You are on page 1of 11

ISBT Science Series (2019) 0, 1–11

© 2019 International Society of Blood Transfusion


INVITED REVIEW DOI: 10.1111/voxs.12529

Anti-platelet agents: past, present and future


Dermot Cox
Molecular & Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland

The discovery of the role of platelets in cardiovascular disease led to the intro-
duction of aspirin as a therapeutic agent for heart disease. Aspirin has proven to
be effective in preventing cardiovascular events but the fact that not all patients
benefit from aspirin has led to the search for more effective agents. Numerous
agents have been discovered including thromboxane synthase inhibitors, phos-
phodiesterase inhibitors, GPIIb/IIIa antagonists, P2Y12 antagonists and PAR-1
antagonists. However, all faced two challenges – their cost-benefit relationship in
comparison with aspirin and the risk of bleeding that is often associated with
greater efficacy. In this paper, I review the performances of these different classes
of anti-platelet agent and discuss the potential future for novel anti-thrombotics.
I also highlight the role of platelets in immunology and discuss the potential for
targeting the immune properties of platelets.
Received: 12 July 2019,
revised 22 August 2019, Key words: anti-platelet agents, aspirin, GPIIb/IIIa antagonists, P2Y12 inhibitors,
accepted 27 August 2019 immunology, thrombosis, FcgammaRIIa, infection.

the latest technologies including genomics and pro-


Introduction
teomics have been deployed to discover the ultimate anti-
The discovery of the role of platelets in thrombosis and platelet agents. Below I will review the mechanism of
the realization that this was a major factor in the patho- action of the different anti-platelet agents and discuss
genesis of a myocardial infarction clearly identified plate- their clinical effectiveness. I will also discuss the proper-
lets as a very significant drug target [1]. As ties of the ultimate anti-platelet agent and whether it is
cardiovascular disease is the major cause of death in the worth pursuing this elusive goal.
world this has led to major pharmaceutical industry
resources being committed to developing the ultimate
Inhibition of COX pathway
anti-platelet agent especially since it is a lifelong chronic
disease affecting a large number of people worldwide.
Aspirin
This search for the perfect anti-platelet agent has been
on-going for 50-years and has been affected by the major The first successful anti-platelet agent was aspirin [2]
changes in the drug discovery process. Anti-platelet which was discovered based on anecdotal accounts of
agents were initially discovered by anecdote – where bleeding being associated with its use [3]. The long path
drugs known to cause bleeding as a side-effect were used. to understanding the mechanism of action of aspirin also
Some agents were discovered using chemistry-led drug provided great insight into platelet function. The key dis-
discovery – where drugs were screened for in vivo anti- covery that aspirin inhibited prostaglandin synthesis [4]
thrombotic activity with no knowledge of how they allowed some of the key signalling pathway in platelets
worked. More recently, drugs were discovered by target- to be characterised (Fig. 1)[5].
led discovery strategies where drugs were designed to tar- Platelet activation by most agonists leads to the activa-
get specific platelet receptors/enzymes. Currently, all of tion of phospholipase A2 which cleaves arachidonic acid
from the inner platelet membrane [6]. This arachidonic
acid is a substrate for cyclooxygenase (COX) which con-
Correspondence: Prof Dermot Cox, MCT, Royal College of Surgeons in verts it into prostaglandin (PG) H2. PGH2 is in turn con-
Ireland, 123 St Stephens Green, Dublin D2, Ireland verted into thromboxane (Tx) A2 – a powerful platelet
Email: dcox@rcsi.ie

1
2 D. Cox

Fig. 1 Platelet activation pathways and the sites of action of anti-platelet agents.

agonist – by the enzyme thromboxane synthase. This cre- patients do worse and would benefit from alternative
ates a feedback loop where the secreted TXA2 activates treatments. It is obvious that if a patient does not respond
further platelets. Aspirin (acetylsalicylic acid) is an irre- to aspirin they will not get its benefit and as a result will
versible inhibitor of COX – it binds to Ser530 in the cat- have a worse prognosis. This in fact has been well estab-
alytic site and acetylates it. As a result, arachidonic acid lished in a meta-analysis [10]. However, the big question
cannot gain access to the catalytic site [7]. is what exactly is aspirin resistance and how should it be
detected?
Aspirin effectiveness There are a number of different types of aspirin resis-
There are many ways of assessing effectiveness of anti- tance: pharmacodynamic, pharmacokinetic and clinical
platelet agents such as mortality (30 days or 90 days) and [11,12]. These are masked by factors such as non-compli-
further cardiovascular events. Often a combined triple ance and drug–drug interactions due to poly-pharmacy.
endpoint of death, myocardial infarction (MI) or urgent All of this is dependent on the assay that is used to detect
revascularization (e.g. need for stenting) is used. There aspirin resistance.
have been many studies that have shown that aspirin is
clinically effective in cardiovascular disease. In particular, Non-compliance. Like any other medication, aspirin
a major meta-analysis by the Antithrombotic Trialists only works when patients take it. As aspirin is usually
Collaboration in 2002 clearly showed a reduction of used after an MI, it would be expected that patients
around 25% in the event rate [8]. The benefit was found would be motivated to take it. However, there is a signifi-
in both stroke and MI patients. cant level of non-compliance among cardiovascular
patients in general [13]. Non-compliance with anti-
Aspirin resistance thrombotics has been found to be around 18% [14]. There
The fact that the benefit of aspirin is only about 25% at are a number of reasons for this non-compliance. Patients
best led to the concept of aspirin resistance [9]. In other are usually on multiple medications with different dosing
words, some patients fail to respond to aspirin and these regimens, and this can easily lead to confusion. This

