You are on page 1of 20

REVIEWS

Therapeutic strategies for thrombosis:


new targets and approaches
Nigel Mackman 1 ✉, Wolfgang Bergmeier2, George A. Stouffer 3
and Jeffrey I. Weitz 4

Abstract | Antiplatelet agents and anticoagulants are a mainstay for the prevention and treatment
of thrombosis. However, despite advances in antithrombotic therapy, a fundamental challenge
is the side effect of bleeding. Improved understanding of the mechanisms of haemostasis and
thrombosis has revealed new targets for attenuating thrombosis with the potential for less bleeding,
including glycoprotein VI on platelets and factor XIa of the coagulation system. The efficacy
and safety of new agents are currently being evaluated in phase III trials. This Review provides an
overview of haemostasis and thrombosis, details the current landscape of antithrombotic agents,
addresses challenges with preventing thromboembolic events in patients at high risk and describes
the emerging therapeutic strategies that may break the inexorable link between antithrombotic
therapy and bleeding risk.

Cardiovascular disease Thrombosis is the leading cause of death worldwide, and endothelial dysfunction10. Specific risk factors for
Disorders of the heart and being responsible for one in four deaths1. The global VTE include genetic factors (such as factor V (FV)
blood vessels. burden is likely to increase with the ageing population Leiden, a prothrombotic variant of FV) or deficiencies
because thrombosis risk increases with age. Thrombosis of endogenous anticoagulants (such as antithrombin or
Platelets
Small enucleate cells in the
can be divided into arterial thrombosis (AT) and venous protein C) as well as non-​genetic factors (for example,
blood involved in clotting. thrombosis (VT). Interestingly, patients with AT are at immobility, surgery and age)8,10. In contrast to arterial
increased risk of VT and vice versa2–6. In addition, var- thrombi, venous thrombi form slowly on an intact, but
ious conditions, such as antiphospholipid syndrome, likely activated, endothelium and are fibrin rich but also
malignancy, infection, and oestrogen use, are associated contain platelets and trapped red blood cells7.
with both AT and VT2. Atrial fibrillation (AF), the most common sustained
Patients with cardiovascular disease (CVD) are at cardiac arrhythmia, is responsible for about 25% of
1
UNC Blood Research Center,
increased risk of AT7. The main acquired risk factors all ischaemic strokes. Stasis of blood in the left atrial
Division of Hematology and for AT are hyperlipidaemia, smoking, diabetes, hyper- appendage or left atrium of patients with AF and other
Oncology, Department of tension, chronic kidney disease and obesity8. Rupture factors can trigger thrombus formation11. These thrombi
Medicine, University of North of atherosclerotic plaques leads to the formation of a can break off and travel from the left atrium to lodge
Carolina at Chapel Hill,
thrombus — a process commonly referred to as ath- in the brain, coronary arteries, abdominal arteries or
Chapel Hill, NC, USA.
erothrombosis9 (Fig. 1). By contrast, eroded plaques are peripheral arteries. While the left atrial appendage is
2
UNC Blood Research Center,
Department of Biochemistry
not normally associated with the formation of occlud- the most common site for thrombus formation, patients
and Biophysics, University of ing thrombi because they do not present a highly pro- with AF have haematological findings consistent with
North Carolina at Chapel Hill, thrombotic surface. Arterial thrombi are platelet rich, a prothrombotic state12. Strokes in patients with AF are
Chapel Hill, NC, USA. but are stabilized by crosslinked fibrin (Fig. 1). Clinical more likely to be fatal or disabling than those in patients
3
McAllister Heart Institute, manifestations of atherothrombosis primarily occur in without AF because the thrombi are larger.
Division of Cardiology, the heart (myocardial infarction (MI)), brain (ischae- Antithrombotic agents fall into three classes — anti-
Department of Medicine,
University of North Carolina
mic stroke) and legs (peripheral artery disease (PAD), platelet agents, anticoagulants and fibrinolytic agents.
at Chapel Hill, Chapel Hill, with its attendant risk of limb ischaemia, gangrene Because of the preponderance of platelets in arterial
NC, USA. and amputation). thrombi, antiplatelet agents are the mainstay for the pre-
4
Thrombosis & Venous thrombi occur in deep veins most often in the vention and treatment of AT. There are four main types
Atherosclerosis Research legs or arms, a process known as deep vein thrombosis10. of FDA-​approved antiplatelet agents — a cyclooxygen-
Institute and Department of These clots can break off, travel to the lungs and lodge ase inhibitor (aspirin), P2Y12 antagonists (for example,
Medicine, McMaster
University, Hamilton, Canada.
in pulmonary arteries — a process known as pulmonary clopidogrel, prasugrel and ticagrelor), αIIbβ3 antago-
✉e-​mail: nmackman@ embolism (PE). Deep vein thrombosis and PE are col- nists (for example, abciximab) and a protease-​activated
med.unc.edu lectively known as venous thromboembolism (VTE). receptor 1 (PAR1) antagonist (vorapaxar) (Box 1, Fig. 2).
https://doi.org/10.1038/ VTE is triggered by one or more factors of Virchow’s Antiplatelet therapy is also effective as secondary
s41573-020-0061-0 triad — sluggish blood flow, blood hypercoagulability prevention in patients with ischaemic stroke (Box 2).

Nature Reviews | Drug Discovery


Reviews

Blood vessel Endothelial


cells

Lumen

RBC Neutrophil
Platelet/fibrin thrombus

NETs
FVa–FXa Thrombin

TxA2, ADP
Fibrin

FVIIIa–FIXa Platelet

Plaque rupture
FXI

TF–FVIIa FXIIa Collagen/vWF

Nucleic acid

Fig. 1 | Formation of an occlusive arterial thrombosis. Rupture of an atherosclerotic plaque exposes collagen
and von Willebrand factor (vWF), which bind and activate platelets. Released second messengers, such as ADP and
thromboxane A2 (TxA2), further activate platelets. Tissue factor (TF) within the plaque binds factor VIIa (FVIIa), and this
complex activates the clotting cascade, resulting in fibrin formation. In addition, FXII is activated by nucleic acid and
other negatively charged surfaces and contributes to thrombin generation. Neutrophils are incorporated into the
thrombus and extrude neutrophil extracellular traps (NETs). Red blood cells (RBCs) are also incorporated into thrombi.

Anticoagulants are the cornerstone for the prevention there is widespread underuse of anticoagulants by clin­
and treatment of VT because they attenuate the forma- icians and patients because of concerns with bleeding,
tion of fibrin, which predominates in venous thrombi. particularly in the elderly. One study analysed the use of
Anticoagulants are also the main therapeutic modality oral anticoagulation in eligible patients with AF between
used to prevent stroke in patients with AF. There are four 2008 and 2014 and found that there was a significant
classes of FDA-​approved anticoagulants — heparins, increase in use rates from 52.4% to 60.7%16, correspond-
including low-​molecular-​weight heparin (LMWH), ing with an increase in the use of DOACs (0 to 25.8%)
vitamin K antagonists (VKAs; such as warfarin), direct during this time period. However, despite this increase,
thrombin inhibitors (DTIs; such as bivalirudin and dab- up to 40% of patients with AF at risk for stroke fail to
igatran) and direct FXa inhibitors (such as apixaban, receive anticoagulation therapy, which may be the result
edoxaban and rivaroxaban) (Box 3, Fig. 2). Oral FXa and of bleeding concerns.
thrombin inhibitors are called direct oral anticoagulants There are a large number of agents targeting plate-
(DOACs). Finally, fibrinolytic agents are used to degrade lets and components of the coagulation cascade that
arterial and venous thrombi13. are claimed to reduce thrombosis without affecting
All current antithrombotic agents are associated haemostasis; however, most have only been studied in
Haemostasis
with an increased risk of bleeding because platelets and preclinical models17–20. This review provides an over-
Arrest of bleeding after crosslinked fibrin are essential components of haemo- view of the current landscape of emerging antiplatelet
damage to a blood vessel. static plugs that seal leaks in the vasculature14,15. Indeed, and anticoagulant targets and agents in development,

www.nature.com/nrd
Reviews

particularly those that are currently being studied the blood, where they circulate for 7–10 days. All com-
in clinical trials. Ongoing challenges in the field and ponents of the coagulation cascade, except tissue factor
potential strategies to address them are discussed. (TF), are present in the blood of healthy individuals21,28.
Under normal conditions, endogenous platelet inhib-
Overview of haemostasis and thrombosis itors and anticoagulants prevent activation of the clot-
Haemostasis ting system29–32. For instance, the vascular endothelium
The mammalian haemostatic system has evolved to inhibits platelet activation via multiple mechanisms
respond rapidly to vascular injury and minimize blood that include release of prostaglandin I2 (also known as
loss by forming a haemostatic plug consisting of plate- prostacyclin) and nitric oxide as well as expression of
lets and crosslinked fibrin21–27. Platelets are the major ectonucleotidases (which degrade the platelet activators
cellular component of the haemostatic system, inter- ATP and ADP)32,33. Activation of the coagulation cascade
acting closely with components of the coagulation is negatively regulated by various anticoagulant proteins
cascade, namely coagulation proteins and fibrinogen/ that act at different points in the cascade29,30. For instance,
fibrin. Platelets are small enucleate cells produced by tissue factor pathway inhibitor (TFPI) — present on
mega­karyocytes in the bone marrow and released into the endothelium and in the blood — inhibits FXa, FVa

Box 1 | A brief history of antiplatelet agents


there are currently four major types of antiplatelet agents used for the prevention and treatment of arterial thrombosis,
namely aspirin (acetylsalicylic acid), P2Y12 antagonists, αiibβ3 (also known as GPiib/iiia) antagonists and a Par1 inhibitor
(Fig. 2).
in 1956, it was proposed that the anti-​inflammatory agent aspirin could protect individuals from heart attacks, and,
several years later, two groups identified the mechanism by which aspirin inhibits platelet activation and aggregation10.
aspirin irreversibly acetylates cyclooxygenase 1, which is used by platelets to generate the second messenger thromboxane
a2 — a potent platelet agonist. the first clinical trials evaluating the use of aspirin for secondary prevention in patients
with myocardial infarction were reported in 1974 (ref.223). since then, aspirin has been successfully used both as a single
agent and in combination with other antiplatelet or anticoagulant agents.
the second class of antiplatelet agents is the P2Y12 antagonists. P2Y12 is a major aDP receptor on the platelet
surface and ADP released from activated platelets binds to P2Y12 on adjacent platelets, thereby propagating platelet
activation and aggregation.
in 1972, studies into the anti-​inflammatory effects of a thienopyridine revealed that thienopyridine derivatives exhibit
antiplatelet effects. the first-​generation thienopyridine, ticlopidine, was used globally in 1991 for the primary and secondary
prevention of stroke and to prevent stent thrombosis224. the second-​generation thienopyridine, clopidogrel, entered
clinical trials in 1987 and was approved by the FDa in 1997. all thienopyridines are oral prodrugs that are metabolized
in the liver by cytochrome P450 (CYP) enzymes, mainly 2C9 and 2C19. about 30% of patients do not metabolize clopidogrel
sufficiently to achieve a therapeutic antiplatelet effect, mainly owing to CYP2C19 polymorphisms225. the problem of
clopidogrel resistance led to the development of a third-​generation thienopyridine, prasugrel (FDa approved in 2009),
which relies less on CYP-​mediated metabolism and produces less variability in response than clopidogrel226.
ticagrelor and cangrelor belong to a new generation of reversible P2Y12 antagonists218,227,228. ticagrelor is an oral atP
analogue that competes with aDP for P2Y12 binding. ticagrelor was approved in europe in 2010 and in the usa in 2011.
Cangrelor is a rapid on and rapid off reversible P2Y12 inhibitor. it is administered intravenously and was approved by the
FDa in 2015. Oral aDP receptor antagonists are used to treat patients with coronary artery disease (CaD), particularly as
an adjunct to aspirin in dual antiplatelet treatment (DaPt) after percutaneous coronary intervention.
the third class of antiplatelet agents targets the integrin αiibβ3 (ref.229). Platelet activation leads to the conversion of
αiibβ3 into an active state that then mediates platelet aggregation by binding fibrinogen and other ligands230. the first
agent targeting αiibβ3 was a monoclonal antibody, abciximab, which was approved by the FDa in 1994 (ref.229). Other
inhibitors of αiibβ3 include eptifibatide (a cyclic peptidomimetic) and tirofiban (a non-​peptide derivative of tyrosine)229.
these agents are administered intravenously and are mainly used in patients at high risk following percutaneous coronary
intervention, such as those with st-​segment elevation myocardial infarction. attempts to develop oral agents in this class
failed due to prothrombotic effects231,232.
the last class of FDa-​approved agents is a protease-​activated receptor 1 (Par1) antagonist. thrombin activates platelets
by cleavage of both Par1 and Par4 (ref.233). vorapaxar is an oral, slowly reversible Par1 inhibitor derived from a natural
product, himbacine234. addition of the Par1 antagonist vorapaxar to standard of care (single antiplatelet therapy or DaPt)
for secondary prevention in patients with CaD and peripheral artery disease decreased the risk of cardiovascular death or
ischaemic events but increased intracranial haemorrhage (NCt00526474)95. a second trial in patients with acute coronary
syndrome (traCer) was terminated early because of an increase in intracranial haemorrhage in patients with a history
of stroke (NCt00527943)96. together, these two studies demonstrate that triple antiplatelet therapy is associated with a
significant increase in bleeding. Despite the increase in intracranial haemorrhage, the FDa concluded that the benefits
of vorapaxar outweighed the risks and approved vorapaxar in 2014 for secondary prevention in patients with CaD or
peripheral artery disease combined with single antiplatelet therapy or DaPt (excluding patients with a history of stroke
or transient ischaemic attack). the efficacy of vorapaxar as sole antiplatelet therapy is unknown. Despite its approval,
vorapaxar is not used, presumably because of concerns with bleeding. atopaxar is a reversible, orally administered Par1
inhibitor that was tested in several phase ii clinical trials. However, its development was halted, in part owing to liver
toxicity (NCt01000727; NCt00312052)235–237.
Despite targeting different receptors or a second messenger, the major side effect of all currently available antiplatelet
agents is bleeding.

Nature Reviews | Drug Discovery


Reviews

Platelet cascade Coagulation cascade


Platelet agonists Extrinsic/intrinsic pathways

Aspirin Plasma clotting cascade

• Apixaban
TxA2 ADP Thrombin FXa
• Clopidogrel • Betrixaban
• Prasugrel • Edoxaban
• Ticagrelor AT • Rivaroxaban
• Cangrelor P2Y12
VKA
• Vorapaxar PAR1 Heparins
• Abciximab AT
αIIbβ3 Thrombin • Bivalirudin
• Eptifibatide
• Argatroban
• Tirofiban
• Dabigatran
Platelet aggregation Fibrin

Haemostatic plug/thrombus

Fig. 2 | Targets of current antithrombotic drugs. Antiplatelet agents act either by preventing the formation of second
messengers (thromboxane A2 (TxA2)), blocking receptors (P2Y12 and PAR1) involved in platelet activation or inhibiting
platelet aggregation by binding to the integrin αIIbβ3. Anticoagulant agents act by either inhibiting post-​translational
modification of coagulation proteins, such as factor Xa (FXa) and prothrombin, inhibiting FXa and thrombin by increasing
the activity of antithrombin (AT), by directly inhibiting FXa or by directly inhibiting thrombin. VKA, vitamin K antagonist.

