration inhibitory fac- 85. Glynn RJ, Danielson E, Fonseca FA, et al. A randomized trial of rosuvastatin in the
tor is induced by thrombin and factor Xa in endothelial cells. J Biol Chem 2004; prevention of venous thromboembolism. N Engl J Med 2009; 360: 1851–1861.
279: 13729–13737. 86. Ramcharan AS, Van Stralen KJ, Snoep JD, et al. HMG-CoA reductase inhibitors,
71. Busch G, Seitz I, Steppich B, et al. Coagulation factor Xa stimulates interleukin-8 other lipid-lowering medication, antiplatelet therapy, and the risk of venous
release in endothelial cells and mononuclear leukocytes: implications in acute thrombosis. J Thromb Haemost 2009; 7: 514–520.
myocardial infarction. Arterioscler Thromb Vasc Biol 2005; 25: 461–466. 87. Sørensen HT, Horvath-Puho E, Søgaard KK, et al. Arterial cardiovascular events,
72. Borensztajn K, Stiekema J, Nijmeijer S, et al. Factor Xa stimulates proinflamma- statins, low-dose aspirin and subsequent risk of venous thromboembolism: a
tory and profibrotic responses in fibroblasts via protease-activated receptor-2 ac- population-based case-control study. J Thromb Haemost 2009; 7: 521–528.
tivation. Am J Pathol 2008; 172: 309–320. 88. Undas A, Celinska-Lowenhoff M, Brummel-Ziedins KE, et al. Simvastatin given
73. Blanc-Brude OP, Chambers RC, Leoni P, et al. Factor Xa is a fibroblast mitogen via for 3 days can inhibit thrombin generation and activation of factor V and enhance
binding to effector-cell protease receptor-1 and autocrine release of PDGF. Am J factor Va inactivation in hypercholesterolemic patients. Arterioscler Thromb Vasc
Physiol Cell Physiol 2001; 281: C681–C689. Biol 2005; 25: 1524–1525.
74. Ko FN, Yang YC, Huang SC, et al. Coagulation factor Xa stimulates platelet-de- 89. Sen-Banerjee S, Mir S, Lin Z, et al. Kruppel-like factor 2 as a novel mediator of sta-
rived growth factor release and mitogenesis in cultured vascular smooth muscle tin effects in endothelial cells. Circulation 2005; 112: 720–726.
cells of rat. J Clin Invest 1996; 98: 1493–1501. 90. Masamura K, Oida K, Kanehara H, et al. Pitavastatin-induced thrombomodulin
75. Joo SS, Won TJ, Kim JS, et al. Inhibition of coagulation activation and inflam- expression by endothelial cells acts via inhibition of small G proteins of the Rho
mation by a novel Factor Xa inhibitor synthesized from the earthworm Eisenia family. Arterioscler Thromb Vasc Biol 2003; 23: 512–517.
andrei. Biol Pharm Bull 2009; 32: 253–258. 91. Albert MA, Danielson E, Rifai N, et al. Effect of statin therapy on C-reactive pro-
76. Walenga JM, Jeske WP, Hoppensteadt D, et al. Factor Xa inhibitors: today and tein levels: the pravastatin inflammation/CRP evaluation (PRINCE): a random-
beyond. Curr Opin Investig Drugs 2003; 4: 272–281. ized trial and cohort study. J Am Med Assoc 2001; 286: 64–70.
77. Ragosta M, Gimple LW, Gertz SD, et al. Specific factor Xa inhibition reduces res- 92. Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients
tenosis after balloon angioplasty of atherosclerotic femoral arteries in rabbits. with atrial fibrillation. N Engl J Med 2009; 361: 1139–1151.
Circulation 1994; 89: 1262–1271. 93. Eriksson BI, Borris LC, Friedman RJ, et al. Rivaroxaban versus enoxaparin for
78. Schwartz RS, Holder DJ, Holmes DR, et al. Neointimal thickening after severe cor- thromboprophylaxis after hip arthroplasty. N Engl J Med 2008; 358: 2765–2775.
onary artery injury is limited by a short-term administration of a factor Xa in- 94. Kakkar AK, Brenner B, Dahl OE, et al. Extended duration rivaroxaban versus
hibitor. Results in a porcine model. Circulation 1996; 93: 1542–1548. short-term enoxaparin for the prevention of venous thromboembolism after
79. Laurent M, Varin R, Joimel U, et al. A novel mechanism of action of rivaroxaban: total hip arthroplasty: a double-blind, randomised controlled trial. Lancet 2008;
inhibition of monocyte and macrophage procoagulant activity and consequence 372: 31–39.
on inflammatory process. Blood (ASH Annual Meeting Abstracts) 2009; 114: 95. Lassen MR, Ageno W, Borris LC, et al. Rivaroxaban versus enoxaparin for throm-
Abstract 3124. boprophylaxis after total knee arthroplasty. N Engl J Med 2008; 358: 2776–2786.
80. Mann KG, Butenas S, Brummel K. The dynamics of thrombin formation. Arte- 96. Lassen MR, Gallus AS, Pineo GF, et al. The ADVANCE-2 study: a randomized
rioscler Thromb Vasc Biol 2003; 23: 17–25. double-blind trial comparing apixaban with enoxaparin for thromboprophylaxis
81. Stangier J, Rathgen K, Stahle H, et al. The pharmacokinetics, pharmacodynamics after total knee replacement. J Thromb Haemost 2009; 7 (Suppl 2): Abstract
and tolerability of dabigatran etexilate, a new oral direct thrombin inhibitor, in LB-MO-005.
healthy male subjects. Br J Clin Pharmacol 2007; 64: 292–303. 97. Turpie AGG, Lassen MR, Davidson BL, et al. Rivaroxaban versus enoxaparin for
82. Bar-Shavit R, Kahn A, Fenton JW, et al. Chemotactic response of monocytes to thromboprophylaxis after total knee arthroplasty (RECORD4): a randomised
thrombin. J Cell Biol 1983; 96: 282–285. trial. Lancet 2009; 373: 1673–1680.
83. Scholz M, Vogel JU, Hover G, et al. Thrombin stimulates IL-6 and IL-8 expression 98. Olsson SB, Executive Steering Committee of the SPORTIF III Investigators.
in cytomegalovirus-infected human retinal pigment epithelial cells. Int J Mol Stroke prevention with the oral direct thrombin inhibitor ximelagatran com-
Med 2004; 13: 327–331. pared with warfarin in patients with non-valvular atrial fibrillation (SPORTIF
84. Patrono C, Baigent C, Hirsh J, et al. Antiplatelet drugs: American College of Chest III): randomised controlled trial. Lancet 2003; 362: 1691–1698.
Physicians evidence-based clinical practice guidelines (8th Edition). Chest 2008; 99. Ludwig RJ. Therapeutic use of heparin beyond anticoagulation. Curr Drug Dis-
133: 199S–233S. cov Technol 2009; 6: 281–289.

Thrombosis and Haemostasis 105.4/2011 © Schattauer 2011

Downloaded from www.thrombosis-online.com on 2015-12-28 | IP: 54.152.109.166


For personal or educational use only. No other uses without permission. All rights reserved.
View publication stats

You might also like