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
Future of anti-platelet agents 3

problem can be resolved by using blister packaging where independent or a platelet-mediated event that is COX-in-
all of the tablets for a specific time are in a single pack- dependent. Platelet agonists can be classed into weak
age [13]. Another problem is lack of knowledge. Patients agonists which act through COX and strong agonists such
often think that aspirin is not important as it is available as thrombin which activate platelets through phospholi-
in supermarkets! However, in our experience education is pase (PL)C-mediated cleavage of inositol triphosphate
helpful here. This involves a nurse, doctor or pharmacist (IP3) and diacylglycerol (DAG) from the membrane. IP3
explaining to the patient what aspirin does and why it is binds to an intracellular Ca2+ channel increasing intracel-
so important. Another reason for non-compliance is lular Ca2+ levels through release from the endoplasmic
adverse effects. A patient may stop taking aspirin as it reticulum. DAG ultimately activates protein kinase C
upsets their stomach. It is important to identify this and which activates further downstream events [5]. Clearly,
provide appropriate treatment such as a co-administration activation through this pathway will not be affected by
of a proton pump inhibitor or misoprostol [15]. aspirin. Furthermore, many agonists work through both
Pharmacodynamic resistance occurs where there are pathways – COX activation at low doses and PLC activa-
detectable levels of aspirin in the blood but no detectable tion at high doses. The existence of this alternative path-
pharmacological response. A number of studies have way has led to the search for the next-generation anti-
shown evidence of true resistance to aspirin often due to platelet agent.
polymorphisms in COX [16] or other platelet enzymes/re-
ceptors [17] and reviewed by Goodman et al. [18]. How-
Targeting thromboxane
ever, the incidence of this resistant phenotype seems to
be low. Studies where patients weight is taken in consid- One approach to dealing with aspirin resistance was to
eration show that all patients respond suggesting that the target other parts of the PLA2 signalling pathway. Two
incidence of pharmacodynamic resistance is likely to be targets that were selected were the downstream enzyme
less than 1% [19,20]. thromboxane synthase which like aspirin would totally
Pharmacokinetic resistance occurs when a patient takes block the synthesis of thromboxane A2. The other target
their aspirin and there is no detectable aspirin in the was the thromboxane A2 receptor which could be blocked
blood. This appears to be the major cause of aspirin non- with small molecule antagonists. Some agents were also
response after non-compliance. Primarily, it is due to discovered that had activity on both the thromboxane
under-dosing. Most patients are on low-dose enteric- synthase enzyme and receptor [27].
coated aspirin but this is not bioequivalent with plain Neither thromboxane receptor antagonists (e.g. seratro-
aspirin [21,22]. In fact, different preparations of enteric dast, terutroban) nor synthase inhibitors have shown ben-
coating are not even bioequivalent with each other [23]. efit over aspirin. So, while effective they could never
This reduced bioavailability of enteric-coated aspirin is compete with aspirin on cost. This is not surprising. Once
particularly a problem in heavier patients where aspirin issues of weight and aspirin bioavailability are taken into
dose (mg/kg) is likely to fall below the minimum effective consideration virtually all patients show complete inhibi-
dose [19]. While it is know that evidence for aspirin non- tion (>97% inhibition) of thromboxane production [21].
response is assay-dependent with very poor agreement Thus, blocking either the thromboxane synthase enzyme
between different assays the association of weight with or receptor is likely to achieve more than 97% inhibition
poor response appears to be true with all assays [24]. A and unlikely that any further inhibition would be clini-
further cause of pharmacokinetic resistance is drug–drug cally relevant.
interactions. Both non-steroidal anti-inflammatories
(NSAIDs) and aspirin bind to the same site on COX; how-
GPIIb/IIIa antagonists
ever, NSAIDs are reversible inhibitors while aspirin is
irreversible. Aspirin has a very short half-life while As the thrombi that precipitate an MI are platelet-rich
NSAIDs have much longer half-lives. If a patient takes an this led researchers to investigate how these thrombi are
NSAID before aspirin then the aspirin cannot bind to formed. The discovery that the platelets in these thrombi
COX and is cleared from the circulation [25]. This can be are bound together by fibrinogen binding to specific
resolved by taking aspirin an hour or two before NSAIDs. receptors on platelets led to the prospect for a new class
A similar problem can exist with statins [26]. of anti-thrombotic agent. As fibrinogen is a divalent
Clinical resistance occurs when a patient has a cardio- molecule, when it binds to its receptor on two different
vascular event while on aspirin. If laboratory tests show platelets it cross-links them which is the basis of platelet
that the patient’s platelets are fully inhibited by aspirin aggregation. In the mid-1980s, the platelet receptor for
this is not aspirin resistance. It is evidence that the fibrinogen was identified as a heterodimer of glycopro-
patient had a cardiovascular event that is platelet- teins (GP) IIb and IIIa [28,29]. It soon became clear that