and the TF–FVIIa initiating complex34. The endothelial exhibit different degrees of bleeding45–48. During haemo-
protein C receptor–thrombomodulin–thrombin path- stasis, FIX is activated by the TF–FVIIa complex and, to
way generates activated protein C (APC), which cleaves a lesser extent, by the thrombin–FXIa pathway21 (Fig. 4).
and inactivates the cofactors FVa and FVIIIa28. Finally, Thrombin is the central protease in the coagulation
antithrombin inhibits most of the coagulation proteases, cascade49. It activates the cofactors FV and FVIII, con-
particularly FXa and thrombin35. verts soluble fibrinogen to fibrin monomers, which then
Both platelets and the coagulation system are acti- polymerize to form fibrin strands that tie the platelet
vated by vascular injury. Adhesion of platelets to the aggregates together, and activates FXIII, which is a trans-
injury site is mediated by transient bridging of the gly- glutaminase that crosslinks and stabilizes fibrin. Finally,
coprotein (GP) Ib–V–IX complex on the platelet sur- thrombin and other agonists activate platelets that pro-
face to subendothelial collagen via von Willebrand vide a negatively charged surface for the assembly of
factor (vWF)36 (Fig. 3). Two platelet receptors — α2β1 the intrinsic tenase (FVIIIa–FIXa) and prothrombinase
and GPVI — are critical for platelet activation and (FVa–FXa) protease/cofactor complexes that enhance
accumulation on collagen37 (Fig. 3). Adherent platelets thrombin generation by several orders of magnitude.
secrete ADP and thromboxane A2, which act as soluble The fibrinolytic system removes haemostatic plugs
agonists that recruit and activate circulating platelets27 from the vasculature after they have sealed the wound50.
(Fig. 3). Thrombin is also a potent activator of platelets The plasminogen activators tissue plasminogen activator
and mediates crosstalk between the platelet and coagu- (tPA) and urokinase plasminogen activator convert plas-
lation pathways. Activated platelets express an activated minogen to plasmin, which degrades fibrin. Fibrinolysis
form of the integrin αIIbβ3 on their surface, which binds is inhibited by plasminogen activator inhibitor 1 (PAI1),
fibrinogen and other ligands to link adjacent platelets which inhibits the plasminogen activators α2 antiplas-
together, a process known as platelet aggregation38,39 min, (a plasmin inhibitor) and thrombin activatable
(Fig. 3). Finally, αIIbβ is also critical for platelet-​mediated fibrinolysis inhibitor51.
clot retraction, a process that helps seal the injury site
and initiate wound healing40. Thrombosis
Vascular injury exposes TF, a transmembrane protein Excessive activation of platelets and coagulation leads to
that serves as a receptor for FVII/FVIIa21. The TF–FVIIa the formation of thrombi in blood vessels that partially
complex is the physiological trigger of blood coagula- or completely block blood flow23,26. For instance, rupture
tion. TF is expressed by adventitial cells that surround of an atherosclerotic plaque exposes blood to collagen,
blood vessels and forms a haemostatic envelope to vWF and TF and leads to atherothrombosis9 (Fig. 1). TF is
limit blood loss41. TF surrounding blood vessels, except also induced in peripheral blood mononuclear cells after
in the brain, has bound FVII that allows rapid initiation total knee arthroplasty and likely contributes to VTE52.
of coagulation after vessel injury42,43. Components of Indeed, TF appears to play a role in all forms of throm-
the intrinsic pathway (FVIII, FIX and FXI) are required bosis, with the exception of those triggered by artificial
for amplification of thrombin generation because the surfaces such as mechanical heart valves21. FXII contrib-
TF–FVIIa complex is rapidly inactivated by TFPI44. utes to thrombosis but not haemostasis and undergoes
Indeed, individuals with deficiencies of these proteins autoactivation on negatively charged surfaces, such as

www.nature.com/nrd
Reviews

silica or kaolin (the FXII activators used in the activated Clopidogrel used to be the most commonly used
partial thromboplastin time (aPTT) assay), and bioma- P2Y12 antagonist in DAPT, but this is being replaced
terials such as catheters, mechanical heart valves and with the more potent agents prasugrel and ticagre-
extracorporeal circuits. More recently, natural negatively lor. Occasionally, αIIbβ3 antagonists are used for ACS
charged polyphosphates — RNA and DNA released from and/or PCI, but these agents are used less than they
damaged or activated cells, inorganic phosphate released were in the past.
from activated platelets and the DNA portion of neutro- Approved antiplatelet agents either prevent the
phil extracellular traps (NETs) extruded from activated generation of second messengers (thromboxane A2),
neutrophils — have been shown to activate FXII53,54. inhibit receptors activated by soluble agonists (P2Y12,
In addition, NETs, which are present in both AT and PAR1) or block the binding of fibrinogen and other
VT in humans54,55, have been shown to contribute to VT ligands to αIIbβ3 (Box 1, Fig. 2). However, because all
in a mouse model56. Studies with mice also showed that these pathways are required for platelets to maintain
FXII and FXI contribute to AT, VT and atherothrombo- haemostasis, the major side effect of existing antiplatelet
sis57–60. Likewise, FXII or FXI knockdown or inhibition agents is bleeding. The risk of major bleeding (generally
attenuates catheter thrombosis in rabbits and clotting on defined as bleeding that requires medical attention) is
arteriovenous shunts in baboons53,61. Furthermore, FXIIa approximately 1% per year with aspirin and all the P2Y12
inhibition prevented clotting to the same extent as hep- inhibitors15,67. In general, the use of antiplatelet agents
arin in a rabbit extracorporeal membrane oxygenation that are more potent than aspirin or the combination
(ECMO) model but was associated with less bleeding62. of antiplatelet agents, are associated with a concomitant
Because the formation of haemostatic plugs and throm- increase in bleeding67. There are no guidelines on the
botic clots are regulated by many of the same pathways, reversal of antiplatelet therapy68. In general, it is thought
it is difficult to separate protective haemostasis from that platelet transfusion is ineffective at reversing the
pathologic thrombosis. antiplatelet effects of P2Y12 inhibitors69,70. One study
Dysregulation of the fibrinolytic system can con- found that platelet transfusions restored platelet function
tribute to thrombosis. For instance, elevated levels of in patients treated with aspirin but not in those treated
PAI1 and thrombin activatable fibrinolysis inhibitor in with clopidogrel71. However, the Platelet Transfusion
disorders such as obesity are associated with thrombo- versus Standard of Care after Acute Stroke due to
sis63,64. In addition, a polymorphism in the PAI1 gene is Spontaneous Cerebral Haemorrhage Associated with
associated with VTE65. Antiplatelet Therapy (PATCH) trial found that platelet
It is important to remember that bleeding and transfusion increased the risk of death compared with
thrombosis do not usually occur in the same vascular a non-​transfused group72. Ticagrelor is the only plate-
territories. Haemostatic plugs repair the vascular wall let inhibitor that has a reversal agent — a monoclonal
and have little impact on blood flow, whereas patho- antibody fragment called PB2453 that binds to ticagre­
logical thrombi grow without stopping, leading to vas- lor and its metabolite with high affinity70. Aspirin and
cular occlusion and clinical manifestations. Increased P2Y12 inhibitors, such as clopidogrel and prasugrel, have
amounts of soluble accessible matrix components and short half-​lives in the circulation but bind to their tar-
soluble activators of platelets and procoagulants, as well gets in an irreversible manner such that their inhibitory
as loss of endothelial inhibitory mechanisms in diseased effects last the lifetime of the platelets. Transfusion of
vessels, could be responsible for the deregulated growth
of pathogenic thrombi. Antithrombotics need to be pow- Box 2 | Antiplatelet therapy for ischaemic stroke
erful enough to prevent thrombi in such prothrombotic
antiplatelet therapy is effective as secondary prevention
environments, yet of course they can cause bleeding in
in patients with non-​cardioembolic transient ischaemic
damaged healthy vessels. attack or non-​cardioembolic ischaemic stroke, but the
most effective agents and optimal duration remain
Clinical experience with antithrombotics controversial. the 2014 Guidelines for the Prevention
Antiplatelet agents of stroke in Patients with stroke and transient ischaemic
AT is prevented and treated with antiplatelet agents20,66 attack238 recommended an aspirin dose of 325 mg per day
(Box 1, Fig. 2). These agents can be divided into paren- (with the option of aspirin plus clopidogrel for 90 days)
teral and oral agents. Parenteral agents, such as αIIbβ3 in patients with a stroke or transient ischaemic attack
antagonists or cangrelor, are generally used to prevent caused by 50% to 99% stenosis of a major intracranial
or treat acute thrombosis. Single antiplatelet treatment artery. these recommendations are supported by a
2002 meta-​analysis from the antithrombotic trialists
(SAPT) with oral agents (aspirin or a P2Y12 inhibitor
Collaboration, which analysed 195 randomized controlled
such as clopidogrel) is generally used for secondary trials comparing antiplatelet therapy, primarily aspirin,
prevention in patients with stable coronary artery dis- with placebo in the prevention of stroke, myocardial
ease (CAD), PAD, non-​cardioembolic stroke or transient infarction and vascular death239. in the subset of patients
ischaemic attack (TIA). By contrast, patients with acute with prior cerebrovascular disease, antiplatelet therapy
coronary syndrome (ACS) and/or undergoing percuta- reduced the risk of secondary stroke, myocardial infarction
neous coronary intervention (PCI) receive short-​term or vascular death by 22%. For most patients with ischaemic
treatment with an anticoagulant (heparin or a parenteral stroke, the long-​term use of aspirin plus clopidogrel does
DTI such as bivalirudin) and then dual antiplatelet not have a greater benefit for stroke prevention than
treatment (DAPT) — the combination of aspirin and either agent alone but does substantially increase the risk
of bleeding complications.
a P2Y12 inhibitor — for secondary prevention (Table 1).

Nature Reviews | Drug Discovery


Reviews

Box 3 | A brief history of anticoagulants


there are four major classes of anticoagulants used for the prevention and treatment of venous thromboembolism (vte)
and the prevention of stroke in patients with atrial fibrillation (aF), namely heparins (parenteral), vitamin K antagonists
(vKas; oral), direct thrombin inhibitors (Dtis; parenteral and oral) and direct Factor Xa (FXa) inhibitors (oral)196 (Fig. 2).
the first anticoagulant, heparin (also referred to as unfractionated heparin), was originally isolated from liver in 1918
(ref.240). a protocol for its purification was described 15 years later241. Heparin is a naturally occurring glycosaminoglycan
with a negative ionic charge synthesized by basophils and mast cells. Heparin was first used as an anticoagulant
in humans in 1937 (refs242,243). the heparin family of anticoagulants now includes heparin, low-​molecular-​weight
heparin (LMwH), which is generated by chemical or enzymatic degradation of heparin, and fondaparinux (a synthetic
pentasaccharide)244, all of which are administered parenterally245. enoxaparin was the first LMwH to be approved by the
FDa in 1993 (with a generic version gaining approval in 2010)246, while fondaparinux was approved in 2001 (ref.247).
several additional LMwH preparations have been approved by the FDa and have antithrombotic activities like that of
enoxaparin yet with distinct biochemical and pharmacological profiles.
Heparin has several limitations, including heparin-​induced thrombocytopenia (Hit), a limb-​threatening and
life-​threatening side effect in which antibodies form against complexes of heparin and platelet factor 4 and induce
platelet activation and a prothrombotic state. Fortunately, the risk of Hit is lower with LMwH than with heparin and
almost non-​existent with fondaparinux248. a challenge with the use of heparin is the significant patient-​to-​patient
variation in its anticoagulant effect. By contrast, LMwH and fondaparinux have more predictable effects on coagulation
and thus can be administered in weight-​based doses without the need for coagulation monitoring249.
all heparins exert their anticoagulant effects by enhancing the activity of antithrombin, a naturally occurring inhibitor
of coagulation250. Heparin enhances antithrombin inhibition of both FXa and thrombin, whereas LMwHs are more
selective for FXa, and fondaparinux only inhibits FXa. therefore, heparins are indirect FXa and thrombin inhibitors.
Notably, positively charged protamine sulfate completely reverses the anticoagulant activity of heparin but only partly
reverses the anticoagulant activity of LMwH and has no effect on fondaparinux activity251.
vKas were the first oral anticoagulants. the anticoagulant activity of dicoumarol (3,3’-methylene-​bis[4-​hyroxycoumarin])
was discovered in 1940 (ref.252), and warfarin was approved by the FDa in 1954 (ref.253). Like all 4-​hyroxycoumarin drugs,
dicoumarol and warfarin are competitive inhibitors of vitamin K epoxide reductase; this leads to a reduction in vitamin K,
which is required for post-​translational modification of glutamic acid residues to carboxyglutamic acid254. several
proteins in the coagulation cascade require this modification to create a positively charged Gla domain, which enables
their binding to negatively charged phospholipids exposed on damaged or activated cells255. vKas induce a transient
prothrombotic state owing to inhibition of anticoagulant proteins, followed by an anticoagulant state due to inhibition
of procoagulants256. vitamin K reverses the effect of warfarin257. vKas interact with multiple drugs and their activity
is affected by dietary vitamin K intake. Because of these interactions, vKas require frequent monitoring and dose
adjustments. in addition, polymorphisms in CYP2C9 and vKOrC1 account for up to 50% of the interindividual variability
of warfarin dosing, which led to a label change in 2007 (refs258,259).
Dtis can be divided into parenteral (bivalirudin (FDa approved in 2000), lepirudin and argatroban) and oral (dabigatran)
agents. Bivalirudin, lepirudin and argatroban are used to treat patients with Hit260. Dabigatran etexilate, a prodrug of
dabigatran, was the first direct oral anticoagulant to be developed and was approved by the FDa in 2008 (ref.261).
idarucizumab is an inhibitory monoclonal antibody fragment that binds dabigatran with an affinity 350-​fold greater than
the affinity of dabigatran for thrombin262–265. idarucizumab was approved by the FDa in 2015 for dabigatran reversal.
there are four direct oral FXa inhibitors (rivaroxaban, apixaban, edoxaban and betrixaban) that were approved by the
FDa between 2008 and 2017 for the prevention and treatment of vte and for the prevention of stroke in patients with
aF196,266. andexanet alfa is a catalytically inactive FXa lacking the membrane-​binding γ-​carboxyglutamic acid domain that
acts as a decoy to bind FXa inhibitors267. andexanet alfa restored haemostasis in patients treated with a FXa inhibitor who
presented with acute major bleeding268. andexanet alfa was FDa-​approved in 2018 for the reversal of all FXa inhibitors269.
as a class, the direct oral anticoagulants are at least as effective as warfarin for stroke prevention in patients with aF or
for the treatment of vte, but they are associated with a 50% reduction in intracranial bleeding and are more convenient
to administer270. Nonetheless, the major side effect of all currently approved anticoagulants is bleeding.

sufficient numbers of normal platelets should replace Another key challenge in the use of current anti-
the inhibited platelets and enable haemostasis. By con- platelet therapies is related to the variability in patient
trast, ticagrelor and its active metabolite bind reversibly response and side effects. Indeed, numerous studies
to P2Y12. Despite the reversible binding, it takes 4 or have demonstrated substantial interpatient variability
5 days for recovery of platelet function after ticagrelor in platelet inhibition in those treated with clopidogrel,
is stopped. Platelet transfusion is ineffective for patients depending on the method of testing and the defini-
treated with ticagrelor because free ticagrelor is available tion of response73. In addition, patients with type 2
to bind to the transfused platelets. Therefore, there is a diabetes have increased platelet reactivity and are less
need for a reversal agent for ticagrelor but not for other responsive to antiplatelet therapy74. While prasugrel
agents. Other concerns with the use of current antiplate- and ticagrelor exhibit less variability than clopidogrel,
let agents are as follows: up to 30% of patients are not prasugrel has been associated with higher rates of
responsive to clopidogrel; prasugrel is contraindicated major bleeding and ticagrelor has been associated
in patients ≥75 years of age, with a weight <60 kg or with with higher rates of non-​coronary artery bypass graft
a prior history of stroke; and ticagrelor can cause dysp- surgery-​related bleeding in head-​to-​head comparisons
noea and is contraindicated in patients with a history of with clopidogrel. In addition, both agents are more
intracranial haemorrhage. expensive than clopidogrel and are associated with

www.nature.com/nrd
Reviews

higher discontinuation rates75. As a result, clopidogrel a minority of patients of European ancestry carry the
remains the most commonly prescribed P2Y12 inhibi- cytochrome P450 (CYP; CYP2C19) loss-​of-​function
tor in clinical practice, accounting for over 50% of filled allele that is associated with decreased platelet respon-
prescriptions after PCI in 2016 (ref.75). siveness to clopidogrel78. This gene–clopidogrel response
Individuals exhibit a broad range of platelet reactiv- relationship was first reported in 2007, but it took more
ity, which makes it difficult for clinicians to know the than a decade to perform a clinical trial. Importantly, the
best agent and dose to use. Attempts have been made POPular Genetics trial found that the use of a CYP2C19
to develop point-​of-​care ADP-​specific platelet reactiv- genotype-​guided strategy (carriers received ticagrelor
ity tests such as PFA-100, VerifyNow, and Multiplate or prasugrel and non-​carriers received clopidogrel) to
Electrode Aggregometry76. A meta-​analysis of 10 clini- guide the selection of oral P2Y12 inhibitor therapy in
cal trials comparing a standard dose of clopidogrel with patients undergoing PCI was non-​inferior to treatment
an intensified dose based on platelet reactivity testing with ticagrelor or prasugrel at 12 months in the primary
showed a reduction in cardiovascular mortality, stent outcomes but with a lower incidence of bleeding79. This
thrombosis and MI; however, this difference was driven suggests a benefit of using a CYP2C19 genotype-​guided
by a decrease in stent thrombosis in the intensified dose strategy for antiplatelet therapy. The success of this trial
group77. These results suggest that more work is needed should lead to avoidance of clopidogrel use in carriers
to improve point-​of-​care platelet reactivity testing, which of the CYP2C19 loss-​of-​function allele, and a CYP219
would allow better use of current antiplatelet agents. genotype-​guided strategy is gaining wider acceptance for
Another approach to improving the use of antiplate- use in clinical practice.
let drugs is genetic testing. For instance, it is known that Another challenge in antiplatelet therapy is defining
the optimal duration of DAPT in patients with ACS,
Soluble agonists particularly those receiving an intracoronary stent.
With first-​generation drug-​eluting stents, long-​term or
Thrombin lifelong DAPT was recommended because of reports
of late stent thrombosis80. With the newer generations of
ADP TxA2 drug-​eluting stents, vascular healing is improved and
BMS-986141 PAR1 PZ-128 P2Y1 stent thrombosis is less common, enabling the duration
PAR4 P2Y12 of DAPT to be reduced, especially in patients at high
BMS-986120
risk of bleeding. The 2016 ACC/AHA Guideline Focused
TP Update on Duration of Dual Antiplatelet Therapy in
Patients with Coronary Artery Disease recommended
6 months of DAPT in patients with stable CAD who
received an intracoronary stent and 12 months for
patients with ACS treated medically or who received
an intracoronary stent81. The 2017 European guidelines
PI3Kβ were similar and recommended 1–6 months (depending
AZD6482 on bleeding risk) of DAPT in patients with stable CAD
and 12 months for patients with ACS82. Several recent
Activation
studies have provided more evidence that 1 month of
BTK DAPT is sufficient in patients with stable CAD at high
bleeding risk 83. Whether the duration of DAPT in
GPVI
patients with ACS can be reduced to less than 1 year is
Fcγ CLEC2 αIIbβ3
ACT017 unknown and will require further study.
These problems associated with existing antiplatelet
agents have led to a search for new platelet targets.
Revacept Collagen
GPIb–V–IX
Anticoagulants
Fibrinogen
Anticoagulants are divided into those that require par-
PDPN
• AJW200 enteral administration and those that are given orally
• ALX-0081 vWF Platelet aggregation (Box 3, Fig. 2). The commonly used parenteral antico-
• ARC1779
agulants include heparin, LMWH, fondaparinux and
Collagen bivalirudin. The commonly used oral anticoagulants
are VKAs and DOACs. Heparins are used as a first line
Adhesion receptors therapy to treat acute thrombotic events. Both parenteral
(such as heparins) and oral agents (such as VKAs and
Fig. 3 | Targets of antiplatelet agents in development. Platelet receptors can be DOACs) are used to prevent and treat VT. Because of
divided into three groups: adhesion receptors (glycoprotein Ib (GPIb) and integrins),
the preponderance of fibrin in venous thrombi and in the
insoluble ligands (GPVI), soluble agonist receptors (thromboxane receptor (TP), P2Y1,
P2Y12, PAR1 and PAR4) and an aggregation receptor (αIIbβ3). In addition, intracellular thrombi that form in the left atrial appendage in patients
kinases (Bruton’s tyrosine kinase (BTK) and phosphoinositide 3-​kinase-​β (PI3Kβ)) are with AF, anticoagulants are the cornerstone for the pre-
required for platelet activation. The names and targets of antiplatelet agents in vention and treatment of VTE and for stroke preven-
development are shown. CLEC2, C-​type lectin-​like 2; PDPN, Podoplanin; TxA2, tion in patients with AF (Box 3, Table 1). Anticoagulants
thromboxane A2; vWF, von Willebrand factor. are also used to prevent clotting on mechanical heart