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
4 D. Cox

GPIIb/IIIa was a member of the expanding super-family The reason for the failure of the oral activity is multi-
of integrin cell adhesion molecules (aIIbb3). GPIIb/IIIa factorial [45,46]. At the heart of the problem is the fact
was a drug target with excellent potential as fibrinogen that all of the agents trigger platelet activation by bind-
binding to GPIIb/IIIa was considered to be the final step ing to GPIIb/IIIa [47,48]. Initially, GPIIb/IIIa appeared to
in platelet aggregation. Thus, no matter how platelets be a simple adhesion molecule but it was subsequently
were activated, fibrinogen binding to GPIIb/IIIa was shown that it is a true receptor and binding agonist (RGD
always going to occur. As a result, these agents were is the natural agonist, and most small molecules are ana-
referred to as ‘super-aspirin’ in the media. Furthermore, logues) leads to platelet activation [49]. This was further
the fibrinogen sequence that supported binding was iden- complicated by low bioavailability and poor pharmacoki-
tified as Arg-Gly-Asp (RGD) [30] which provided a hit netics which led to significant under-dosing. As a result,
compound for a drug discovery programme. In one of the all orally active GPIIb/IIIa antagonists were dropped.
first examples of target-led drug discovery, multiple com-
panies set out to discover GPIIb/IIIa antagonists.
P2Y12 antagonists
The first GPIIb/IIIa antagonist was abciximab, a mono-
clonal antibody directed against the GPIIIa subunit and With the failure of GPIIb/IIIa antagonist, there was still a
was one of the first monoclonal antibodies to be commer- need for a drug to take over from aspirin. One possibility
cialized [31]. A second approach used the fact that viper was ticlopidine [50]. This agent was discovered through a
venoms contain very potent RGD-containing GPIIb/IIIa chemistry-led drug discovery programme. Thus, it had
antagonists (disintegrins) [32] which are used to prevent been discovered to be an anti-thrombotic in vivo although
clot formation after a snake bite. Using these as the start- it had no activity in vitro. It was not known how it
ing point led to the discovery of the cyclic-peptide eptifi- worked. While clinically effective, its side-effect profile
batide [33] and the non-peptide tirofiban [34]. These was a problem especially the neutropenia [51,52]. How-
agents all act by binding to GPIIb/III thereby preventing ever, with the failure of oral GPIIb/IIIa antagonist it now
fibrinogen from binding. became the centre of attention.
As most MI patients would be given aspirin prior to Further studies on ticlopidine showed that it was a
arriving at hospital it was clear that clinical trials with pro-drug and that it was metabolized into an active agent
aspirin na€ıve patients would be too difficult aspirin was that bound irreversibly to an ADP receptor on platelets –
co-administered with the GPIIb/IIIa antagonist in all clin- P2Y12. Retaining the active component, a new pro-drug
ical trials. As a result, they are clinically used with was developed. This was clopidogrel and is metabolized
aspirin. to the active form by cytochrome P450 enzymes in the
The clinical trials with these agents were very successful liver. It has the advantage over ticlopidine of not causing
for MI, and a meta-analysis confirmed that they were more neutropenia [53].
effective than aspirin alone [35]. However, as none of these There was initial scepticism about targeting P2Y12 as it
agents were orally active their market was always going to was only one receptor on platelets. While GPIIb/IIIa
be restricted to the acute management of MI. In stroke, antagonists had the potential to block aggregation due to
these agents were found to have little clinical benefit with any agonist, clopidogrel was restricted to ADP activation
increased bleeding risk [36]. At their peak, none of the only – what about collagen, thrombin etc.? Furthermore,
agents had annual sales of more than $300 million and by P2Y12 was a weak agonist and not even the only ADP
2012 the combined sales for all three agents were less than receptor as there was also a P2Y1 receptor as well [54].
$250 million. Currently, they are only used in certain high- However, it turns out that once activated platelets secrete
risk situations [37] and thus far from being a commercial ADP from their granules and this ADP activates other
blockbuster. platelets via P2Y12 thereby recruiting them into the
The solution to this problem was to develop orally growing thrombus. Thus, P2Y12 is a critical receptor in
active agents. Using RGD as a hit compound led to the thrombosis [55].
discovery of xemilofiban and orbofiban [38]. Screening The importance of P2Y12 in thrombosis has been sup-
small molecule libraries led to sibrafiban [39] and the dis- ported by the many clinical trials that have shown that
covery that pentamidine had anti-GPIIb/IIIa activity clopidogrel is a very effective anti-platelet agent for acute
[40,41] led to the discovery of FK633 [42,43]. However, patients [56] and patients undergoing percutaneous coro-
there were surprising results in the clinical trials of all of nary intervention (e.g. angioplasty and stenting) [57],
these agents. Rather than showing a benefit similar to the although it is probably no better than aspirin for stable
IV compounds a meta-analysis showed that all of these disease [58]. Similar to GPIIb/IIIa antagonists, it is not a
compounds led to an increase in cardiovascular mortality replacement for aspirin and is normally used as part of
[44]. dual-therapy with aspirin. Clopidogrel became the 2nd

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
Future of anti-platelet agents 5

biggest selling drug with annual sales of $9 billion in by PKA inhibits platelet activation [66]. While PGI2 and
2010. its analogues are potent anti-platelet agents they are also
potent vasodilators and this vasodilatory activity is very
unpleasant for patients and thus they are not clinically
Clopidogrel resistance
used as anti-platelet agents. Another natural anti-platelet
While highly successful it soon became clear that some agent is nitric oxide (NO) produced by the endothelium. It
patients failed to respond to clopidogrel therapy. It was diffuses into the platelet where it activates a soluble gua-
subsequently found to be due to a failure to convert nyl cyclase leading to an increase in cGMP. This in turn
clopidogrel into the active form. This is due to a poly- activates PKG leading to downstream protein phosphory-
morphism in the CYPP450 enzymes that metabolize clopi- lation [67].
dogrel. Patients with the polymorphism are under- Both cAMP and cGMP are metabolized by phosphodi-
expressers of the enzyme and thus fail to respond to esterase (PDE). Cilostazol is a PDE3 inhibitor, and by
clopidogrel [59]. blocking PDE, it prolongs the activity of cAMP and thus
Prasugrel was one solution to this problem. It was the is an effective anti-platelet agent [68]. Clinical trials have
same active agent as clopidogrel but a different pro-drug supported the effectiveness of cilostazol [69]. Another
that was not affected by the CYP450 polymorphisims agent with PDE inhibitory activity is dipyridamole which
[60]. While a very effective agent with no evidence of is primarily used for the treatment of stroke [70].
resistance it had one major drawback – price. Clopidogrel
is off-patent and as a result much cheaper than the newer
Protease-activated receptor (PAR) inhibitors
prasugrel. Because of the pressures of drug pricing health
authorities preferred to use clopidogrel as first line and Activation of the coagulation system generates thrombin
restrict prasugrel to confirmed cases of clopidogrel resis- which is a very potent platelet activator. Thrombin is a
tance. As a result, by 2016 annual sales of prasugrel were proteolytic enzyme and acts on a unique family of pro-
only $500 million, far below the expected sales. tease-activated receptors (PAR). Thrombin cleaves a ter-
Another approach to the problem of clopidogrel resis- minal peptide from the receptor, and the newly exposed
tance was to develop direct inhibitors of P2Y12. These terminal becomes the agonist peptide and binds to the
were not pro-drugs and were reversible. Reversibility had binding site. There are four members of the family, and
a major advantage during intervention. Clopidogrel was two (PAR-1 and PAR-4) are expressed on human plate-
the drug of choice for use during stenting but sometimes lets. PAR receptors are G-protein-coupled receptors, and
there are complications during stenting, and the patient their activation leads to platelet activation via PLC and
needs to go for by-pass surgery. However, surgeons are thus is independent of aspirin [71]. This led to the devel-
often reluctant to operate on a clopidogrel-treated patient, opment of specific inhibitors of PAR-1 for use as anti-
and as an irreversible inhibitor, it can take some time for platelet agents [72]. Vorapaxar [73] and atopaxar are two
its effects to be reversed or require a platelet transfusion such agents and have been shown to be effective in clini-
[61]. A reversible antagonist would have the advantage of cal trials [74]. However, there is no evidence that they are
rapid reversibility and thus ideal for use during interven- more effective than other agents and have a bleeding risk
tion. Ticagrelor and cangrelor are reversible antagonists associated with them. There is also interest in developing
of P2Y12 [62]. Ticagrelor has proven to be more effective PAR-4 antagonists as anti-platelet agents [75]. As PAR 1
than prasugrel in diabetes [63] and probably than clopi- and 4 are not restricted to platelets some of the potential
dogrel as well [64] and has achieved blockbuster status beneficial effects of these agents may be due to inhibition
with sales in excess of $1 billion annually. On the other of PAR on other cells.
hand, the future of cangrelor is uncertain [65].
Most effective anti-platelet agent
Phosphodiesterase inhibitors
Recently network meta-analyses have been published that
As an MI is due to unwanted platelet activation, it is compared multiple anti-platelet regimens for effective-
important that the body has systems to prevent this ness. In the case of stroke, both cilostazol and clopidogrel
unwanted platelet activation. This is achieved through were found to reduce stroke recurrence and composite
another prostaglandin – prostacyclin (PGI2). It binds to events compared to aspirin or dipyridamole. Dipyridamole
Gas receptor on platelets leading to an increase in cAMP was found to have a good safety profile but had poor
production by adenyl cyclase. This in turn leads to the efficacy. Combining clopidogrel with aspirin improved
activation of Protein kinase A (PKA) which phosphory- efficacy but increased bleeding risk [76]. In the case of
lates many downstream proteins. Protein phosphorylation MI, all of the agents and combinations of agents were

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
6 D. Cox

found to be more effective than placebo. However, when growing awareness that platelets are key components of
compared to aspirin mono-therapy none of the agents the immune system. This is clear in that they express
were more effective except for the addition of the factor immune-associated receptors such as Toll-like receptors
X inhibitor rivaroxaban to the clopidogrel/aspirin combi- (TLR), FccRIIa and CLEC-2 on their surface [86,87]. These
nation [77]. receptors have been shown to be functional on platelets
and engagement of these receptors leads to platelet acti-
vation. However, the activation pathways associated with
Improved anti-platelet agents
these receptors is different to that of the haemostasis-as-
So, with the clear effectiveness of existing anti-platelet sociated receptors. The nature of the activation is also dif-
agents and with at best incremental increases in efficacy ferent [88,89]. For instance, TLR-mediated activation of
of newer agents the search for newer agents has focused platelets does not seem to trigger platelet aggregation
on developing agents that are effective without causing unlike that seen with ADP or thrombin. Instead TLR-me-
bleeding [78–80]. Experience would suggest that this is a diated activation of platelets leads to platelet binding to
difficult proposition. All of the studies have shown that neutrophils which leads to the formation of neutrophil
effectiveness of anti-platelet agents is directly associated extracellular traps (NET) [90].
with increased bleeding risk. So, is it possible to discover Activated platelets also lead to secretion of antimicro-
an agent that inhibits the thrombotic pathways but not bial peptides that kill bacteria [91]. When there is a
the haemostatic pathways? breach in the vasculature (a cut for instance), platelets are
In theory, this is possible. GPIb is the von Willebrand the first responders binding to the exposed extracellular
factor (VWF) receptor on platelets. It plays an important matrix and forming platelet aggregates to prevent further
role in the adhesion of platelets to the damaged endothe- blood loss. They also secrete antimicrobial peptides to
lium as well as subsequent platelet activation [81]. This is sterilize the area and also secrete chemokines and cytoki-
an important first step in thrombosis formation, and what nes that attract immune cells to the area and activate
makes it an important drug target is that this interaction them.
only occurs under high shear conditions. Thus, it does While this immune regulatory activity of platelets is
not play a significant role in the venous system or in a very important it can sometimes cause problems. When
healthy arterial system. It is particularly important in ste- bacteria gain access into the blood they start to grow
nosed arteries such as atherosclerotic coronary arteries. causing a bacteraemia. This leads to platelet activation
Thus, a GPIb antagonist has the potential to block throm- and secretion of antimicrobial peptides. However, some
bosis in atherosclerotic vessels and not healthy vessels strains of bacteria are resistant to platelet-derived antimi-
and thus have little impact on bleeding [82,83] although crobial peptides. In this case, the bacteria continue to
Bernard-Soulier disease (deficiency in GPIb) is associated grow and platelets continue to become activated. These
with a bleeding tendency [84]. However, over the last activated platelets either get cleared in the spleen or form
30 years researchers have been trying to discover GPIb aggregates. The result of the continued bacterial growth
antagonists and none progressed to the market. It is not and continued platelet activation leads to thrombocytope-
clear why all of these projects failed and whether it is nia. Platelet aggregate formation can lead to clot forma-
ever going to be possible to develop such an inhibitor. tion in the microvasculature, a condition known as
However, the cost of developing new agents is very disseminated intravascular coagulation (DIC). This can
high. Most of the agents have produced only modest ben- lead to ischaemic damage which as it spreads causes
efits over aspirin and thus hard to justify their cost. Cur- organ failure as the bacteraemia progresses to sepsis. Pla-
rently, a combination of aspirin and clopidogrel is very telet granules are full of serotonin, and the ongoing pla-
effective and inexpensive. Some of the other agents such telet activation causes the release of large quantities of
as prasugrel and ticagrelor while expensive can be used serotonin which causes vasodilation ultimately leading to
for those patients that fail to respond in combination with septic shock [92].
a CYP450 screening assay [85]. With the cost of a suc- The key role of platelets in the pathogenesis of sepsis is
cessful drug discovery programme at approximately $1 clear from meta-analysis of anti-platelet agents in sepsis
billion, it is hard to see how a new anti-thrombotic dis- although the conclusions are limited by the fact that the
covery programme is financially viable. studies are all retrospective. A network meta-analysis of
critically ill patients showed an odds ratio of 081 for in-
hospital mortality in patients on anti-platelet agents. The
Novel anti-platelet agents
OR for sepsis was 081 as well, and the mortality benefits
While platelets are critical components of haemostasis were associated with sepsis [93]. This is despite the fact
that is not their only function. Recently, there is a that the primary reason for being on an anti-platelet