Nature Reviews | Drug Discovery


Reviews

PolyP, nucleic acid unaffected by genetic polymorphisms or variations in


dietary vitamin K intake and there are fewer drug–drug
interactions. Therefore, DOACs are more convenient to
FXIIa CSL312
administer than VKAs. In randomized clinical trials that
included over 100,000 patients, DOACs were shown to
• IONIS-416858
• Osocimab Intrinsic
be at least as effective as VKAs for the prevention of
Extrinsic TF FVIIa FXIa • AB023 stroke in patients with AF and for the treatment of VTE
pathway pathway
• BMS-986177 but were associated with less bleeding, and in particular
less intracranial bleeding. Given their similar efficacy,
FIXa greater safety and convenience compared with VKAs,
DOACs are now given preference over VKAs for AF and
VTE indications in recent guidelines87.
FXa Nevertheless, although DOACs are a major advance
over VKAs, there are several issues with DOACs, including
Common increased bleeding in some patient populations, their ina-
pathway bility to be used in certain patients and a lack of efficacy
Platelet
activation Thrombin FXIIIa in others. For instance, rates of gastrointestinal bleeding
are higher with DOACs than with VKAs. Studies compar-
ing either edoxaban or rivaroxaban versus dalteparin for
Fibrin monomers
the prevention of recurrent VTE in patients with cancer
observed an increase in bleeding in patients with gas-
Crosslinked fibrin trointestinal cancers treated with DOACs compared to
dalteparin88. Furthermore, even though the risk of major
Fig. 4 | Targets of anticoagulant agents in development. The coagulation cascade can
be divided into three parts: extrinsic (tissue factor (TF), factor VIIa (FVIIa)), intrinsic (FXIIa,
bleeding is 50% lower with DOACs than with VKAs, the
FXIa, FIXa and FVIIIa) and the common pathway (FXa, FVa and thrombin). Activation of annual rates of major bleeding with DOACs can be as
the coagulation cascade leads to the generation of thrombin. Thrombin plays a central high as 2–3% in elderly patients with AF89. All DOACs
role in the coagulation cascade by cleaving fibrinogen to fibrin monomers, activating are cleared through the kidneys and, therefore, there is a
FXIII, which then crosslinks fibrin monomers, and by activating the cofactors FV and FVIII. risk of drug accumulation and bleeding in patients with
In addition, thrombin is a potent activator of platelets. All coagulation factors except renal dysfunction. All DOACs have different recommen-
FXII are required for haemostasis. Components of the extrinsic and common pathways dations for use in patients with impaired renal function,
are essential for haemostasis, whereas components of the intrinsic pathway are required with a dose reduction for renal impairment. However,
to amplify thrombin generation. Anticoagulant agents in development target either FXII, apixaban has recently been licenced for patients with
FXI, FIX or activation of FXI by FXIIa.
end-​stage renal disease (ESRD)90. Like VKAs, DOACs
pass through the placenta, which could pose a risk to
valves or on extracorporeal circuits such as those used the foetus; consequently, DOACs are contraindicated
for haemodialysis, cardiopulmonary bypass surgery in pregnancy. In addition, unlike VKAs, DOACs may
and ECMO. pass into breast milk and are therefore also contraindi-
Although VKAs are effective, they are difficult to cated in nursing mothers. There is a black box warning
manage because of within-​patient and between-​patient against the use of DOACs in patients with mechanical
differences in dosing. These differences reflect a combi- heart valves because a study comparing dabigatran with
nation of factors, including common genetic polymor- warfarin for this indication showed a trend for more
phisms in CYP2C9 and vitamin K epoxide reductase thromboembolic and bleeding events with dabigatran91.
complex subunit 1 (VKORC1) (which account for ~30% Consequently, VKAs, such as warfarin, remain the only
of the interindividual variation) that affect the metabo- approved anticoagulants for this patient population.
lism of VKAs, variations in dietary intake of vitamin K In addition, VKAs remain the anticoagulant of choice for
and multiple drug interactions84,85. Because of this vari- patients with triple-​positive antiphospholipid syndrome
ability, frequent monitoring of the international normal- (positive for lupus anticoagulant, anticardiolipin and
ized ratio and dose adjustments to maintain it within the anti-​β-​glycoprotein I antibodies) following the early ter-
therapeutic range are necessary. The many limitations mination of a study comparing rivaroxaban and warfarin
of VKAs prompted a long search for oral anticoagulants in such patients due to excess thromboembolic events in
that were at least as effective as VKAs, but could be the rivaroxaban arm (NCT02157272)92. Finally, DOACs
administered in fixed doses without the need for routine also have more drug–drug interactions than LMWHs93.
coagulation monitoring. Therefore, there is still a need for safer anticoagu-
DOACs overcome many of the limitations of VKAs86. lants, particularly those with minimal renal clearance
DOACs are direct anticoagulants because they bind and the capacity to attenuate clotting induced by medi-
to the active site of their target enzyme and block its cal devices such as catheters, heart valves, left ventricle
activity. Thus, dabigatran inhibits thrombin, whereas assist devices and extracorporeal circuits.
apixaban, betrixaban, edoxaban and rivaroxaban
inhibit FXa. In contrast to VKAs, DOACs are given in Combination antiplatelet and anticoagulant therapy
fixed doses without monitoring. Such dosing is possible ACS, CAD and PAD. More potent antiplatelet agents
because DOACs produce a more predictable anticoag- and/or the use of multiple antiplatelet therapies sig-
ulant response than VKAs because their metabolism is nificantly increase the risk of bleeding, which suggests

www.nature.com/nrd
Reviews

that we have reached the limit for targeting platelets event in 2013. However, it has not been widely adopted
alone in patients with CVD94–96. Fibrin is also present owing to concerns with bleeding.
in AT. The concept of combining antiplatelet and anti- DPI has also been investigated in patients with stable
coagulant therapy to prevent thromboembolic events in CAD and PAD. The standard therapy for these patients
patients with CVD was examined more than 10 years is a single antiplatelet agent, either aspirin or clopi-
ago. A meta-​analysis of 10 trials concluded that addition dogrel. The Cardiovascular Outcomes for People Using
of a VKA to aspirin decreased MI and ischaemic stroke Anticoagulation Strategies (COMPASS) trial compared
in patients with ACS compared with aspirin alone, but the effect of three treatments: aspirin alone, rivaroxaban
increased major bleeding97. However, these studies do alone and the combination of the two agents in patients
not reflect current practice because they were performed with stable CAD or PAD100–102. The trial was stopped early
before the widespread use of PCI and DAPT. owing to the overwhelming benefit of DPI compared
Because DOACs are associated with a lower risk of with aspirin alone on the primary outcome (the com-
intracranial bleeding compared with VKAs, the concept posite of cardiovascular death, MI or stroke) being signif-
of dual pathway inhibition (DPI) — combined anti- icantly reduced100,102. Although major bleeding (mostly
platelet and anticoagulant therapy — was revisited. Two gastrointestinal bleeding) was significantly increased in
trials determined the effect of adding a FXa inhibitor to the aspirin plus rivaroxaban group compared with the
standard antiplatelet therapy in patients with ACS. The aspirin alone group, there was no significant increase in
first trial, APPRAISE-2, which administered apixaban fatal bleeding or intracranial bleeding. Notably, the net
at the same dose used to prevent stroke in patients with clinical benefit of DPI was greater in patients with PAD
AF or to treat patients with VTE98, resulted in signifi- compared with patients with CAD, as well as in patients
cantly increased major bleeding without reducing the with PAD with the highest risk of atherothrombotic
primary outcome of cardiovascular death. The second events102. In 2018, the FDA approved the combination
trial, ATLAS ACS 2–TIMI 51 (ref.99), which used two of aspirin and rivaroxaban for patients with CAD or
doses of rivaroxaban that were lower than those used PAD. The results of the COMPASS trial are exciting and
to treat or prevent VTE or for stroke in patients with support the notion that a combination of an antiplate-
AF, the primary end point of cardiovascular death, MI let and anticoagulant agent may be more efficacious in
and ischaemic stroke was reduced with both doses of reducing cardiovascular death, MI or stroke than a single
rivaroxaban compared with standard antiplatelet ther- agent for some indications. The VOYAGER trial is cur-
apy alone. However, as expected, rivaroxaban increased rently evaluating the effect of rivaroxaban on acute limb
rates of major bleeding and intracranial bleeding. The ischaemia or major amputations as well as cardiovascular
European Commission approved the use of rivaroxaban death, MI and stroke in patients with PAD undergoing
for the secondary prevention after an acute coronary revascularization procedures (NCT02504216).

Table 1 | use of current antithrombotics


indication subtype Agent comments
Arterial thrombosis Coronary stable CAD ASA or ASA + riva Need for new agents
ACS DAPT
PCI high risk AP + AC (heparin or DTI)
Cerebral ASA
Peripheral ASA or ASA + riva
VTE – AC (DOAC, VKA, heparins) Guidelines give preference to
DOAC vs VKA treatment
AF – AC (DOAC, VKA) Guidelines give preference to
DOAC vs VKA treatment
APS – AP + AC –
Medical devices — mechanical – VKA Black box warning against the
heart valves and LVADs use of DOACs for use with
mechanical vent valve
Extracorporeal circuits — – Heparin FXIIa and FXIa inhibitors may be
cardiopulmonary bypass, associated with less bleeding
haemodialysis and ECMO
Prevention of recurrent VTE in – AC (DOAC, LMWH, VKA) DOAC > LMWH > VKA
patients with cancer
Prevention of recurrent stroke – ASA Heparin, LMWH and heparinoids
have failed; need for new agents
AC, anticoagulant; ACS, acute coronary syndrome; AF, arterial fibrillation; AP, antiplatelet; APS, antiphospholipid syndrome;
ASA, acetylsalicyclic acid (aspirin); CAD, coronary artery disease; DAPT, dual antiplatelet treatment; DOAC, direct oral anticoagulant;
DTI, direct thrombin inhibitor; ECMO, extracorporeal membrane oxygenation; F, factor; LMWH, low-​molecular-​weight heparin;
LVAD, left ventricular assist device; PCI, percutaneous coronary intervention; riva, rivaroxaban; VKA, vitamin K antagonist;
VTE, venous thromboembolism.

Nature Reviews | Drug Discovery


Reviews

Interestingly, addition of the PAR1 inhibitor vora- APC has both anticoagulant and cytoprotective
paxar to SAPT or DAPT for patients with CAD or PAD activity via activation of PAR1 on the endothelium108,109.
also reduced major adverse limb events and revascu- 3K3A-​APC is a variant of APC with full cytoprotective
larization in patients with PAD, but it did not reduce activity but significantly reduced anticoagulant activity.
mortality95. Further studies are needed to better under- This variant was protective in a mouse stroke model110.
stand the mechanism by which combined antiplatelet A phase II study showed that addition of 3K3A-​APC to
and anticoagulant therapy as well as PAR1 inhibition is tPA for the treatment of acute stroke was associated with
protective in patients with CAD or PAD. It is possible a trend towards less bleeding, possibly owing to protec-
that DPI reduces both thrombosis and PAR-​dependent tion of the endothelium111. An ongoing phase II trial
vascular inflammation. is comparing the effect of 3K3A-​APC with placebo in
tPA treatment, medical thrombectomy or both in acute
Patients with CVD and AF. Patients with CVD and ischaemic stroke (NCT02222714).
AF require both antiplatelet agents and anticoagulants Current guidelines recommend systemic throm-
as they are at risk for both AT and VT. In the past, bolytics for the treatment of massive PE but not for
patients with AF who experience an ACS or underwent sub-​massive PE unless the patient deteriorates after
PCI were treated with DAPT plus an anticoagulant beginning anti-​coagulation and the bleeding risk is low87.
(labelled as triple therapy). Five recent studies (WOEST, Interestingly, the MOPETT trial showed that low-​dose
PIONEER AF-​PCI, RE-​DUAL PCI, AUGUSTUS and tPA significantly reduced the combined end point of
ENTRUST-​AF-​PCI) examined the effects of using SAPT death plus recurrent PE in patients with moderate PE112.
or DAPT plus an anticoagulant (VKA or DOAC). A Another study found that both half-​dose systemic throm-
meta-​analysis of four of these studies103 compared the bolysis and ultrasound-​facilitated catheter-​directed
efficacy and safety of the different groups. There were no fibrinolysis were beneficial in the treatment of patients
statistically significant differences in the rates of major with PE but systemic thrombolysis was more cost effec-
adverse cardiovascular events (MACE) amongst the dif- tive113. More recent trials have suggested that low-​dose
ferent groups, but there was more bleeding in the VKA tPA delivered directly into the pulmonary arteries by a
plus DAPT group. In addition, strategies omitting aspi- catheter that provides high-​frequency, low-​power ultra-
rin caused less bleeding, without a significant difference sound (termed ultrasound-​enhanced thrombolysis)
in MACE, compared with strategies including aspirin. can decrease right ventricle dilation, reduce pulmonary
This indicates that DOACs are safer than VKAs when arterial pressures and decrease thrombus burden114.
used in combination with antiplatelet agents and that Thrombolytic agents are also a beneficial treatment
aspirin might not be necessary in patients treated with a for acute ST-​segment elevation myocardial infarction,
DOAC or VKA and a P2Y12 inhibitor. although their use has largely been supplanted by PCI.
The AFFIRE study of 2,236 patients in Japan found
that rivaroxaban monotherapy was non-​inferior to Antiplatelet drugs in development
dual therapy (rivaroxaban plus an antiplatelet agent) in A number of antiplatelet targets and associated agents
patients with AF and CAD, with neither requiring inter- are currently being evaluated in clinical trials (Table 2).
vention for more than 1 year after revascularization104.
The primary efficacy end point was stroke, systemic Inhibitors of thrombin-​mediated platelet activation
embolism, MI, unstable angina requiring revasculari- Thrombin activates human platelets by proteolytic
zation and all-​cause mortality, while the primary safety cleavage of two G protein-​coupled receptors, PAR1 and
end point was major bleeding. The study was stopped PAR4115,116 (Fig. 3). PAR1 is activated by low concentra-
early because of higher rates of all-​cause mortality in tions of thrombin (sub-​nanomolar) and produces a rapid
the dual therapy arm. Therefore, a single anticoagulant, but transient signal, whereas PAR4 requires ~10-​fold
rivaroxaban, was superior to dual therapy in this patient more thrombin for its activation and mediates pro-
population. longed signalling that is required for stable thrombus
formation. Early studies focused on developing PAR1
Fibrinolytic therapy antagonists and culminated in the FDA approval of vora-
Acute ischaemic stroke is treated with fibrinolytic ther- paxar in 2014 (Box 1). However, vorapaxar is a slowly
apy and/or mechanical thrombectomy. tPA is approved reversible inhibitor that may interfere with thrombin
for use within 3 h of onset and there are data that out- activation of platelets during haemostasis. Therefore,
comes are improved when administered within 4.5 h of recent efforts have been made to develop rapidly revers-
the onset of symptoms105. As with antithrombotics, the ible PAR1 antagonists and PAR4 inhibitors that may
major side effect of fibrinolytics is bleeding. Studies have have less effect on haemostasis. Importantly, PAR1 and
shown that treatment of MI and PE with systemic throm- PAR4 are expressed on numerous cell types in the vas-
bolysis is associated with a 1–1.5% risk of intracerebral culature, including endothelial cells117. Therefore, inhi-
bleeding106,107. tPA also affects blood–brain barrier per- bition of PAR1 and PAR4 may have effects on cells other
meability13. The ongoing MOST trial (NCT03735979) than platelets.
is determining whether combining tPA with either
the direct thrombin inhibitor argatroban or with the PAR1 antagonists. PZ-128 is a pepducin and there-
αIIbβ3 antagonist eptifibatide is superior to tPA alone fore represents a second class of PAR1 inhibitors
in improving the 90-​day Rankin score for patients with with a distinct mechanism of action. Pepducins are
acute ischaemic stroke. membrane-​ tethered, cell-​ p enetrating lipopeptides

www.nature.com/nrd
Reviews

Table 2 | Antiplatelet agents in development


Name/company Agent/route of Target Disease clinical trial Trial iD comments
administration
PZ-128/Tufts Medical Pepducin/IV PAR1 NA Phase I NCT01806077 Completed; safety study
Center
BMS-986120/ Small molecule/oral PAR4 NA Phase I NCT02439190 Completed; safety study
Bristol-​Myers Squibb
BMS-986141/ Small molecule/oral PAR4 NA Phase I NCT02341638 Completed; safety study
Bristol-​Myers Squibb
Recurrent stroke Phase II NCT02671416 Study terminated early
Revacet/Advance Cor Fusion protein/IV GPVI ligand Stable CAD Phase I NCT01042964 Completed; safety study
undergoing PCI
Phase II NCT03312855 Completed; no data
Symptomatic Phase II NCT01645306 Ongoing
carotid stenosis
ACT017/Acticor Biotech Antibody/IV GPVI NA Phase I NCT03803007 Completed; safety study
ARC1779/Archemix DNA aptamer/IV vWF NA Phase I NCT00432770 Completed; safety study
Cerebral Phase II NCT00742612 Completed; reduced cerebral
embolization embolization
AZD6482/AstraZeneca Small molecule/IV PI3Kβ NA Phase I NCT00688714 Completed; safety study
Phase I NCT00853450 Completed; safety study
Isoquercetin/Beth Israel Small molecule/oral ND NA Phase I NCT01722669 Completed; safety study
Isoquercetin/NHLBI Small molecule/oral ND VTE in advanced Phase II/III NCT02195232 Completed; reduced D-​dimer
cancer and P-​selectin
CAD, coronary artery disease; GPVI, glycoprotein VI; IV, intravenous; NA, not available; ND, not determined; NHLBI, National Heart, Lung, and Blood Institute; PAR,
protease-​activated receptor; PCI, percutaneous coronary intervention; PI3Kβ, phosphoinositide 3-​kinase β; vWF, von Willebrand factor; VTE, venous thromboembolism.