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
Future of anti-platelet agents 7

agent is for secondary prevention of MI or stroke. Thus, With viruses, it is more complex as there are multiple
these patients did better despite being sicker. A study of receptors involved [95]. FccRIIa certainly mediates some
over 500 000 insurance patients showed that prior use of virus-platelet interactions such as with Dengue [105].
aspirin was associated with between 2% and 12% reduc- Other key receptors are DC-SIGN [105], CLEC-2 and TLRs
tion in mortality [94]. This is not far from the benefit of [106]. Unlike bacteria-platelet interactions, there has been
aspirin in cardiovascular disease. little research into virus-platelet interactions partly
Platelets are also involved with the immune response because some of the viruses such as VHF viruses require
to viral infections. Viruses such as HIV bind to platelets BSL4 conditions and thus are very difficult to work with.
leading to thrombocytopenia. There are also the viral However, with future research in this area it should be
haemorrhagic fevers (VHF), which are viral infections due possible to better characterize virus-platelet interactions
to four families of viruses – Arenaviridae, Bunyaviridae, and identify novel targets.
Filoviridae and Flaviviridae. These include viruses such as
Dengue, Ebola, Yellow fever and Lassa. These are all asso-
Conclusions
ciated with thrombocytopenia and can cause a coagu-
lopathy similar to DIC in sepsis [95,96]. Anti-platelet drug discovery has been an area of mixed
In both sepsis and VHF, the DIC is a major contributor results. Aspirin, a drug known for centuries proved to be
to mortality and morbidity. Managing the coagulopathy an effective agent for treatment of cardiovascular disease
is an important strategy in treating patients. In the case and despite all of the money spent of drug discovery in
of sepsis, there can be a significant delay in identifying this area it is still the most important agent in clinical
the appropriate antibiotics especially with the growing use. Identified as a COX inhibitor, all attempts to generate
incidence of antibiotic resistant strains of bacteria. By improved agents that target the COX pathway have failed
controlling the coagulopathy, an anti-platelet agent can to produce an agent with an improved cost-benefit pro-
buy time to identify the best antibiotic to use. In the case file. GPIIb/IIIa antagonists were the next category of anti-
of VHF, there are no drugs but if patients survive the platelet agent that promised that target-led drug discov-
coagulopathy their immune system can clear the infec- ery could produce a new super-aspirin. However, while
tion. An anti-platelet agent may play a key role in showing a small clinical benefit over aspirin their lack of
managing the VHF DIC. oral activity and increase in bleeding has meant that they
The problem with using anti-platelet agents in sepsis/ are a niche class of drugs used for high-risk interventions
VHF is that by their very nature they inhibit platelet only. P2Y12 antagonists are the real success story in
function and thereby increase the risk of bleeding. In a thrombosis. Using a chemistry-led approach, ticlopidine
patient with thrombocytopenia due to infection this was discovered and further refined to improve its safety
increased bleeding risk is much more significant. Thus, profile leading to clopidogrel. At its peak, it was the 2nd
anti-platelet agents preserve platelet number over platelet biggest selling drug in the world and now that it is off-
function. However, as both viruses and bacteria activate patent it is the drug of choice for prevention of events
platelets using receptors entirely distinct from those that post-stenting although usually used in conjunction with
mediate haemostasis it would be possible to discover aspirin. Further attempts to improve on clopidogrel failed
agents that inhibit pathogen-induced platelet activation on the cost-benefit profile although ticagrelor as a rever-
only. sible P2Y12 antagonist has been a commercial success.
Virtually, all bacteria that have been shown to acti- Thus, it is clear that the biggest hurdle for any new anti-
vate platelets do so in an FccRIIa-dependent manner and platelet agent is the cost-benefit profile in comparison to
blocking FccRIIa has been shown to prevent the platelet aspirin/clopidogrel combination. While there maybe
activation [97]. This is true of both Gram-positive bacte- agents that work better when this combination does not
ria such as Staphylococcus aureus [98], Streptococcus work this will always be a minor market.
sanguinis [99] and Streptococcus gordonii [100] and The other big challenge in anti-thrombotic therapy is
Gram-negative such as Escherichia coli [101] and Heli- the safety profile. While some agents such as ticlopidine
cobacter pylori [102]. There are two approaches to have specific adverse effects the increased risk of bleeding
inhibiting FccRIIa – monoclonal antibodies and small is common to all anti-platelet agents. Bleeding risk is a
molecules. Currently, an anti-FccRIIa monoclonal anti- problem with aspirin, especially cerebral bleeding, and as
body is entering phase I clinical trials [103]. Small mole- a result, it is primarily used for secondary prevention
cule antagonists have been discovered and while they [107]. The ideal anti-platelet agent would be one that tar-
may not have sufficient potency to be used clinically it gets thrombosis over haemostasis and would thus mini-
shows that small molecules have the ability to block the mize the bleeding risk. However, while many agents have
receptor [104]. promised to deliver in this area none have been

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
8 D. Cox

successful. While, theoretically possible it remains to be patients with coronary heart disease on confirmed aspirin
seen if such an agent can be discovered that also has a adherence. J Atheroscler Thromb, 2014; 21:239–247
good cost-benefit profile compared with aspirin/clopido- 11 Weber A-A, Przytulski B, Schanz A, et al.: Towards a defini-
grel. tion of aspirin resistance: a typological approach. Platelets,
2002; 13:37–40
So, while the cost-benefit and safety profiles are the
12 Cox D: Aspirin resistance: a nebulous concept. J Clin Phar-
two biggest challenges for a new anti-platelet agent there
macol Pharmacoepidemiol, 2010; 1:39–47
remains one area where there is scope for drug discovery. 13 Lee JK, Grace KA, Taylor AJ: Effect of a pharmacy care pro-
There is significant evidence that platelets are significant gram on medication adherence and persistence, blood pres-
players in the immune system where they play a major sure, and low-density lipoprotein cholesterol: a randomized
role in the response to infection (DIC). Considering that controlled trial. JAMA, 2006; 296:2563–2571
sepsis is the number one cause of in-hospital mortality 14 De Schryver EL, van GijnJ, Kappelle LJ, et al.: Non-adher-
there is the potential for commercial success in this thera- ence to aspirin or oral anticoagulants in secondary preven-
peutic area. As the receptors involved in the immune tion after ischaemic stroke. J Neurol, 2005; 252:1316–1321
functions of platelets are not involved in haemostasis any 15 Newton JL: Improving the gastrointestinal tolerability of
agent targeting them would be unlikely to cause bleeding aspirin in older people. Clin Interv Aging, 2006; 1:33–39
16 Maree AO, Curtin RJ, Chubb A, et al.: Cyclooxygenase-1
problems. These novel agents would also be highly speci-
haplotype modulates platelet response to aspirin. J Thromb
fic for pathogen-mediated platelet activation and thus
Haemost, 2005; 3:2340–2345
likely more effective than aspirin/clopidogrel. Thus, drugs 17 Bajrangee A, Ryan N, Vangjeli C, et al.: Impact of genetic
that target the immune function of platelets are most variation in the 5-HT transporter and receptor on platelet
likely to be cost-effective with a good safety profile. function in patients with stable CAD taking aspirin. Thromb
Res, 2016; 146:51–55
18 Goodman T, Ferro A, Sharma P: Pharmacogenetics of
Conflict of interests aspirin resistance: a comprehensive systematic review. Br J
The author declare no conflict of interests. Clin Pharmacol, 2008; 66:222–232
19 Peace A, McCall M, Tedesco T, et al.: The role of weight and
enteric coating on aspirin response in cardiovascular
References patients. J Thromb Haemost, 2010; 8:2323–2325
1 Fitzgerald DJ, Roy L, Catella F, et al.: Platelet activation in 20 Maree AO, Curtin RJ, Dooley M, et al.: Platelet response to
unstable coronary disease. N Engl J Med, 1986; 315:983– low-dose enteric-coated aspirin in patients with stable car-
989 diovascular disease. J Am Coll Cardiol, 2005; 46:1258–1263
2 Cairns JA, Gent M, Singer J, et al.: Aspirin, sulfinpyrazone, 21 Cox D, Maree AO, Dooley M, et al.: Effect of enteric coating
or both in unstable angina. N Engl J Med, 1985; 313:1369– on antiplatelet activity of low-dose aspirin in healthy volun-
1375 teers. Stroke, 2006; 37:2153–2158
3 Weiss HJ, Aledort LM, Kochwa S: The effect of salicylates 22 Grosser T, Maree AO, Dooley M, et al.: Drug resistance and
on the hemostatic properties of platelets in man. J Clin pseudoresistance. Circulation, 2013; 127:377–385
Investig, 1968; 47:2169–2180 23 Cox D, Fitzgerald DJ: Lack of bioequivalence among low-
4 Vane JR, Botting RM: The mechanism of action of aspirin. dose, enteric-coated aspirin preparations. Clin Pharmacol
Thromb Res, 2003; 6:255 Ther, 2018; 103:1047–1051
5 Stalker TJ, Ma P, Wannemacher KM, et al.: Platelet Signal- 24 McCall M, Peace A, Tedesco A, et al.: Weight as an assay-
ing, in Gresele P, Born GVR, Patrono C, Page CP (eds): Anti- independent predictor of poor response to enteric aspirin in
platelet Agents. Berlin, Heidelberg, Springer Berlin cardiovascular patients. Platelets, 2019. In press. https://doi.
Heidelberg. 2012:59–85. org/10.1080/09537104.2019.1667495
6 Puri RN: Phospholipase A2: its role in ADP- and thrombin- 25 Catella-Lawson F, Reilly MP, Kapoor SC, et al.: Cyclooxyge-
induced platelet activation mechanisms. Int J Biochem Cell nase Inhibitors and the Antiplatelet Effects of Aspirin. N
Biol, 1998; 30:1107–1122 Engl J Med, 2001; 345:1809–1817
7 Fuster V, Sweeny JM: Aspirin: a historical and contempo- 26 Tacconelli S, Dovizio M, Di Francesco L, et al.: Reduced
rary therapeutic overview. Circulation, 2011; 123:768–778 variability to aspirin antiplatelet effect by the coadministra-
8 Antithrombotic Trialists C: Collaborative meta-analysis of tion of statins in high-risk patients for cardiovascular dis-
randomised trials of antiplatelet therapy for prevention of ease. Clin Pharmacol Ther, 2018; 104:111–119
death, myocardial infarction, and stroke in high risk 27 Fontana P, Zufferey A, Daali Y, et al.: Antiplatelet therapy:
patients. BMJ, 2002; 324:71–86 targeting the TxA2 pathway. Cardiovasc Transl Res, 2014;
9 Patrono C: Aspirin resistance: definition, mechanisms and 7:29–38
clinical read-outs. J Thromb Haemost, 2003; 1:1710–1713 28 Marguerie GA, Thomas-Maison N, Larrieu MJ, et al.: The
10 Li J, Song M, Jian Z, et al.: Laboratory aspirin resistance interaction of fibrinogen with human platelets in a plasma
and the risk of major adverse cardiovascular events in milieu. Blood, 1982; 59:91–95