that target the cytoplasmic domain of receptors118,119. haemostatic plug formation. A variety of agents, such as
PZ-128 was developed as a rapidly acting, reversible inhibitory antibodies, pepducins and small molecules,
antiplatelet agent to prevent ischaemic events associated have been developed against PAR4.
with PCI. The safety, efficacy and pharmacokinetics of BMS-986120 is a potent and reversible PAR4-​specific
PZ-128 were evaluated in a phase I study in patients small molecule antagonist123 (Fig. 3, Table 2) that blocks
with vascular disease or who had two or more CAD platelet activation by γ-​thrombin or a PAR4 activa-
risk factors (NCT01806077)120. Intravenous adminis- tion peptide. In a non-​human primate thrombosis
tration of PZ-128 inhibited PAR1-​dependent platelet model, BMS-986120 and clopidogrel decreased throm-
activation ex vivo but not activation by other agonists, bus weight to a similar extent but, unlike clopidogrel,
such as ADP, and did not cause bleeding. Further tri- which prolonged the bleeding time, BMS-986120
als are planned with PZ-128 in high-​risk patients with had little effect123. In a phase I study, orally adminis-
CAD or ACS undergoing PCI (A. Kuliopulos, personal tered BMS-986120 had a half-​life of 4 h and selectively
communication). inhibited PAR4-​induced platelet activation ex vivo
Another class of PAR1 inhibitors is the non-​peptidic (NCT02439190) 124. Importantly, administration of
small molecules named parmodulins, which also target BMS-986120 to healthy volunteers did not cause spon-
intracellular domains121. Mechanistic studies demon- taneous bleeding but reduced thrombus growth ex vivo
strated that parmodulins selectively interfere with to a similar extent as aspirin plus clopidogrel. BMS-
Gαq but not Gα12/13 signalling downstream of PAR1. 986141, which is more potent than BMS-986120, was
Consequently, parmodulins inhibit PAR1-​mediated also evaluated in healthy volunteers (NCT02341638).
platelet integrin activation and aggregation. In addition, In addition, a phase II trial was initiated to determine if
parmodulins have also been shown to inhibit proin- BMS-986141 reduced recurrent stroke compared with
flammatory signalling in endothelial cells while pre- placebo in patients with a recent stroke or TIA, all of
serving cytoprotective signalling in endothelial cells122. whom received aspirin (NCT02671461). However, the
No clinical trials have yet been conducted with these study was terminated early for undisclosed reasons.
new agents. A pepducin against PAR4 and an inhibitory PAR4
monoclonal antibody have been generated but neither
PAR4 antagonists. Inhibition of PAR4 blocks the sus- has been evaluated in humans118,119,125.
tained activation of platelets by high concentrations
of thrombin116 — conditions that are more likely to be Antagonists of GPVI collagen-​mediated platelet
present during atherothrombosis — while leaving tran- activation
sient PAR1 signalling intact. The rationale for targeting GPVI is a promising new target for antiplatelet therapy126
PAR4 is that this receptor may be critical for platelet– (Fig. 3). It is the main platelet receptor for collagen127,
platelet cohesion during AT while PAR1 signalling alone and recent studies suggest that it also binds fibrin128,129.
mediates thrombin-​induced platelet adhesion during However, the interaction between GPVI and fibrin is

Nature Reviews | Drug Discovery


Reviews

controversial, and one study concluded that dimeric atherosclerotic arteries and arterioles. Studies in pre-
GPVI is not a receptor for fibrin128,130–133. clinical animal models suggested that the vWF-​GPIbα
Ruptured atherosclerotic plaques are rich in collagen interaction is more important in arterial thrombosis and
and thus provide a robust surface for GPVI-​mediated ischaemic stroke than during haemostatic plug forma-
platelet adhesion. Indeed, several preclinical studies tion145,146. However, one must be cautious when extra­
suggest that GPVI is critical for platelet adhesion to polating results from preclinical models because they do
plaque material under flow conditions134,135 and to rup- not necessarily equate to results in the clinical setting.
tured plaques in vivo134,136. However, the role of type I Increased vWF levels and/or activity can lead to
and type III fibrillar collagen in thrombosis triggered thrombotic microangiopathy, which is thrombosis
by plaque erosion is less clear. Patients lacking func- in capillaries and arterioles due to endothelial injury.
tional GPVI generally exhibit only a mild bleeding For instance, in thrombotic thrombocytopenia pur-
diathesis137 except when they have moderate to severe pura (TTP) there is a reduction in the level of the
thrombocytopenia138. Taken together, these results sug- protease ADAMTS13 (inherited or acquired), which
gest that targeting GPVI may reduce thrombosis caused cleaves ultra-​large vWF multimers into smaller forms.
by ruptured or possibly eroded atherosclerotic plaques Accumulation of these large vWF multimers leads to
with less bleeding than that produced by conventional life-​threatening microvascular thrombosis and thrombo­
antiplatelet agents. However, it remains to be seen how cytopenia147. A similar phenotype is observed in patients
important GPVI is for embolic stroke. with type 2B von Willebrand’s disease, who express
a mutant form of vWF that binds spontaneously to
Inhibitors of GPVI–collagen/fibrin interaction. Two GPIbα148.
agents have been developed to inhibit the function vWF inhibitors that are being investigated include
of GPVI. Revacept targets the GPVI ligand collagen, ARC1779, which is a DNA-​based aptamer, two mono­
whereas ACT017 targets GPVI itself (Fig. 3). clonal antibodies (AJW200 and 82D6A3) and two
Revacept is a fusion protein that contains the extra- bivalent nanobodies (caplacizumab (ALX-0081) and
cellular domain of GPVI fused to a human Fc immuno­ ALX-0681) 149–153. Caplacizumab was demonstrated
globulin region139. Revacept binds to subendothelial to be well tolerated with no signs of bleeding in two
collagen to prevent platelet adhesion and activation. phase I clinical trials (NCT0289733 and NCT03172208).
A recent study found that revacept did not bind to Moreover, caplacizumab was associated with a lower
fibrin132. In a mouse stroke model, revacept improved incidence of recurrence of TTP compared with pla-
the efficacy of tPA without increasing bleeding140. cebo in a phase III trial154, leading to its FDA approval
In addition, revacept dose-​d ependently inhibited in 2019 for adult patients with acquired TTP in com-
collagen-​induced platelet activation without impairing bination with plasma exchange and immunosuppres-
haemostasis in a phase I trial (NCT01042964)141. A com- sive therapy. ARC1779 has a high affinity for the A1
pleted phase II trial evaluated the efficacy of revacept in domain of vWF 150; a phase I trial showed that the
patients with symptomatic carotid artery stenosis, ama­ aptamer dose-​dependently inhibited platelet function
urosis fugax, TIA or stroke (NCT01645306). However, no ex vivo (NCT00432770) 150 whereas a phase II trial
results have been reported. A second ongoing phase II (NCT00742612) showed that it also reduced cerebral
trial is comparing the effect of two doses of revacept embolization after carotid endarterectomy155.
versus placebo on top of DAPT in patients with stable The clinical development of GPIbα inhibitors has
CAD undergoing PCI (NCT03312855)142. lagged behind drugs targeting the vWF A1 domain.
ACT017 is a humanized Fab fragment that has Various agents, including two anti-​GPIbα antibodies
a high antigen-​binding specificity and affinity for (SZ2 and h6B4-​Fab), a snake toxin (anfibatide) and
GPVI143. Injection of ACT017 into macaque monkeys a fusion protein that contains the amino terminus of
dose-​dependently inhibited collagen-​induced plate- GPIbα fused to human IgG1 Fc, showed promise in
let activation ex vivo without inducing thrombocyto­ preclinical testing149,156–158. It should be noted that loss
penia or bleeding144. A phase I study in healthy subjects of GPIbα is associated with increased platelet clearance,
evaluating the pharmacokinetics, pharmacodynam- and this must be avoided when targeting GPIbα159.
ics, safety and tolerability of ACT017 demonstrated Despite the development of several agents that inhibit
favourable safety and tolerability profiles and inhibi- the interaction between vWF and GPIbα, these agents
tion of collagen-​induced platelet aggregation without have not been tested as antiplatelet agents in patients
effects on platelet count or platelet GPVI expression with CVD (except for a small study in patients under-
(NCT03803007)144. A phase II study in patients with going carotid endarterectomy). It will be interesting to
stroke is planned (NCT03803007). Therefore, revacept see if there will be future trials of these agents in patients
and ACT017 represent novel antiplatelet agents that with CVD.
may reduce thrombosis with less bleeding than current
antiplatelet agents. Inhibiting PI3Kβ
The phosphoinositide 3-​kinase-​β (PI3Kβ) member
Inhibitors of vWF–GP1bα-mediated platelet activa- of the PI3K family of lipid kinases plays an impor-
tion. Platelets are captured at sites of vascular injury by tant role in signalling downstream of various platelet
interaction of GPIbα with vWF exposed on the extra- receptors and is required for stable platelet adhesion
cellular matrix (Fig. 3). This interaction is particularly under shear stress160 (Fig. 3). Indeed, p110β-​deficient
important under high shear stress, such as is found in mice have reduced arterial thrombosis161. Two PI3Kβ

www.nature.com/nrd
Reviews

inhibitors, AZD6482 and MIPS-9922, were shown to thrombocytopenia and sepsis174. ML355 is a 12-​LOX
reduce thrombosis in dog and mouse models with min- inhibitor that shows >50-​fold selectivity compared with
imal effects on bleeding162,163. AZD6482 was well toler- other LOX paralogs175. In mice, ML355 was protective
ated in healthy volunteers (NCT00688714)162 and, in against microvascular arterial thrombosis with no effect
combination with aspirin, exhibited greater antiplatelet on tail bleeding175. In vitro, ML355 shows comparable
activity and less bleeding than clopidogrel plus aspirin inhibitory activity towards aggregation of human and
(NCT00688714)164. Further studies are needed to deter- murine platelets, suggesting that studies in mice can be
mine if targeting PI3Kβ will be an effective strategy to translated to humans.
inhibit thrombosis without increasing bleeding. Bruton’s tyrosine kinase (BTK) is expressed in
platelets and is a critical component of the signalling
Inhibiting PDI downstream to GPVI. The small molecule inhibitor,
Thiol isomerases are oxidoreductases best known for ibrutinib, is an oral irreversible BTK inhibitor used to
their role in protein folding. Protein disulfide isomerase treat patients with B cell malignancies176. One of the side
(PDI), a member of the thiol isomerase family, is secreted effects of ibrutinib is low-​grade bleeding events, which
by vascular cells and modifies targets involved in throm- occur in ~35% of treated patients177. Ex vivo studies on
bus formation165. Mechanistic studies demonstrated an collagen-​induced and plaque material-​induced platelet
important role for PDI in platelet activation. The proposed aggregation under static and flow conditions support
mechanism of action includes reshuffling of disulfide the notion that ibrutinib inhibits platelet activation178. The
bonds within key surface receptors such as αIIbβ3 inte- effects of BTK inhibitors on platelet function and their
grin. The flavonol quercetin-3-​rutinoside (isoquercetin) usefulness as antithrombotic agents are currently under
was the first identified PDI inhibitor with antithrombotic investigation.
activity. Administration of isoquercetin protected mice in
several models of thrombosis166. As it is commonly con- Development of new anticoagulants
sumed in fruits, vegetables and food supplements, iso- All current anticoagulant agents either directly or indi-
quercetin quickly moved into phase I (NCT01722669)167. rectly target components of the common pathway —
A subsequent phase II trial demonstrated that isoquerce- FXa and thrombin (Box 3, Figs 2,4). The goal of this
tin decreased levels of D-​dimer and soluble P-​selectin strategy is to partially inhibit these proteases to reduce
in patients with advanced cancer and no patients devel- thrombosis. However, these proteases are essential for
oped VTE (NCT02195232)168. Further phase III trials haemostasis and therefore bleeding is the primary side
are planned with isoquercetin in patients with cancer effect of current anticoagulants. Table 3 shows a list of
(J. Zwicker, personal communication). anticoagulant agents and their targets that have been
evaluated in clinical trials.
Other antiplatelet strategies
Several additional antiplatelet strategies are emerging. Inhibiting the TF–FVIIa complex
For example, targeting the active conformation of αIIbβ3 The TF–FVIIa complex plays an essential role in haemo-
with a single-​chain variable fragment has been shown to stasis but also contributes to most forms of thrombosis21.
inhibit thrombosis without an increase in bleeding time169. This complex can be inhibited by the endogenous inhib-
This has led to the concept that antithrombotic agents itor TFPI or by antibodies, pharmacological agents (such
can be delivered to developing thrombi, which in theory as active site-​inhibited FVIIa) and proteins expressed
should require a lower concentration of the antithrom- by blood-​sucking ticks (ixolaris) and worms (nematode
botic and therefore be associated with less bleeding. anticoagulant protein C2 (NAPc2))21. The antithrom-
Indeed, single-​chain variable fragments have been botic effect of these inhibitors has been tested in preclin-
coupled to a variety of antithrombotic agents, including ical and clinical studies. NAPc2 suppressed prothrombin
the FXa inhibitor tick anticoagulant peptide170, and all F1.2 and reduced ischaemia without increasing bleed-
conjugates have exhibited antithrombotic activity in pre- ing in patients with non-​ST-​segment elevation ACS
clinical studies without affecting haemostasis20. Another (NCT00116012)179. However, no further studies have
approach is to target intracellular interactions that are been performed. Active-​site inhibited FVIIa was eval-
required for αIIbβ3 activation. One study found that dis- uated in patients with acute lung injury/acute respira-
rupting the binding of the intracellular domain of αIIbβ3 tory distress syndrome but was stopped early because of
to Gα13 with a myristoylated peptide did not inhibit initial increased bleeding and mortality180.
platelet adhesion or primary aggregation, but inhibited
thrombosis in a mouse model171. Targeting the intrinsic coagulation pathway
Platelets contain an oxygenase called 12(S)- Components of the intrinsic pathway (FXII, FXI, FIX
lipoxygenase (12-​LOX) that converts arachidonic acid and FVIII) are attractive targets for the development of
released from phospholipids into bioactive metabolites anticoagulants because their inhibition is less likely to
(12(S)-hydroperoxyeicosatetraenoic acid and hydro­ cause bleeding compared with anticoagulants that tar-
eicosatetraenoic acid)172. 12-​LOX was found to regulate get the TF–FVIIa complex or common pathway181–184.
various platelet responses, including integrin activation However, there is a trade-​off between efficacy and safety.
and secretion, in response to stimulation with collagen Earlier trials focused on targeting FVIII or FIX but these
or thrombin173. In addition, 12-​LOX is required for agents were halted owing to a lack of efficacy and an
FcγRIIa-mediated platelet activation during immune- allergic reaction to the conjugate as well as for addi-
mediated thrombosis in patients with heparin-​induced tional unknown reasons. We have included a discussion

Nature Reviews | Drug Discovery


Reviews

Table 3 | clinical trials of anticoagulant agents


Name/company Agent/route of Target Disease clinical trial Trial iD comments
administration
TB-402/ThrombGenics Antibody/IV FVIII NA Phase I NCT00612196 Completed; safety study
TKA Phase II NCT00793234 Completed; reduced VTE
THA Phase II NCT01344954 Completed; no data
REG1/NHLBI/Regado RNA Aptamer/IV FIX NA Phase I NCT00113997 Completed; safety study
Biosciences
CAD Phase II NCT01848106 Terminated early; allergic reaction
ACS Phase III NCT00932100 Terminated early; allergic reaction
TTP889/vTv Therapeutics Small molecule/oral FIX Hip repair Phase II NCT00119457 Completed; no effect
LVAD Phase II NCT00909298 Terminated
Ionis-416858/Ionis ASO/SC FXI TKA Phase II NCT02553889 Ongoing
Pharmaceuticals
ESRD on Phase II NCT03358030 Ongoing
haemodialysis
BAY-1213790/Bayer Antibody/IV FXI TKA Phase II NCT03276143 Ongoing
ESRD on Phase II NCT03787368 Ongoing
haemodialysis
MAA868/Novartis Antibody/IV FXI AF Phase II NCT04213807 Ongoing
BMS-986177/BMS Small molecule/oral FXI Recurrent stroke Phase I NCT02608970 Completed; safety study
Phase II NCT03766581 Ongoing
TKA Phase II NCT03891524 Ongoing
AB023/Aronora Antibody/IV FXI ESRD on Phase I NCT03097341 Completed; safety study
haemodialysis
Phase II NCT03612856 Ongoing
CSL312/CSL Behring Antibody/IV FXII Hereditary Phase II NCT03712228 Ongoing
angioedema
ACS, acute coronary syndrome; AF, atrial fibrillation; ASO, antisense oligonucleotide; CAD, coronary artery disease; ESRD, end-​stage renal disease; F, factor;
IV, intravenous; LVAD, left ventricular assist device; NA, not available; NHLBI, National Heart, Lung, and Blood Institute; SC, subcutaneous; THA, total hip arthroplasty;
TKA, total knee arthroplasty; VTE, venous thromboembolism.

of FVIII and FIX inhibitors herein because this infor- in 261 patients who had undergone surgery for hip frac-
mation may provide insights into the future success or ture187, there was little difference in the primary efficacy
failure of targeting FXI or FXII. outcome of VTE between patient groups. There were
Because individuals with congenital deficiency of no major bleeding events and two clinically relevant
FXII do not bleed44, targeting FXII should be safe, but non-​major bleeding events with TTP889. Because of
efficacy may be reduced because TF–FVIIa-​dependent the lack of efficacy compared with placebo, develop-
and thrombin-​FXIa prothrombotic pathways will not be ment of TTP889 was halted. TTP889 was also admin-
inhibited. By contrast, targeting FXI will inhibit throm- istered to patients with left ventricular assist devices to
bin generation by blocking both FXIIa-​dependent and determine if it would reduce levels of thrombin activa-
thrombin-​dependent activation of FXI. tion markers in the plasma, but no results have been
reported (Table 3).
FVIII inhibitors. TB‐402 is a synthetic monoclonal SB249417 is a monoclonal antibody against FIX
antibody against FVIII that inhibits FVIII activity by that binds to the membrane-​binding carboxylation-​
80–90%. In a phase I study, TB-402 prolonged the aPTT carboxyglutamic (Gla) domain of human FIX. SB249417
in a dose-​dependent manner, and, with higher doses, attenuated thrombosis and infarct volume in rat mod-
the effect lasted up to 56 days185. Furthermore, in a els of arterial thrombosis and stroke, respectively188,189.
phase II study in patients undergoing knee replacement In addition, SB249417 demonstrated rapid and dose-
surgery186, a single postoperative intravenous TB‐402 dependent prolongation of the aPTT in cynomolgus
infusion resulted in a lower rate of VTE than with the monkeys and humans190,191 but was not carried forward
approved LMWH enoxaparin. The rates of major and into a phase I trial for unknown reasons.
clinically relevant non-​major bleeding with the two low- Pegnivacogin (REG1) is a FIXa-​directed inhibitory
est doses of TB-402 were the same as with enoxaparin. RNA aptamer. A strength of using an RNA aptamer
However, despite these promising results, development is that it can be rapidly inhibited by a complementary
of TB-402 was halted for unknown reasons. aptamer anivamersen (RB007). In phase I studies, intra-
venous REG1 prolonged the aPTT and activated clot-
FIX inhibitors. TTP889 is a small molecule oral inhib- ting time in a dose-​dependent manner, and these effects
itor of FIXa. In a study comparing a once daily dose of were rapidly reversed with RB007 (ref.192). In the phase II
TTP889 with placebo for extended thromboprophylaxis RADAR study, 640 patients with non-​ST-​s egment

www.nature.com/nrd
Reviews

Antisense oligonucleotides elevation ACS with planned early cardiac catheteriza- (NCT02608970). Currently, BMS-986177 is being
(ASOs). Antisense tion via femoral access were randomized to REG1 with compared with enoxaparin for thromboprophylaxis
oligonucleotides reduce RB007 reversal or to heparin193. At least 50% reversal after elective knee replacement surgery in a phase II
protein expression by binding of REG1 was needed to prevent bleeding after sheath clinical trial (NCT03891524). In addition, it is being
to mRNA and increasing its
degradation.
removal, indicating that targeting FIX is associated with compared with placebo in a phase II trial for second-
bleeding. Allergic reactions were noted in a small num- ary prevention in patients with stroke or high-​risk TIA
ber of patients. In the phase III REGULATE PCI trial, (NCT03766581). In the latter study, all patients receive
patients undergoing PCI for non-​ST-​segment elevation aspirin plus clopidogrel for 21 days followed by aspirin
ACS were randomized to REG1 with 80% RB007 rever- alone thereafter. Evidence of new stroke or intracra-
sal or to bivalirudin194. The study was stopped early after nial bleeding on repeat brain imaging are the primary
partial enrolment owing to severe allergic reactions194,195. efficacy and safety outcomes, respectively.
The rate of the primary efficacy end point, a composite of AB023 (xisomab) is a humanized version of 14E11,
all-​cause death, MI, stroke and unplanned target lesion an antibody that binds to the apple 2 domain of FXI
re-​vascularization, was 7% and 6% with REG1 and biv- and inhibits FXIIa-​mediated FXI activation but not
alirudin, respectively. Major bleeding occurred in <1% of thrombin-​mediated FXI activation203. AB023 has no
patients in both groups. The allergic reactions with REG1 effect on FXIa activity. Therefore, even though it binds
have been attributed to the polyethylene glycol conjugate FXI and not FXII, AB023 serves as an inhibitor of
used to prolong the half-​life of the aptamer. However, FXIIa-​dependent activation of coagulation. In a phase I
this adverse reaction halted the development of REG1. study, AB023 prolonged the aPTT in a dose-​dependent
manner204. AB023 exhibits a short half-​life with low doses,
FXI inhibitors. The most exciting new target for anticoag- which likely reflects rapid binding of AB023 to plasma
ulant therapy is FXI. There are several different develop- FXI. However, once FXI is saturated with high doses of
ment programmes that involve antisense oligonucleotides AB023, the half-​life of the free antibody increases, likely
(ASOs), monoclonal antibodies, aptamers and small reflecting its slower clearance (NCT03097341)204. A
molecules196,197 (Table  3). Small molecule inhibitors phase II trial is currently evaluating the safety and effi-
can be delivered orally or parenterally, whereas ASOs, cacy of AB023 compared with heparin in patients with
antibodies and aptamers require parenteral adminis- ESRD while on haemodialysis (NCT03612856). The level
tration. ASOs reduce protein expression indirectly by of clotting in the circuit and plasma will be assessed.
binding to target mRNA and causing its degradation. A It is notable that three trials are analysing the effect
disadvantage of this approach is that it takes 3 to 4 weeks of FXIa inhibitors on thrombosis in patients with ESRD
of ASO treatment to lower FXI levels into the therapeu- on haemodialysis. However, TF has been shown to
tic range. By contrast, FXI can be inhibited rapidly by contribute to haemodialysis-​induced thrombosis, rais-
antibodies, aptamers and small molecules. ing concerns about the efficacy of inhibiting FXI205.
A particularly exciting agent in development is IONIS- Nevertheless, we are eagerly awaiting results from these
416858, an ASO directed against FXI198. A phase II trial trials to see the efficacy and safety of targeting FXIa.
evaluated two subcutaneous doses of IONIS-416858 for
postoperative thromboprophylaxis in patients undergo- FXII inhibitors. The most advanced inhibitor of FXII
ing elective knee replacement surgery199, with the higher is 3F7, a fully humanized monoclonal antibody. 3F7
dose demonstrating superiority to enoxaparin in terms attenuated thrombosis to the same extent as heparin in
of VTE with no increase in bleeding. Two phase II stud- a rabbit ECMO model62. CSL312 is an affinity matured
ies are currently assessing the safety and tolerability of variant of 3F7. Notably, no thrombosis trials have been
IONIS-416858 in patients with ESRD on haemodialysis reported for CSL312. Instead, CSL312 is being evaluated
(NCT02553889 and NCT03358030). in a phase II trial in patients with hereditary angioedema
Osocimab (BAY-1213790) is the most advanced anti- (NCT03712228) because FXIIa also leads to the genera-
​FXIa antibody200. In a phase II trial called FOXTROT, tion of bradykinin and increased vascular permeability in
BAY-1213790 was compared with enoxaparin for these patients. Importantly, patients with a point mutation
throm­b oprophylaxis in patients undergoing knee in FXII that leads to increased autoactivation of FXII pres-
arthroplasty (NCT03276143). The study showed that ent with hereditary angioedema type III owing to exces-
preoperative BAY-1213790 was superior to enoxaparin sive activation of the bradykinin-​forming kallikrein–kinin
for reducing asymptomatic DVT201. In addition, a small pathway but not with thrombosis206. Indeed, the role of
phase II study is assessing the safety and tolerability of FXII in thrombosis in humans is still unclear44.
BAY-1213790 in patients with ESRD on haemodialysis
(NCT0378368). Other anticoagulant strategies
MAA868 is an antibody that binds to both FXI and An emerging approach to inhibit the activation of FXII
FXIa202. It reduced thrombosis in a mouse ferric chloride is to block its activators. Nucleic acid scavengers and
carotid artery model and prolonged the aPTT in cyno- polyphosphate inhibitors have been shown to reduce
molgus monkeys. In addition, a phase I trial reported that thrombosis in mouse models207,208. Studies have shown
a single subcutaneous dose of MAA868 prolonged the that administration of DNase I, which degrades NETs
aPTT for 4 weeks202. MAA868 is currently being evaluated and cell-​f ree DNA, reduces venous thrombosis in
in a phase II trial in patients with AF (NCT04213807). tumour-​bearing and non-​tumour-​bearing mice209–212.
BMS-986177 is a small molecule oral inhibitor Pulmozyme® is a commercial DNase used as an inhaled
of FXIa200 that was well tolerated in a phase I study therapy to remove extracellular DNA from the lungs

Nature Reviews | Drug Discovery


Reviews

of patients with cystic fibrosis. One study evaluated different patients, leading to thrombosis at a variety of
the effects of intravenous followed by subcutaneous sites. Thrombosis may be triggered by the activation
dosing of recombinant DNase I in patients with lupus of platelets, of the coagulation system and/or of the
nephritis213. DNase I was well tolerated but did not endothelium. For instance, patients with myeloprolifer-
reduce serum markers of disease. To date, these agents ative neoplasia often have splanchnic vein thrombosis215.
have not been evaluated in thrombosis trials. In patients with CVD, the primary trigger is rupture or
erosion of atherosclerotic plaques, whereas the triggers
Challenges developing antithrombotic agents in patients with PAD are not known. In terms of VTE,
One of the major challenges in the development of triggers range from artificial surfaces to inflammation
new antithrombotic drugs is that we already have a induced by surgery, which likely induces TF expression
rich armamen­tarium of antiplatelet and anticoagulant on monocytes10,52. This means that, in contrast to VKAs
agents (Fig. 2, Boxes 1,3). Therefore, new agents must and inhibitors of the common pathway, a new anticoag-
demonstrate either superior efficacy and safety or ulant, such as a FXII antagonist, may be effective for one
equivalent efficacy and improved safety compared with indication but not for others.
existing agents.
Another challenge is the expense of the large clini- Outlook
cal trials required to assess new agents, particularly with Thrombosis is the leading cause of death in the world.
antiplatelet agents. For instance, the PAR1 antagonist Although major progress has been made in developing
vorapaxar was assessed in the TRACER trial, which effective antiplatelet agents (such as P2Y12 antagonists
involved 12,944 patients with ACS, and in the TRA for AT) and anticoagulants (such as the DOACs for
2P–TIMI 50 trial, which involved 26,449 patients with a VT), incomplete protection and bleeding complications
history of MI, ischaemic stroke or PAD95,96. However, the associated with the use of currently available drugs call
TRA 2P–TIMI 50 trial included a rather diverse group of for the development of novel therapeutic approaches
patients who may have had different underlying diseases. and the identification of new targets.
There are similar challenges with VTE trials. Guidelines Notably, the PROSPECT study showed that the
recommending VKAs for stroke prevention in AF were cumulative rate of MACE in patients with ACS was
based on randomized trials comparing them with aspirin 20.4% at 3 years216. In addition, three major trials (CURE,
or no treatment in less than 3,000 patients. By contrast, TRITON and PLATO) have demonstrated that, irrespec-
guidelines giving preference to DOACs over VKAs are tive of the combination of drugs, about 10% of patients
based on data in over 70,000 patients, and the recom- with ACS treated with DAPT are not protected94,217,218.
mendation to use the combination of low-​dose rivarox- Furthermore, the risk of recurrence after stopping anti-
aban plus aspirin over aspirin alone in selected patients coagulants in patients with unprovoked VTE is about
with CAD or PAD is based on a trial that enrolled over 10% at 1 year. Recent studies have shown that, compared
27,000 patients100. Therefore, head-​to-​head studies com- with placebo, aspirin significantly reduces recurrent
paring new anticoagulants with DOACs will require large VTE219–221, which suggested a role for platelets in VTE.
numbers of patients. To avoid this, niche indications need However, a subsequent study showed that the FXa inhib-
to be identified such that the new agents can be compared itor rivaroxaban exhibited superior efficacy to aspirin in
with placebo or standard-​of-​care agents that are known preventing recurrent VTE for extended thromboprophy-
to be problematic. Examples of such indications include laxis and reduced the rate of recurrent VTE222. Similar
the use of FXII inhibitors to prevent central venous cath- to DAPT, the optimal duration of anticoagulation after
eter thrombosis in patients with cancer, an indication VTE is unclear.
where LMWH and VKAs have failed; the prevention of Improved strategies to prevent recurrent ischaemic
recurrent stroke in patients with acute ischaemic stroke, stroke (Box 2), where the current treatment is an anti-
an indication where heparin, LMWH and heparinoids platelet agent, are also needed. Ongoing trials are eval-
have failed; the prevention of stroke in patients with AF uating the effect of other available antithrombotics as
undergoing haemodialysis, an indication where there well as a variant of APC together with tPA on the treat-
is equipoise as to whether the benefits of VKAs out- ment of acute ischaemic stroke. Trials are also being
weigh their harms214; and the prevention or treatment of conducted to determine if PAR4 and FXI inhibitors —
VTE in patients with active cancer, an indication where novel antithrombotic agents — can reduce recurrent
DOACs are superior to placebo or LMWH, respectively, stroke. FXI antagonists are further being evaluated in
but cause more bleeding. Therefore, thoughtful selec- preventing thrombosis in patients with ESRD on hae-
tion of indications and creative trial designs are needed modialysis, a complication where the intrinsic pathway
to gain approval for the next generation of anticoagu- of coagulation has a well-​documented role.
lants. However, the selected patient population needs A more personalized therapeutic approach to throm-
to be large enough for the pharmaceutical companies to bosis prevention would be beneficial. Indeed, one trial
make a profit on their investment. Of note, it would be found that genotyping patients for CYP2C19 genetic
interesting to determine if a FXIIa antagonist can reduce variation improved antiplatelet therapy with P2Y12
thrombosis associated with a mechanical heart valve. antagonists. The development of similar genetic and
Another consideration is determining in which spe- functional tests for predicting or measuring a patient’s
cific indication the new agent may be most effective. response to antiplatelet or anticoagulant therapy will be
Thrombosis has been divided into AT and VT. However, critical for providing the best possible treatment to indi-
there are numerous different triggers of thrombosis in viduals. New challenges includes the global epidemics of

www.nature.com/nrd
Reviews

obesity, metabolic syndrome and diabetes. For instance, possibility of them exhibiting reduced antithrombotic
patients with type 2 diabetes exhibit increased platelet potency must be considered.
reactivity and are less responsive to antiplatelet therapy74. Despite these challenges and considerations, the new
Major challenges in the establishment of novel thera- generation of antiplatelet and anticoagulant drugs holds
pies include the demonstration of non-​inferiority of new great promise and the results of ongoing clinical trials
agents compared with approved agents and the high cost are eagerly awaited.
associated with such large trials. However, while these
new agents are expected to cause less bleeding, the Published online xx xx xxxx