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
Future of anti-platelet agents 9

29 Nachman RL, Leung LL: Complex formation of platelet 46 Dermot C: Oral GPIIb/IIIa antagonists: what went wrong?
membrane glycoproteins IIb and IIIa with fibrinogen. J Clin Curr Pharm Des, 2004; 10:1587–1596
Investig, 1982; 69:263–269 47 Cox D, Smith R, Quinn M, et al.: Evidence of platelet activa-
30 Hawiger J, Kloczewiak M, Bednarek MA, et al.: Platelet tion during treatment with a GPIIb/IIIa antagonist in
receptor recognition domains on the alpha chain of human patients presenting with acute coronary syndromes. J Am
fibrinogen: structure-function analysis. Biochemistry, 1989; Coll Cardiol, 2000; 36:1514–1519
28:2909–2914 48 Armstrong PC, Peter K: GPIIb/IIIa inhibitors: from bench to
31 Investigators E: Use of a monoclonal antibody directed bedside and back to bench again. Thromb Haemost, 2012;
against the platelet glycoprotein IIb/IIIa receptor in high-risk 107:808–814
coronary angioplasty. N Engl J Med, 1994; 330:956–961 49 Shen B, Delaney MK, Du X: Inside-out, outside-in, and
32 Huang TF, Hsu CC, Kuo YJ: Anti-thrombotic agents derived inside-outside-in: G protein signaling in integrin-mediated
from snake venom proteins. Thromb J, 2016; 14(Suppl 1):18 cell adhesion, spreading, and retraction. Curr Opin Cell Biol,
33 Tcheng JE, Harrington RA, Kottke-Marchant K, et al.: Multi- 2012; 24:600–606
center, randomized, double-blind, placebo-controlled trial of 50 Defreyn G, Bernat A, Delebassee D, et al.: Pharmacology of
the platelet integrin glycoprotein IIb/IIIa blocker integrelin ticlopidine: a review. Semin Thromb Hemost, 1989; 15:159–
in elective coronary intervention. Circulation, 1995; 166
91:2151–2157 51 Hass WK, Easton JD, Adams HP Jr, et al.: A randomized
34 Platelet Receptor Inhibition in Ischemic Syndrome Manage- trial comparing ticlopidine hydrochloride with aspirin for
ment Study I: A comparison of aspirin plus tirofiban with the prevention of stroke in high-risk patients. N Engl J Med,
aspirin plus heparin for unstable angina. N Engl J Med, 1989; 321:501–507
1998; 338:1498–1505 52 Gent M, Blakely JA, Easton JD, et al.: The Canadian Ameri-
35 Boersma E, Harrington RA, Moliterno DJ, et al.: Platelet gly- can Ticlopidine Study (Cats) in thromboembolic stroke. Lan-
coprotein IIb/IIIa inhibitors in acute coronary syndromes: a cet, 1989; 333:1215–1220
meta-analysis of all major randomised clinical trials. Lancet, 53 Savi P, Herbert J-M: Clopidogrel and ticlopidine: P2Y12
2002; 359:189–198 adenosine diphosphate-receptor antagonists for the preven-
36 Ciccone A, Motto C, Abraha I, et al.: Glycoprotein IIb-IIIa tion of atherothrombosis. Semin Thromb Hemost, 2005;
inhibitors for acute ischaemic stroke. Cochrane Database 31:174–183
Syst Rev, 2014: CD005208 54 Wijeyeratne YD, Heptinstall S: Anti-platelet therapy: ADP
37 Pasala T, Sattayaprasert P, Bhat PK, et al.: Clinical and eco- receptor antagonists. Br J Clin Pharmacol, 2011; 72:647–
nomic studies of eptifibatide in coronary stenting. Ther Clin 657
Risk Manag, 2014; 10:603–614 55 Koupenova M, Ravid K: Biology of platelet purinergic recep-
38 Anders R, Kleiman J, Nicholson N, et al.: Xemilofiban/Orbo- tors and implications for platelet heterogeneity. Front Phar-
fiban: insight into drug development. Cardiovasc Drug Rev, macol, 2018; 9:37
2001; 19:116–132 56 CAPRIE Steering Committee: A randomised, blinded, trial of
39 Modi NB, Novotny W, Reimann JD, et al.: Pharmacokinetics clopidogrel versus aspirin in patients at risk of ischaemic
and pharmacodynamics of sibrafiban, an orally administered events (CAPRIE). Lancet, 1996; 348:1329–1339
IIb/IIIa antagonist, in patients with acute coronary syn- 57 Mehta SR, Yusuf S, Peters RJ, et al.: Effects of pretreatment
drome. J Clin Pharmacol, 1999; 39:675–684 with clopidogrel and aspirin followed by long-term therapy
40 Cox D, Motoyama Y, Seki J, et al.: Pentamidine: a non-pep- in patients undergoing percutaneous coronary intervention:
tide GPIIb/IIIa antagonist--in vitro studies on platelets from the PCI-CURE study. Lancet, 2001; 358:527–533
humans and other species. Thromb Haemost, 1992; 68:731– 58 Yuan J, Xu GM, Ding J: Aspirin versus clopidogrel
736 monotherapy for the treatment of patients with stable coro-
41 Cox D, Motoyama Y, Seki J, et al.: Pentamidine is a specific, nary artery disease: a systematic review and meta-analysis.
non-peptide, GPIIb/IIIa antagonist. Thromb Haemost, 1996; Adv Ther, 2019; 36:2062–2071
75:503–509 59 Warlo EMK, Arnesen H, Seljeflot I: A brief review on resis-
42 Aoki T, Cox D, Senzaki K, et al.: The anti-platelet and anti- tance to P2Y12 receptor antagonism in coronary artery dis-
thrombotic effects of FK633, a peptide-mimetic GPIIb IIIa ease. Thromb J, 2019; 17:11
antagonist. Thromb Res, 1996; 81:439–450 60 Niitsu Y, Jakubowski JA, Sugidachi A, et al.: Pharmacology
43 Cox D, Seki J: Characterization of the binding of FK633 to of CS-747 (prasugrel, LY640315), a novel, potent antiplatelet
the platelet fibrinogen receptor. Thromb Res, 1998; 91:129– agent with in vivo P2Y12 receptor antagonist activity. Semin
136 Thromb Hemost, 2005; 31:184–194
44 Chew DP, Bhatt DL, Sapp S, et al.: Increased mortality with 61 Nagalla S, Sarode R: Role of platelet transfusion in the
oral platelet glycoprotein IIb/IIIa antagonists. Circulation, reversal of anti-platelet therapy. Transfus Med Rev, 2019;
2001; 103:201–206 33:92–97
45 Cox D, Brennan M, Moran N: Integrins as therapeutic tar- 62 Gioel Gabrio S, Parisi R, Mirabella F, et al.: P2Y12 inhibi-
gets: lessons and opportunities. Nat Rev Drug Discovery, tors: pharmacologic mechanism and clinical relevance. Car-
2010; 9:804 diovasc Hematol Agents Med Chem, 2013; 11:101–105