1. Wendelboe, A. M. & Raskob, G. E. Global burden of thrombosis and cardiovascular disease. Thromb. 45. Puy, C., Rigg, R. A. & McCarty, O. J. The hemostatic
thrombosis: epidemiologic aspects. Circ. Res. 118, Haemost. 117, 1265–1271 (2017). role of factor XI. Thromb. Res. 141, S8–S11 (2016).
1340–1347 (2016). The paper describes the coagulation protease 46. Lillicrap, D. The molecular pathology of hemophilia A.
2. Prandoni, P. Venous and arterial thrombosis: two cascade and its role in thrombosis, thrombo- Transfus. Med. Rev. 5, 196–206 (1991).
aspects of the same disease? Eur. J. Intern. Med. 20, inflammation and atherosclerosis. 47. Tjarnlund-Wolf, A. & Lassila, R. Phenotypic
660–661 (2009). 24. Broos, K., Feys, H. B., De Meyer, S. F., Vanhoorelbeke, K. characterization of haemophilia B - Understanding
3. Prandoni, P. et al. Venous thromboembolism and the & Deckmyn, H. Platelets at work in primary hemostasis. the underlying biology of coagulation factor IX.
risk of subsequent symptomatic atherosclerosis. Blood Rev. 25, 155–167 (2011). Haemophilia 25, 567–574 (2019).
J. Thromb. Haemost. 4, 1891–1896 (2006). 25. Stalker, T. J., Welsh, J. D. & Brass, L. F. Shaping the 48. Bolton-Maggs, P. H. & Pasi, K. J. Haemophilias A and B.
4. Reich, L. M. et al. Prospective study of subclinical platelet response to vascular injury. Curr. Opin. Hematol. Lancet 361, 1801–1809 (2003).
atherosclerosis as a risk factor for venous 21, 410–417 (2014). 49. Mann, K. G. Thrombin formation. Chest 124 (Suppl. 3),
thromboembolism. J. Thromb. Haemost. 4, 26. Stegner, D. & Nieswandt, B. Platelet receptor signaling 4–10 (2003).
1909–1913 (2006). in thrombus formation. J. Mol. Med. 89, 109–121 50. Loskutoff, D. J. & Curriden, S. A. The fibrinolytic
5. Braekkan, S. K. et al. Family history of myocardial (2011). system of the vessel wall and its role in the control of
infarction is an independent risk factor for venous 27. Bergmeier, W. & Stefanini, L. Platelets at the vascular thrombosis. Ann. N. Y. Acad. Sci. 598, 238–247
thromboembolism: the Tromso study. J. Thromb. interface. Res. Pract. Thromb. Haemost. 2, 27–33 (1990).
Haemost. 6, 1851–1857 (2008). (2018). 51. Wiman, B. & Collen, D. On the mechanism of the
6. Bova, C. et al. Incidence of arterial cardiovascular 28. Esmon, C. T. Regulation of blood coagulation. Biochim. reaction between human alpha 2-antiplasmin and
events in patients with idiopathic venous Biophys. Acta 1477, 349–360 (2000). plasmin. J. Biol. Chem. 254, 9291–9297 (1979).
thromboembolism. A retrospective cohort study. 29. Esmon, C. T. The endothelial cell protein C receptor. 52. Johnson, G. J., Leis, L. A. & Bach, R. R. Tissue factor
Thromb. Haemost. 96, 132–136 (2006). Thromb. Haemost. 83, 639–643 (2000). activity of blood mononuclear cells is increased after
7. Mackman, N. Triggers, targets and treatments for 30. Mackman, N. Tissue-specific hemostasis in mice. total knee arthroplasty. Thromb. Haemost. 102,
thrombosis. Nature 451, 914–918 (2008). Arterioscler. Thromb. Vasc. Biol. 25, 2273–2281 728–734 (2009).
The paper describes the mechanisms of arterial and (2005). 53. Grover, S. P. & Mackman, N. Intrinsic pathway of
venous thrombosis and targets for antithrombotics. 31. Rosenberg, R. D. & Aird, W. C. Vascular-bed-specific coagulation and thrombosis. Arterioscler. Thromb.
8. Previtali, E., Bucciarelli, P., Passamonti, S. M. hemostasis and hypercoagulable states. N. Engl. Vasc. Biol. 39, 331–338 (2019).
& Martinelli, I. Risk factors for venous and arterial J. Med. 340, 1555–1564 (1999). 54. Thalin, C., Hisada, Y., Lundstrom, S., Mackman, N.
thrombosis. Blood Transfus. 9, 120–138 (2011). 32. Stefanini, L. & Bergmeier, W. Negative regulators of & Wallen, H. Neutrophil extracellular traps: villains
9. Owens, A. P. III & Mackman, N. Role of tissue factor platelet activation and adhesion. J. Thromb. Haemost. and targets in arterial, venous, and cancer-associated
in atherothrombosis. Curr. Atheroscler. Rep. 14, 16, 220–230 (2018). thrombosis. Arterioscler. Thromb. Vasc. Biol. 39,
394–401 (2012). 33. Robson, S. C. et al. Ectonucleotidases of CD39 family 1724–1738 (2019).
10. Wolberg, A. S. et al. Venous thrombosis. Nat. Rev. modulate vascular inflammation and thrombosis in 55. Martinod, K. & Wagner, D. D. Thrombosis: tangled up
Dis. Prim. 1, 15006 (2015). transplantation. Semin. Thromb. Hemost. 31, 217–233 in NETs. Blood 123, 2768–2776 (2014).
11. Khan, A. A. & Lip, G. Y. H. The prothrombotic state in (2005). 56. Martinod, K. et al. Neutrophil histone modification by
atrial fibrillation: pathophysiological and management 34. Wood, J. P., Ellery, P. E., Maroney, S. A. & Mast, A. E. peptidylarginine deiminase 4 is critical for deep vein
implications. Cardiovasc. Res. 115, 31–45 (2019). Biology of tissue factor pathway inhibitor. Blood 123, thrombosis in mice. Proc. Natl Acad. Sci. USA 110,
12. Watson, T., Shantsila, E. & Lip, G. Y. Mechanisms of 2934–2943 (2014). 8674–8679 (2013).
thrombogenesis in atrial fibrillation: Virchow’s triad 35. Rosenberg, R. D. & Rosenberg, J. S. Natural 57. Kokoye, Y. et al. A comparison of the effects of factor XII
revisited. Lancet 373, 155–166 (2009). anticoagulant mechanisms. J. Clin. Invest. 74, 1–6 deficiency and prekallikrein deficiency on thrombus
13. Medcalf, R. L. Fibrinolysis: from blood to the brain. (1984). formation. Thromb. Res. 140, 118–124 (2016).
J. Thromb. Haemost. 15, 2089–2098 (2017). Overview of how anticoagulant mechanisms 58. Kuijpers, M. J. et al. Factor XII regulates the
14. Lin, L. et al. Clinical and safety outcomes of oral balance procoagulants in different vascular beds. pathological process of thrombus formation on
antithrombotics for stroke prevention in atrial 36. Bryckaert, M., Rosa, J. P., Denis, C. V. & Lenting, P. J. ruptured plaques. Arterioscler. Thromb. Vasc. Biol. 34,
fibrillation: a systematic review and network Of von Willebrand factor and platelets. Cell Mol. 1674–1680 (2014).
meta-analysis. J. Am. Med. Dir. Assoc. 16, 1103.e1–19 Life Sci. 72, 307–326 (2015). 59. van Montfoort, M. L. et al. Factor XI regulates
(2015). 37. Nieswandt, B. & Watson, S. P. Platelet-collagen pathological thrombus formation on acutely ruptured
15. Doktorova, M. & Motovska, Z. Clinical review: interaction: is GPVI the central receptor? Blood 102, atherosclerotic plaques. Arterioscler. Thromb. Vasc.
bleeding–a notable complication of treatment in 449–461 (2003). Biol. 34, 1668–1673 (2014).
patients with acute coronary syndromes: incidence, The paper describes the role of platelet GPVI in 60. Mackman, N. New targets for atherothrombosis.
predictors, classification, impact on prognosis, and haemostasis and arterial thrombosis. Arterioscler. Thromb. Vasc. Biol. 34, 1607–1608
management. Crit. Care 17, 239 (2013). 38. Stefanini, L. & Bergmeier, W. RAP GTPases and (2014).
16. Marzec, L. N. et al. Influence of direct oral platelet integrin signaling. Platelets 30, 41–47 61. Yau, J. W. et al. Selective depletion of factor XI or
anticoagulants on rates of oral anticoagulation for (2019). factor XII with antisense oligonucleotides attenuates
atrial fibrillation. J. Am. Coll. Cardiol. 69, 2475–2484 39. Shattil, S. J. & Newman, P. J. Integrins: dynamic catheter thrombosis in rabbits. Blood 123, 2102–2107
(2017). scaffolds for adhesion and signaling in platelets. Blood (2014).
17. Plow, E. F., Wang, Y. & Simon, D. I. The search for new 104, 1606–1615 (2004). The paper shows that knockdown of either FXII
antithrombotic mechanisms and therapies that may 40. Huang, Y. et al. Arf6 controls platelet spreading and or FXI but not FVII prolonged the time to catheter
spare hemostasis. Blood 131, 1899–1902 (2018). clot retraction via integrin αIIbβ3 trafficking. Blood 127, occlusion in a rabbit model.
18. Grover, S. P., Bergmeier, W. & Mackman, N. Platelet 1459–1467 (2016). 62. Larsson, M. et al. A factor XIIa inhibitory antibody
signaling pathways and new inhibitors. Arterioscler. 41. Drake, T. A., Morrissey, J. H. & Edgington, T. S. provides thromboprotection in extracorporeal circulation
Thromb. Vasc. Biol. 38, e28–e35 (2018). Selective cellular expression of tissue factor in without increasing bleeding risk. Sci. Transl. Med. 6,
19. Fredenburgh, J. C., Gross, P. L. & Weitz, J. I. Emerging human tissues. Implications for disorders of 222ra217 (2014).
anticoagulant strategies. Blood 129, 147–154 (2017). hemostasis and thrombosis. Am. J. Pathol. 134, The paper shows that a FXII antibody inhibited
20. McFadyen, J. D., Schaff, M. & Peter, K. Current and 1087–1097 (1989). thrombosis in an extracorporeal circuit as
future antiplatelet therapies: emphasis on preserving 42. Hoffman, M. et al. Tissue factor around dermal effectively as heparin without increasing bleeding
haemostasis. Nat. Rev. Cardiol. 15, 181–191 (2018). vessels has bound factor VII in the absence of injury. in a rabbit model.
The paper summarizes antiplatelet trials in J. Thromb. Haemost. 5, 1403–1408 (2007). 63. Vilahur, G., Ben-Aicha, S. & Badimon, L. New insights
patients with cardiovascular disease and new 43. Hoffman, M. & Monroe, D. M. Tissue factor in brain is into the role of adipose tissue in thrombosis. Cardiovasc.
antiplatelet agents. not saturated with factor VIIa: implications for factor Res. 113, 1046–1054 (2017).
21. Grover, S. P. & Mackman, N. Tissue factor: an essential VIIa dosing in intracerebral hemorrhage. Stroke 40, 64. Dellas, C. & Loskutoff, D. J. Historical analysis of PAI-1
mediator of hemostasis and trigger of thrombosis. 2882–2884 (2009). from its discovery to its potential role in cell motility
Arterioscler. Thromb. Vasc. Biol. 38, 709–725 (2018). 44. Gailani, D. & Renne, T. The intrinsic pathway of and disease. Thromb. Haemost. 93, 631–640 (2005).
22. Gremmel, T., Frelinger, A. L. III & Michelson, A. D. coagulation: a target for treating thromboembolic 65. Huang, G., Wang, P., Li, T. & Deng, X. Genetic association
Platelet physiology. Semin. Thromb. Hemost. 42, disease? J. Thromb. Haemost. 5, 1106–1112 (2007). between plasminogen activator inhibitor-1 rs1799889
191–204 (2016). The paper provides an overview of the intrinsic polymorphism and venous thromboembolism: evidence
23. Ten Cate, H., Hackeng, T. M. & Garcia de Frutos, P. pathway of coagulation and the potential for FXI from a comprehensive meta-analysis. Clin. Cardiol. 42,
Coagulation factor and protease pathways in and FXII as targets for new anticoagulants. 1232–1238 (2019).

Nature Reviews | Drug Discovery


Reviews

66. Patrono, C. et al. Antiplatelet agents for the treatment 88. Al-Samkari, H. & Connors, J. M. The role of direct oral neuroprotection. Blood 132, 159–169 (2018).
and prevention of coronary atherothrombosis. J. Am. anticoagulants in treatment of cancer-associated The paper describes the beneficial effects of an
Coll. Cardiol. 70, 1760–1776 (2017). thrombosis. Cancers 10, E271 (2018). activated protein C variant in stroke.
67. Serebruany, V. L., Malinin, A. I., Eisert, R. M. 89. Levi, M. Management of bleeding in patients treated 109. Griffin, J. H., Mosnier, L. O., Fernandez, J. A. &
& Sane, D. C. Risk of bleeding complications with with direct oral anticoagulants. Crit. Care 20, 249 Zlokovic, B. V. 2016 Scientific Sessions Sol Sherry
antiplatelet agents: meta-analysis of 338,191 (2016). distinguished lecturer in thrombosis: thrombotic
patients enrolled in 50 randomized controlled trials. 90. Mavrakanas, T. A., Samer, C. F., Nessim, S. J., Frisch, G. stroke: neuroprotective therapy by recombinant-
Am. J. Hematol. 75, 40–47 (2004). & Lipman, M. L. Apixaban pharmacokinetics at steady activated protein C. Arterioscler. Thromb. Vasc. Biol.
68. Raimondi, P., Hylek, E. M. & Aronis, K. N. Reversal state in hemodialysis patients. J. Am. Soc. Nephrol. 28, 36, 2143–2151 (2016).
agents for oral antiplatelet and anticoagulant 2241–2248 (2017). 110. Mosnier, L. O., Zlokovic, B. V. & Griffin, J. H.
treatment during bleeding events: current strategies. 91. Eikelboom, J. W. et al. Dabigatran versus warfarin in Cytoprotective-selective activated protein C therapy for
Curr. Pharm. Des. 23, 1406–1423 (2017). patients with mechanical heart valves. N. Engl. J. Med. ischaemic stroke. Thromb. Haemost. 112, 883–892
69. Nagalla, S. & Sarode, R. Role of platelet transfusion in 369, 1206–1214 (2013). (2014).
the reversal of anti-platelet therapy. Transfus. Med. Rev. The trial comparing dabigatran with warfarin in 111. Lyden, P. et al. Randomized, controlled, dose
33, 92–97 (2019). patients with mechanical heart valves was stopped escalation trial of a protease-activated receptor-1
70. Bhatt, D. L. et al. Antibody-based ticagrelor reversal early owing to increased rates of thromboembolic agonist in acute ischemic stroke: Final results of the
agent in healthy volunteers. N. Engl. J. Med. 380, and bleeding complications with dabigatran. RHAPSODY trial: a multi-center, phase 2 trial using a
1825–1833 (2019). 92. Pengo, V. et al. Rivaroxaban vs warfarin in high-risk continual reassessment method to determine the
71. Taylor, G., Osinski, D., Thevenin, A. & Devys, J. M. patients with antiphospholipid syndrome. Blood 132, safety and tolerability of 3K3A-APC, a recombinant
Is platelet transfusion efficient to restore platelet 1365–1371 (2018). variant of human activated protein C, in combination
reactivity in patients who are responders to aspirin The trial showed that the use of rivaroxaban in with tissue plasminogen activator, mechanical
and/or clopidogrel before emergency surgery? high-risk patients with antiphospholipid syndrome thrombectomy or both in moderate to severe acute
J. Trauma. Acute Care Surg. 74, 1367–1369 (2013). was associated with increased rates of thrombosis ischemic stroke. Ann. Neurol. 85, 125–136 (2019).
72. Baharoglu, M. I. et al. Platelet transfusion versus compared with warfarin. 112. Sharifi, M. et al. Moderate pulmonary embolism
standard care after acute stroke due to spontaneous 93. Leentjens, J., Peters, M., Esselink, A. C., Smulders, Y. treated with thrombolysis (from the “MOPETT” Trial).
cerebral haemorrhage associated with antiplatelet & Kramers, C. Initial anticoagulation in patients with Am. J. Cardiol. 111, 273–277 (2013).
therapy (PATCH): a randomised, open-label, phase 3 pulmonary embolism: thrombolysis, unfractionated 113. Sharifi, M. et al. Retrospective comparison of
trial. Lancet 387, 2605–2613 (2016). heparin, LMWH, fondaparinux, or DOACs? Br. J. Clin. ultrasound facilitated catheter-directed thrombolysis
73. O’Donoghue, M. & Wiviott, S. D. Clopidogrel Pharmacol. 83, 2356–2366 (2017). and systemically administered half-dose thrombolysis
response variability and future therapies: clopidogrel: 94. Wiviott, S. D. et al. Prasugrel versus clopidogrel in in treatment of pulmonary embolism. Vasc. Med. 24,
does one size fit all? Circulation 114, e600–e606 patients with acute coronary syndromes. N. Engl. 103–109 (2019).
(2006). J. Med. 357, 2001–2015 (2007). 114. Piazza, G. et al. A prospective, single-arm, multicenter
74. Angiolillo, D. J. et al. Insulin therapy is associated with 95. Morrow, D. A. et al. Vorapaxar in the secondary trial of ultrasound-facilitated, catheter-directed, low-dose
platelet dysfunction in patients with type 2 diabetes prevention of atherothrombotic events. N. Engl. J. Med. fibrinolysis for acute massive and submassive pulmonary
mellitus on dual oral antiplatelet treatment. J. Am. Coll. 366, 1404–1413 (2012). embolism: the SEATTLE II study. JACC Cardiovasc.
Cardiol. 48, 298–304 (2006). 96. Tricoci, P. et al. Thrombin-receptor antagonist vorapaxar Interv. 8, 1382–1392 (2015).
75. Dayoub, E. J. et al. Trends in platelet adenosine in acute coronary syndromes. N. Engl. J. Med. 366, 115. Nakanishi-Matsui, M. et al. PAR3 is a cofactor for
diphosphate P2Y12 receptor inhibitor use and 20–33 (2012). PAR4 activation by thrombin. Nature 404, 609–613
adherence among antiplatelet-naive patients after 97. Rothberg, M. B., Celestin, C., Fiore, L. D., Lawler, E. (2000).
percutaneous coronary intervention, 2008–2016. & Cook, J. R. Warfarin plus aspirin after myocardial 116. Nieman, M. T. Protease-activated receptors in
JAMA Intern. Med. 178, 943–950 (2018). infarction or the acute coronary syndrome: meta-analysis hemostasis. Blood 128, 169–177 (2016).
76. Paniccia, R., Priora, R., Liotta, A. A. & Abbate, R. with estimates of risk and benefit. Ann. Intern. Med. 117. Coughlin, S. R. Thrombin signalling and
Platelet function tests: a comparative review. Vasc. 143, 241–250 (2005). protease-activated receptors. Nature 407, 258–264
Health Risk Manag. 11, 133–148 (2015). 98. Alexander, J. H. et al. Apixaban with antiplatelet (2000).
77. Aradi, D. et al. Efficacy and safety of intensified therapy after acute coronary syndrome. N. Engl. J. Med. 118. Covic, L., Misra, M., Badar, J., Singh, C. & Kuliopulos, A.
antiplatelet therapy on the basis of platelet reactivity 365, 699–708 (2011). Pepducin-based intervention of thrombin-receptor
testing in patients after percutaneous coronary 99. Mega, J. L. et al. Rivaroxaban in patients with a recent signaling and systemic platelet activation. Nat. Med. 8,
intervention: systematic review and meta-analysis. acute coronary syndrome. N. Engl. J. Med. 366, 9–19 1161–1165 (2002).
Int. J. Cardiol. 167, 2140–2148 (2013). (2012). 119. Kuliopulos, A. & Covic, L. Blocking receptors on the
78. Hulot, J. S. et al. Cytochrome P450 2C19 loss-of- This trial shows that low-dose rivaroxaban reduced inside: pepducin-based intervention of PAR signaling
function polymorphism is a major determinant cardiovascular events in patients with a recent and thrombosis. Life Sci. 74, 255–262 (2003).
of clopidogrel responsiveness in healthy subjects. acute cardiovascular event treated with antiplatelet 120. Gurbel, P. A. et al. Cell-penetrating pepducin therapy
Blood 108, 2244–2247 (2006). agents. targeting PAR1 in subjects with coronary artery
79. Claassens, D. M. F. et al. A genotype-guided strategy 100. Eikelboom, J. W. et al. Rivaroxaban with or without disease. Arterioscler. Thromb. Vasc. Biol. 36,
for oral P2Y12 inhibitors in primary PCI. N. Engl. aspirin in stable cardiovascular disease. N. Engl. J. Med. 189–197 (2016).
J. Med. 381, 1621–1631 (2019). 377, 1319–1330 (2017). 121. Aisiku, O. et al. Parmodulins inhibit thrombus
The paper shows a benefit of using a CYP2C19 The trial shows that the combination of low-dose formation without inducing endothelial injury caused
genotype-guided strategy for the selection of P2Y12 rivaroxaban and aspirin reduced cardiovascular by vorapaxar. Blood 125, 1976–1985 (2015).
inhibitor therapy. events in patients with stable atherosclerotic 122. De Ceunynck, K. et al. PAR1 agonists stimulate
80. McFadden, E. P. et al. Late thrombosis in drug-eluting vascular disease compared with aspirin alone. APC-like endothelial cytoprotection and confer
coronary stents after discontinuation of antiplatelet 101. Connolly, S. J. et al. Rivaroxaban with or without resistance to thromboinflammatory injury. Proc. Natl
therapy. Lancet 364, 1519–1521 (2004). aspirin in patients with stable coronary artery Acad. Sci. USA 115, E982–E991 (2018).
81. Levine, G. N. et al. 2016 ACC/AHA guideline focused disease: an international, randomised, double-blind, 123. Wong, P. C. et al. Blockade of protease-activated
update on duration of dual antiplatelet therapy in placebo-controlled trial. Lancet 391, 205–218 (2018). receptor-4 (PAR4) provides robust antithrombotic
patients with coronary artery disease: a report of the 102. Anand, S. S. et al. Rivaroxaban with or without aspirin activity with low bleeding. Sci. Transl. Med. 9,
American College of Cardiology/American Heart in patients with stable peripheral or carotid artery eaaf5294 (2017).
Association task force on clinical practice guidelines. disease: an international, randomised, double-blind, Strong data in animal models supporting PAR4 as
J. Am. Coll. Cardiol. 68, 1082–1115 (2016). placebo-controlled trial. Lancet 391, 219–229 (2018). an antiplatelet target.
82. Collet, J. P. et al. Case-based implementation of the 103. Lopes, R. D. et al. Safety and efficacy of antithrombotic 124. Wilson, S. J. et al. PAR4 (Protease-Activated Receptor 4)
2017 ESC focused update on dual antiplatelet therapy strategies in patients with atrial fibrillation undergoing antagonism with BMS-986120 inhibits human ex vivo
in coronary artery disease. Eur. Heart J. 39, e1–e33 percutaneous coronary intervention: a network thrombus formation. Arterioscler. Thromb. Vasc. Biol.
(2018). meta-analysis of randomized controlled trials. JAMA 38, 448–456 (2018).
83. Watanabe, H. et al. Very short dual antiplatelet therapy Cardiol. 4, 747–755 (2019). 125. French, S. L. et al. A function-blocking PAR4 antibody is
after drug-eluting stent implantation in patients with 104. Yasuda, S. et al. Antithrombotic therapy for atrial markedly antithrombotic in the face of a hyperreactive
high bleeding risk: insight from the STOPDAPT-2 trial. fibrillation with stable coronary disease. N. Engl. J. Med. PAR4 variant. Blood Adv. 2, 1283–1293 (2018).
Circulation 140, 1957–1959 (2019). 381, 1103–1113 (2019). 126. Martins Lima, A., Martins Cavaco, A. C.,
84. Kuruvilla, M. & Gurk-Turner, C. A review of warfarin 105. Powers, W. J. et al. 2018 guidelines for the early Fraga-Silva, R. A., Eble, J. A. & Stergiopulos, N. From
dosing and monitoring. Proceedings 14, 305–306 management of patients with acute ischemic stroke: patients to platelets and back again: pharmacological
(2001). a guideline for healthcare professionals from the approaches to glycoprotein VI, a thrilling antithrombotic
85. Zhu, Y. et al. Estimation of warfarin maintenance dose American Heart Association/American Stroke target with minor bleeding risks. Thromb. Haemost.
based on VKORC1 (-1639 G>A) and CYP2C9 Association. Stroke 49, e46–e110 (2018). 119, 1720–1739 (2019).
genotypes. Clin. Chem. 53, 1199–1205 (2007). 106. Gurwitz, J. H. et al. Risk for intracranial hemorrhage 127. Rayes, J., Watson, S. P. & Nieswandt, B. Functional
86. Weitz, J. I., Jaffer, I. H. & Fredenburgh, J. C. Recent after tissue plasminogen activator treatment for acute significance of the platelet immune receptors GPVI and
advances in the treatment of venous thromboembolism myocardial infarction. Participants in the National CLEC-2. J. Clin. Invest. 129, 12–23 (2019).
in the era of the direct oral anticoagulants. F1000Res Registry of Myocardial Infarction 2. Ann. Intern. Med. 128. Mammadova-Bach, E. et al. Platelet glycoprotein VI
6, 985 (2017). 129, 597–604 (1998). binds to polymerized fibrin and promotes thrombin
The paper describes the benefits of direct oral 107. Chatterjee, S. et al. Thrombolysis for pulmonary generation. Blood 126, 683–691 (2015).
anticoagulants over vitamin K antagonists for the embolism and risk of all-cause mortality, major bleeding, 129. Alshehri, O. M. et al. Fibrin activates GPVI in human
treatment of venous thromboembolism. and intracranial hemorrhage: a meta-analysis. JAMA and mouse platelets. Blood 126, 1601–1608 (2015).
87. Kearon, C. et al. Antithrombotic therapy for VTE 311, 2414–2421 (2014). 130. Slater, A. et al. Does fibrin(ogen) bind to monomeric
disease: CHEST guideline and expert panel report. 108. Griffin, J. H., Zlokovic, B. V. & Mosnier, L. O. Activated or dimeric GPVI, or not at all? Platelets 30, 281–289
Chest 149, 315–352 (2016). protein C, protease activated receptor 1, and (2019).

www.nature.com/nrd
Reviews

131. Onselaer, M. B. et al. Fibrin and D-dimer bind to 155. Markus, H. S. et al. The von Willebrand inhibitor non-ST-segment elevation acute coronary syndrome:
monomeric GPVI. Blood Adv. 1, 1495–1504 (2017). ARC1779 reduces cerebral embolization after carotid the ANTHEM-TIMI-32 trial. J. Am. Coll. Cardiol. 49,
132. Ebrahim, M. et al. Dimeric glycoprotein VI binds to endarterectomy: a randomized trial. Stroke 42, 2398–2407 (2007).
collagen but not to fibrin. Thromb. Haemost. 118, 2149–2153 (2011). 180. Vincent, J. L., Artigas, A., Petersen, L. C. & Meyer, C.
351–361 (2018). 156. Zheng, L. et al. Therapeutic efficacy of the platelet A multicenter, randomized, double-blind, placebo-
133. Induruwa, I. et al. Platelet collagen receptor glycoprotein Ib antagonist anfibatide in murine models controlled, dose-escalation trial assessing safety and
glycoprotein VI-dimer recognizes fibrinogen and fibrin of thrombotic thrombocytopenic purpura. Blood Adv. efficacy of active site inactivated recombinant factor
through their D-domains, contributing to platelet 1, 75–83 (2016). VIIa in subjects with acute lung injury or acute
adhesion and activation during thrombus formation. 157. Li, T. T. et al. A novel snake venom-derived GPIb respiratory distress syndrome. Crit. Care Med. 37,
J. Thromb. Haemost. 16, 389–404 (2018). antagonist, anfibatide, protects mice from acute 1874–1880 (2009).
134. Schulz, C. et al. Platelet GPVI binds to collagenous experimental ischaemic stroke and reperfusion injury. 181. Wheeler, A. P. & Gailani, D. The intrinsic pathway of
structures in the core region of human atheromatous Br. J. Pharmacol. 172, 3904–3916 (2015). coagulation as a target for antithrombotic therapy.
plaque and is critical for atheroprogression in vivo. 158. Lei, X. et al. Anfibatide, a novel GPIb complex Hematol. Oncol. Clin. North Am. 30, 1099–1114
Basic. Res. Cardiol. 103, 356–367 (2008). antagonist, inhibits platelet adhesion and thrombus (2016).
135. Reininger, A. J. et al. A 2-step mechanism of arterial formation in vitro and in vivo in murine models of 182. Nickel, K. F., Long, A. T., Fuchs, T. A., Butler, L. M.
thrombus formation induced by human atherosclerotic thrombosis. Thromb. Haemost. 111, 279–289 & Renne, T. Factor XII as a therapeutic target in
plaques. J. Am. Coll. Cardiol. 55, 1147–1158 (2010). (2014). thromboembolic and inflammatory diseases.
136. Kuijpers, M. J. et al. Complementary roles of platelets 159. Quach, M. E., Chen, W. & Li, R. Mechanisms of Arterioscler. Thromb. Vasc. Biol. 37, 13–20 (2017).
and coagulation in thrombus formation on plaques platelet clearance and translation to improve platelet 183. Weitz, J. I. & Fredenburgh, J. C. Platelet polyphosphate:
acutely ruptured by targeted ultrasound treatment: storage. Blood 131, 1512–1521 (2018). the long and the short of it. Blood 129, 1574–1575
a novel intravital model. J. Thromb. Haemost. 7, 160. Gratacap, M. P. et al. Regulation and roles of (2017).
152–161 (2009). PI3Kbeta, a major actor in platelet signaling and 184. Kenne, E. & Renne, T. Factor XII: a drug target for safe
137. Nurden, A. T. Clinical significance of altered collagen- functions. Adv. Enzyme Regul. 51, 106–116 (2011). interference with thrombosis and inflammation. Drug.
receptor functioning in platelets with emphasis on 161. Martin, V. et al. Deletion of the p110β isoform of Discov. Today 19, 1459–1464 (2014).
glycoprotein VI. Blood Rev. 38, 100592 (2019). phosphoinositide 3-kinase in platelets reveals its 185. Verhamme, P. et al. Tolerability and pharmacokinetics
138. Arthur, J. F., Dunkley, S. & Andrews, R. K. Platelet central role in Akt activation and thrombus formation of TB-402 in healthy male volunteers. Clin. Ther. 32,
glycoprotein VI-related clinical defects. Br. J. Haematol. in vitro and in vivo. Blood 115, 2008–2013 (2010). 1205–1220 (2010).
139, 363–372 (2007). 162. Nylander, S. et al. Human target validation of 186. Verhamme, P. et al. Single intravenous administration of
139. Gruner, S. et al. Relative antithrombotic effect of phosphoinositide 3-kinase (PI3K)β: effects on platelets TB-402 for the prophylaxis of venous thromboembolism
soluble GPVI dimer compared with anti-GPVI and insulin sensitivity, using AZD6482 a novel PI3Kβ after total knee replacement: a dose-escalating,
antibodies in mice. Blood 105, 1492–1499 (2005). inhibitor. J. Thromb. Haemost. 10, 2127–2136 (2012). randomized, controlled trial. J. Thromb. Haemost. 9,
140. Reimann, A. et al. Combined administration of the 163. Zheng, Z. et al. Discovery and antiplatelet activity of 664–671 (2011).
GPVI-Fc fusion protein Revacept with low-dose a selective PI3Kβ inhibitor (MIPS-9922). Eur. J. Med. 187. Eriksson, B. I. et al. Partial factor IXa inhibition with
thrombolysis in the treatment of stroke. Heart Int. 11, Chem. 122, 339–351 (2016). TTP889 for prevention of venous thromboembolism:
e10–e16 (2016). 164. Nylander, S., Wagberg, F., Andersson, M., Skarby, T. an exploratory study. J. Thromb. Haemost. 6,
141. Ungerer, M. et al. Novel antiplatelet drug revacept & Gustafsson, D. Exploration of efficacy and bleeding 457–463 (2008).
(dimeric glycoprotein VI-Fc) specifically and efficiently with combined phosphoinositide 3-kinase β inhibition 188. Toomey, J. R. et al. Comparing the antithrombotic
inhibited collagen-induced platelet aggregation and aspirin in man. J. Thromb. Haemost. 13, efficacy of a humanized anti-factor IX(a) monoclonal
without affecting general hemostasis in humans. 1494–1502 (2015). antibody (SB 249417) to the low molecular weight
Circulation 123, 1891–1899 (2011). 165. Bekendam, R. H. & Flaumenhaft, R. Inhibition of heparin enoxaparin in a rat model of arterial
142. Schupke, S. et al. Revacept, a novel inhibitor of protein disulfide isomerase in thrombosis. Basic Clin. thrombosis. Thromb. Res. 100, 73–79 (2000).
platelet adhesion, in patients undergoing elective Pharmacol. Toxicol. 119, 42–48 (2016). 189. Toomey, J. R. et al. Inhibition of factor IX(a) is
PCI-Design and rationale of the randomized 166. Jasuja, R. et al. Protein disulfide isomerase inhibitors protective in a rat model of thromboembolic stroke.
ISAR-PLASTER Trial. Thromb. Haemost. 119, constitute a new class of antithrombotic agents. Stroke 33, 578–585 (2002).
1539–1545 (2019). J. Clin. Invest. 122, 2104–2113 (2012). 190. Benincosa, L. J. et al. Pharmacokinetics and
143. Lebozec, K., Jandrot-Perrus, M., Avenard, G., 167. Stopa, J. D. et al. Protein disulfide isomerase inhibition pharmacodynamics of a humanized monoclonal
Favre-Bulle, O. & Billiald, P. Design, development and blocks thrombin generation in humans by interfering antibody to factor IX in cynomolgus monkeys.
characterization of ACT017, a humanized Fab that with platelet factor V activation. JCI Insight 2, e89373 J. Pharmacol. Exp. Ther. 292, 810–816 (2000).
blocks platelet’s glycoprotein VI function without (2017). 191. Chow, F. S. et al. Pharmacokinetic and
causing bleeding risks. MAbs 9, 945–958 (2017). 168. Zwicker, J. I. et al. Targeting protein disulfide pharmacodynamic modeling of humanized anti-factor
144. Voors-Pette, C. et al. Safety and tolerability, isomerase with the flavonoid isoquercetin to improve IX antibody (SB 249417) in humans. Clin. Pharmacol.
pharmacokinetics, and pharmacodynamics of ACT017, hypercoagulability in advanced cancer. JCI Insight 4, Ther. 71, 235–245 (2002).
an antiplatelet GPVI (Glycoprotein VI) Fab. Arterioscler. 125851 (2019). 192. Dyke, C. K. et al. First-in-human experience of an
Thromb. Vasc. Biol. 39, 956–964 (2019). 169. Schwarz, M. et al. Conformation-specific blockade of antidote-controlled anticoagulant using RNA aptamer
145. Bergmeier, W., Chauhan, A. K. & Wagner, D. D. the integrin GPIIb/IIIa: a novel antiplatelet strategy technology: a phase 1a pharmacodynamic evaluation
Glycoprotein Ibα and von Willebrand factor in primary that selectively targets activated platelets. Circ. Res. of a drug-antidote pair for the controlled regulation of
platelet adhesion and thrombus formation: lessons 99, 25–33 (2006). factor IXa activity. Circulation 114, 2490–2497
from mutant mice. Thromb. Haemost. 99, 264–270 170. Hanjaya-Putra, D. et al. Platelet-targeted dual (2006).
(2008). pathway antithrombotic inhibits thrombosis with 193. Povsic, T. J. et al. A phase 2, randomized, partially
146. Denorme, F. & De Meyer, S. F. The VWF-GPIb axis preserved hemostasis. JCI Insight 3, 99329 (2018). blinded, active-controlled study assessing the efficacy
in ischaemic stroke: lessons from animal models. 171. Shen, B. et al. A directional switch of integrin and safety of variable anticoagulation reversal using
Thromb. Haemost. 116, 597–604 (2016). signalling and a new anti-thrombotic strategy. Nature the REG1 system in patients with acute coronary
147. Kremer Hovinga, J. A. et al. Thrombotic 503, 131–135 (2013). syndromes: results of the RADAR trial. Eur. Heart J.
thrombocytopenic purpura. Nat. Rev. Dis. Primers 3, 172. Ikei, K. N. et al. Investigations of human platelet-type 34, 2481–2489 (2013).
17020 (2017). 12-lipoxygenase: role of lipoxygenase products in 194. Lincoff, A. M. et al. Effect of the REG1 anticoagulation
148. Mikhail, S., Aldin, E. S., Streiff, M. & Zeidan, A. platelet activation. J. Lipid Res. 53, 2546–2559 system versus bivalirudin on outcomes after
An update on type 2B von Willebrand disease. (2012). percutaneous coronary intervention (REGULATE-PCI):
Expert. Rev. Hematol. 7, 217–231 (2014). 173. Yeung, J. et al. 12-lipoxygenase activity plays an a randomised clinical trial. Lancet 387, 349–356
149. Firbas, C., Siller-Matula, J. M. & Jilma, B. Targeting important role in PAR4 and GPVI-mediated platelet (2016).
von Willebrand factor and platelet glycoprotein Ib reactivity. Thromb. Haemost. 110, 569–581 (2013). 195. Povsic, T. J. et al. Pre-existing anti-PEG antibodies are
receptor. Expert. Rev. Cardiovasc. Ther. 8, 1689–1701 174. Yeung, J. et al. Platelet 12-LOX is essential for associated with severe immediate allergic reactions to
(2010). FcgγRIIa-mediated platelet activation. Blood 124, pegnivacogin, a PEGylated aptamer. J. Allergy Clin.
150. Gilbert, J. C. et al. First-in-human evaluation of anti 2271–2279 (2014). Immunol. 138, 1712–1715 (2016).
von Willebrand factor therapeutic aptamer ARC1779 175. Adili, R. et al. First selective 12-LOX inhibitor, ML355, 196. Weitz, J. I. & Harenberg, J. New developments in
in healthy volunteers. Circulation 116, 2678–2686 impairs thrombus formation and vessel occlusion anticoagulants: past, present and future. Thromb.
(2007). in vivo with minimal effects on hemostasis. Arterioscler. Haemost. 117, 1283–1288 (2017).
151. Bartunek, J. et al. Novel antiplatelet agents: ALX-0081, Thromb. Vasc. Biol. 37, 1828–1839 (2017). 197. Weitz, J. I. & Fredenburgh, J. C. 2017 scientific sessions
a nanobody directed towards von Willebrand factor. 176. Honigberg, L. A. et al. The Bruton tyrosine kinase sol sherry distinguished lecture in thrombosis: factor xi
J. Cardiovasc. Transl. Res. 6, 355–363 (2013). inhibitor PCI-32765 blocks B-cell activation and is as a target for new anticoagulants. Arterioscler. Thromb.
152. Kageyama, S., Matsushita, J. & Yamamoto, H. Effect of efficacious in models of autoimmune disease and Vasc. Biol. 38, 304–310 (2018).
a humanized monoclonal antibody to von Willebrand B-cell malignancy. Proc. Natl Acad. Sci. USA 107, 198. Younis, H. S. et al. Antisense inhibition of coagulation
factor in a canine model of coronary arterial 13075–13080 (2010). factor XI prolongs APTT without increased bleeding
thrombosis. Eur. J. Pharmacol. 443, 143–149 177. Brown, J. R. et al. Incidence of and risk factors for risk in cynomolgus monkeys. Blood 119, 2401–2408
(2002). major haemorrhage in patients treated with ibrutinib: (2012).
153. Kageyama, S. et al. Pharmacokinetics and an integrated analysis. Br. J. Haematol. 184, 199. Buller, H. R. et al. Factor XI antisense oligonucleotide
pharmacodynamics of AJW200, a humanized 558–569 (2019). for prevention of venous thrombosis. N. Engl. J. Med.
monoclonal antibody to von Willebrand factor, in 178. Busygina, K. et al. Oral Bruton tyrosine kinase 372, 232–240 (2015).
monkeys. Arterioscler. Thromb. Vasc. Biol. 22, inhibitors selectively block atherosclerotic The trial shows that reducing levels of FXI using
187–192 (2002). plaque-triggered thrombus formation in humans. an antisense oligonucleotide is associated with
154. Scully, M. et al. Caplacizumab treatment for acquired Blood 131, 2605–2616 (2018). reduced venous thrombosis in patients undergoing
thrombotic thrombocytopenic purpura. N. Engl. J. Med. 179. Giugliano, R. P. et al. Recombinant nematode total knee arthroplasty without increasing
380, 335–346 (2019). anticoagulant protein c2 in patients with bleeding.

Nature Reviews | Drug Discovery


Reviews

200. Gomez-Outes, A., Garcia-Fuentes, M. & polymorphism on platelet function testing during 250. Barrowcliffe, T. W., Mulloy, B., Johnson, E. A.
Suarez-Gea, M. L. Discovery methods of coagulation- single antiplatelet treatment with clopidogrel. & Thomas, D. P. The anticoagulant activity of heparin:
inhibiting drugs. Expert. Opin. Drug. Discov. 12, Thromb. J. 9, 4 (2011). measurement and relationship to chemical structure.
1195–1205 (2017). 226. Montalescot, G. et al. Prasugrel compared with J. Pharm. Biomed. Anal. 7, 217–226 (1989).
201. Weitz, J. I. et al. Effect of Osocimab in preventing clopidogrel in patients undergoing percutaneous 251. Schroeder, M. et al. Protamine neutralisation of low
venous thromboembolism among patients undergoing coronary intervention for ST-elevation myocardial molecular weight heparins and their oligosaccharide
knee arthroplasty. JAMA 323, 130–139 (2020). infarction (TRITON-TIMI 38): double-blind, randomised components. Anal. Bioanal. Chem. 399, 763–771
202. Koch, A. W. et al. MAA868, a novel FXI antibody with controlled trial. Lancet 373, 723–731 (2009). (2011).
a unique binding mode, shows durable effects on 227. Bonaca, M. P., Braunwald, E. & Sabatine, M. S. 252. Link, K. P. The discovery of dicumarol and its sequels.
markers of anticoagulation in humans. Blood 133, Long-term use of ticagrelor in patients with prior Circulation 19, 97–107 (1959).
1507–1516 (2019). myocardial infarction. N. Engl. J. Med. 373, 253. Pirmohamed, M. Warfarin: almost 60 years old and
203. Cheng, Q. et al. A role for factor XIIa-mediated 1271–1275 (2015). still causing problems. Br. J. Clin. Pharmacol. 62,
factor XI activation in thrombus formation in vivo. 228. Vranckx, P. et al. Ticagrelor plus aspirin for 1 month, 509–511 (2006).
Blood 116, 3981–3989 (2010). followed by ticagrelor monotherapy for 23 months vs 254. Presnell, S. R. & Stafford, D. W. The vitamin
204. Lorentz, C. U. et al. Contact activation inhibitor and aspirin plus clopidogrel or ticagrelor for 12 months, K-dependent carboxylase. Thromb. Haemost. 87,
factor XI antibody, AB023, produces safe, dose- followed by aspirin monotherapy for 12 months after 937–946 (2002).
dependent anticoagulation in a phase 1 first-in-human implantation of a drug-eluting stent: a multicentre, 255. Tie, J. K. & Stafford, D. W. Structural and functional
trial. Arterioscler. Thromb. Vasc. Biol. 39, 799–809 open-label, randomised superiority trial. Lancet 392, insights into enzymes of the vitamin K cycle. J. Thromb.
(2019). 940–949 (2018). Haemost. 14, 236–247 (2016).
205. Lakbakbi, S. et al. Tissue factor expressed by adherent 229. Coller, B. S. Anti-GPIIb/IIIa drugs: current strategies 256. Moualla, H. & Garcia, D. Vitamin K antagonists–current
cells contributes to hemodialysis-membrane and future directions. Thromb. Haemost. 86, concepts and challenges. Thromb. Res. 128, 210–215
thrombogenicity. Thromb. Res. 144, 218–223 (2016). 427–443 (2001). (2011).
206. Bjorkqvist, J. et al. Defective glycosylation of 230. Bennett, J. S. Structure and function of the platelet 257. Hanley, J. P. Warfarin reversal. J. Clin. Pathol. 57,
coagulation factor XII underlies hereditary angioedema integrin αIIbβ3. J. Clin. Invest. 115, 3363–3369 1132–1139 (2004).
type III. J. Clin. Invest. 125, 3132–3146 (2015). (2005). 258. Gage, B. F. & Lesko, L. J. Pharmacogenetics of warfarin:
207. Jain, S. et al. Nucleic acid scavengers inhibit 231. Chew, D. P., Bhatt, D. L. & Topol, E. J. Oral regulatory, scientific, and clinical issues. J. Thromb.
thrombosis without increasing bleeding. Proc. Natl glycoprotein IIb/IIIa inhibitors: why don’t they work? Thrombolysis 25, 45–51 (2008).
Acad. Sci. USA 109, 12938–12943 (2012). Am. J. Cardiovasc. Drugs 1, 421–428 (2001). 259. Futatani, T., Watanabe, C., Baba, Y., Tsukada, S.
208. Travers, R. J., Shenoi, R. A., Kalathottukaren, M. T., 232. Cox, D. Oral GPIIb/IIIa antagonists: what went wrong? & Ochs, H. D. Bruton’s tyrosine kinase is present in
Kizhakkedathu, J. N. & Morrissey, J. H. Nontoxic Curr. Pharm. Des. 10, 1587–1596 (2004). normal platelets and its absence identifies patients
polyphosphate inhibitors reduce thrombosis while 233. Kahn, M. L. et al. A dual thrombin receptor system for with X-linked agammaglobulinaemia and carrier
sparing hemostasis. Blood 124, 3183–3190 (2014). platelet activation. Nature 394, 690–694 (1998). females. Br. J. Haematol. 114, 141–149 (2001).
209. von Bruhl, M. L. et al. Monocytes, neutrophils, and 234. Franchi, F., Rollini, F., Park, Y. & Angiolillo, D. J. 260. Bain, J. & Meyer, A. Comparison of bivalirudin to
platelets cooperate to initiate and propagate venous Platelet thrombin receptor antagonism with vorapaxar: lepirudin and argatroban in patients with
thrombosis in mice in vivo. J. Exp. Med. 209, 819–835 pharmacology and clinical trial development. Future heparin-induced thrombocytopenia. Am. J. Health
(2012). Cardiol. 11, 547–564 (2015). Syst. Pharm. 72 (Suppl. 2), 104–109 (2015).
210. Brill, A. et al. Neutrophil extracellular traps promote 235. O’Donoghue, M. L. et al. Safety and tolerability of 261. Connolly, S. J. et al. Dabigatran versus warfarin in
deep vein thrombosis in mice. J. Thromb. Haemost. atopaxar in the treatment of patients with acute patients with atrial fibrillation. N. Engl. J. Med. 361,
10, 136–144 (2012). coronary syndromes: the lessons from antagonizing 1139–1151 (2009).
211. Leal, A. C. et al. Tumor-derived exosomes induce the the cellular effects of thrombin-acute coronary The trial shows that, in patients with atrial
formation of neutrophil extracellular traps: implications syndromes trial. Circulation 123, 1843–1853 (2011). fibrillation, dabigatran was associated with similar
for the establishment of cancer-associated thrombosis. 236. Wiviott, S. D. et al. Randomized trial of atopaxar in rates of stroke and systemic embolism to those
Sci. Rep. 7, 6438 (2017). the treatment of patients with coronary artery associated with warfarin but with lower rates of
212. Hisada, Y. et al. Neutrophils and neutrophil disease: the lessons from antagonizing the cellular major haemorrhage.
extracellular traps enhance venous thrombosis in mice effect of thrombin-coronary artery disease trial. 262. Pollack, C. V. Jr. et al. Idarucizumab for dabigatran
bearing human pancreatic tumors. Haematologica Circulation 123, 1854–1863 (2011). reversal. N. Engl. J. Med. 373, 511–520 (2015).
105, 218–225 (2020). 237. Goto, S. et al. Double-blind, placebo-controlled 263. Pollack, C. V. Jr. et al. Idarucizumab for dabigatran
213. Davis, J. C. Jr. et al. Recombinant human Dnase I Phase II studies of the protease-activated receptor 1 reversal – full cohort analysis. N. Engl. J. Med. 377,
(rhDNase) in patients with lupus nephritis. Lupus 8, antagonist E5555 (atopaxar) in Japanese patients 431–441 (2017).
68–76 (1999). with acute coronary syndrome or high-risk coronary 264. Hutcherson, T. C., Cieri-Hutcherson, N. E. & Bhatt, R.
214. Konigsbrugge, O. & Ay, C. Atrial fibrillation in patients artery disease. Eur. Heart J. 31, 2601–2613 (2010). Evidence for idarucizumab (Praxbind) in the reversal
with end-stage renal disease on hemodialysis: 238. Kernan, W. N. et al. Guidelines for the prevention of of the direct thrombin inhibitor dabigatran: review
magnitude of the problem and new approach to oral stroke in patients with stroke and transient ischemic following the RE-VERSE AD full cohort analysis. P. T.
anticoagulation. Res. Pract. Thromb. Haemost. 3, attack: a guideline for healthcare professionals from 42, 692–698 (2017).
578–588 (2019). the American Heart Association/American Stroke 265. Pour-Ghaz, I. et al. Transcatheter aortic valve
215. Dentali, F. et al. JAK2V617F mutation for the early Association. Stroke 45, 2160–2236 (2014). replacement with a focus on transcarotid: a review of
diagnosis of Ph-myeloproliferative neoplasms in patients 239. Antithrombotic Trialists Collaboration. Collaborative the current literature. Ann. Transl. Med. 7, 420 (2019).
with venous thromboembolism: a meta-analysis. Blood meta-analysis of randomised trials of antiplatelet 266. Joppa, S. A. et al. A practical review of the emerging
113, 5617–5623 (2009). therapy for prevention of death, myocardial infarction, direct anticoagulants, laboratory monitoring, and
216. Stone, G. W. et al. A prospective natural-history study and stroke in high risk patients. BMJ 324, 71–86 reversal agents. J. Clin. Med. 7, E29 (2018).
of coronary atherosclerosis. N. Engl. J. Med. 364, (2002). 267. Lu, G. et al. A specific antidote for reversal of
226–235 (2011). 240. Hemker, H. C. A century of heparin: past, present and anticoagulation by direct and indirect inhibitors
217. Weber, W. et al. Noninvasive follow-up of GDC-treated future. J. Thromb. Haemost. 14, 2329–2338 (2016). of coagulation factor Xa. Nat. Med. 19, 446–451
saccular aneurysms by MR angiography. Eur. Radiol. 241. Lim, G. B. Milestone 1: discovery and purification of (2013).
11, 1792–1797 (2001). heparin. Nat. Rev. Cardiol. https://doi.org/10.1038/ 268. Connolly, S. J. et al. Full study report of andexanet alfa
218. Wallentin, L. et al. Ticagrelor versus clopidogrel in nrcardio.2017.171 (2017). for bleeding associated with factor Xa Inhibitors.
patients with acute coronary syndromes. N. Engl. 242. Best, C. H. Heparin and thrombosis. Br. Med. J. 2, N. Engl. J. Med. 380, 1326–1335 (2019).
J. Med. 361, 1045–1057 (2009). 977–981 (1938). 269. Heo, Y. A. Andexanet Alfa: first global approval. Drugs
219. Brighton, T. A. et al. Low-dose aspirin for preventing 243. Best, C. H., Cowan, C. & Maclean, D. L. Heparin and 78, 1049–1055 (2018).
recurrent venous thromboembolism. N. Engl. J. Med. the formation of white thrombi. Science 85, 338–339 270. Ruff, C. T. et al. Comparison of the efficacy and safety
367, 1979–1987 (2012). (1937). of new oral anticoagulants with warfarin in patients
220. Becattini, C. et al. Aspirin for preventing the recurrence 244. Walenga, J. M. et al. Fondaparinux: a synthetic with atrial fibrillation: a meta-analysis of randomised
of venous thromboembolism. N. Engl. J. Med. 366, heparin pentasaccharide as a new antithrombotic trials. Lancet 383, 955–962 (2014).
1959–1967 (2012). agent. Expert. Opin. Investig. Drugs 11, 397–407
221. Marik, P. E. & Cavallazzi, R. Extended anticoagulant (2002). Acknowledgements
and aspirin treatment for the secondary prevention of 245. Garcia, D. A., Baglin, T. P., Weitz, J. I. & Samama, M. M. The authors would like to thank Cihan Ay, Jonathan Erlich,
thromboembolic disease: a systematic review and Parenteral anticoagulants: antithrombotic therapy and Steven Grover, Raj Kasthuri, Carolyn Mackman and Dougald
meta-analysis. PLoS One 10, e0143252 (2015). prevention of thrombosis, 9th ed: American College of Monroe III for helpful comments and Yohei Hisada for help
222. Weitz, J. I. et al. Rivaroxaban or aspirin for extended Chest Physicians evidence-based clinical practice with preparing the manuscript.
treatment of venous thromboembolism. N. Engl. J. Med. guidelines. Chest 141 (Suppl. 2), e24–e43 (2012).
376, 1211–1222 (2017). 246. Lee, S. et al. Scientific considerations in the review and Competing interests
223. Elwood, P. C. et al. A randomized controlled trial of approval of generic enoxaparin in the United States. G.A.S. and W.B. have no competing interests. N.M. has a grant
acetyl salicylic acid in the secondary prevention of Nat. Biotechnol. 31, 220–226 (2013). from Bayer and is a consultant for Bayer. J.I.W. is a consultant
mortality from myocardial infarction. Br. Med. J. 1, 247. Wienbergen, H. & Zeymer, U. Management of acute for Bristol-Myers Squibb, Pfizer, Daiichi Sankyo, Bayer,
436–440 (1974). coronary syndromes with fondaparinux. Vasc. Health Janssen, Boehringer Ingelheim, IONIS Pharmaceuticals,
224. Janzon, L. et al. Prevention of myocardial infarction Risk Manag. 3, 321–329 (2007). Merck, Portola, Perosphere, Servier, Anthos and Tetherex.
and stroke in patients with intermittent claudication; 248. Arepally, G. M. Heparin-induced thrombocytopenia.
effects of ticlopidine. Results from STIMS, the Swedish Blood 129, 2864–2872 (2017). Publisher’s note
Ticlopidine Multicentre Study. J. Intern. Med. 227, 249. Nutescu, E. A., Burnett, A., Fanikos, J., Spinler, S. Springer Nature remains neutral with regard to jurisdictional
301–308 (1990). & Wittkowsky, A. Pharmacology of anticoagulants claims in published maps and institutional affiliations.
225. Pettersen, A. A., Arnesen, H., Opstad, T. B. used in the treatment of venous thromboembolism.
& Seljeflot, I. The influence of CYP 2C19*2 J. Thromb. Thrombolysis 41, 15–31 (2016). © Springer Nature Limited 2020

www.nature.com/nrd

You might also like