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
10 D. Cox

63 Franchi F, Rollini F, Aggarwal N, et al.: Pharmacodynamic 81 Ruggeri ZM, Mendolicchio GL: Interaction of von Wille-
comparison of prasugrel versus ticagrelor in patients with brand factor with platelets and the vessel wall. Hamostase-
type 2 diabetes mellitus and coronary artery disease. Circu- ologie, 2015; 35:211–224
lation, 2016; 134:780–792 82 Momi S, Tantucci M, Van Roy M, et al.: Reperfusion of
64 Alexandros B, Telila T, Palla M, et al.: P2Y12 receptor cerebral artery thrombosis by the GPIb–VWF blockade with
antagonists: which one to choose? A systematic review and the Nanobody ALX-0081 reduces brain infarct size in gui-
meta-analysis Curr Pharm Des, 2016; 22:4568–4576 nea pigs. Blood, 2013; 121:5088–5097
65 Waite LH, Phan YL, Spinler SA: Cangrelor: a novel intra- 83 Gresele P, Momi S: Inhibitors of the interaction between
venous antiplatelet agent with a questionable future. Phar- von Willebrand factor and platelet GPIb/IX/V. Handb Exp
macotherapy, 2014; 34:1061–1076 Pharmacol, 2012; 210:287–309
66 Raslan Z, Naseem KM: The control of blood platelets by 84 Diz-K€ €kkaya R: Inherited platelet disorders including
ußcu
cAMP signalling. Biochem Soc Trans, 2014; 42:289–294 Glanzmann thrombasthenia and Bernard-Soulier syndrome.
67 Makhoul S, Walter E, Pagel O, et al.: Effects of the NO/sol- ASH Educ Program Book 2013; 2013:268–275
uble guanylate cyclase/cGMP system on the functions of 85 Cavallari LH, Franchi F, Rollini F, et al.: Clinical implemen-
human platelets. Nitric Oxide, 2018; 76:71–80 tation of rapid CYP2C19 genotyping to guide antiplatelet
68 Schr€or K: The pharmacology of cilostazol. Diabetes Obes therapy after percutaneous coronary intervention. J Transl
Metab, 2002; 4:S14–S19 Med, 2018; 16:92
69 Rogers KC, Oliphant CS, Finks SW: Clinical efficacy and 86 Cox D, Kerrigan SW, Watson SP: Platelets and the innate
safety of cilostazol: a critical review of the literature. Drugs, immune system: mechanisms of bacterial-induced platelet
2015; 75:377–395 activation. J Thromb Haemost, 2011; 9:1097–1107
70 Gresele P, Momi S, Falcinelli E: Anti-platelet therapy: phos- 87 Rayes J, Watson SP, Nieswandt B: Functional significance
phodiesterase inhibitors. Br J Clin Pharmacol, 2011; of the platelet immune receptors GPVI and CLEC-2. J Clin
72:634–646 Invest, 2019; 129:12–23
71 Adams MN, Ramachandran R, Yau MK, et al.: Structure, 88 Semple JW, Italiano JE, Freedman J: Platelets and the
function and pathophysiology of protease activated recep- immune continuum. Nat Rev Immunol, 2011; 11:264–274
tors. Pharmacol Ther, 2011; 130:248–282 89 Kapur R, Zufferey A, Boilard E, et al.: Nouvelle cuisine: pla-
72 Tricoci P: Protease-activated receptor-1 antagonists post- telets served with inflammation. J Immunol, 2015;
percutaneous coronary intervention. Interv Cardiol Clin, 194:5579–5587
2017; 6:57–66 90 Clark SR, Ma AC, Tavener SA, et al.: Platelet TLR4 activates
73 Bharti B, Bharati M: Vorapaxar, a Protease-Activated Recep- neutrophil extracellular traps to ensnare bacteria in septic
tor-1 Antagonist, a Double –Edged Sword!. Recent Adv Car- blood. Nat Med, 2007; 13:463–469
diovasc Drug Discov (Discontinued), 2014; 9:73–77 91 Yeaman MR: Platelets: at the nexus of antimicrobial
74 Al-Khafaji K, Mutyala M, Al-Khafaji N, et al.: Protease-acti- defence. Nat Rev Micro, 2014; 12:426–437
vated receptor 1 inhibitors: novel antiplatelet drugs in pre- 92 Cloutier N, Allaeys I, Marcoux G, et al.: Platelets release
vention of atherothrombosis. Am J Ther, 2017; 24:e730– pathogenic serotonin and return to circulation after immune
e736 complex-mediated sequestration. Proc Natl Acad Sci, 2018;
75 Rwibasira Rudinga G, Khan GJ, Kong Y: Protease-activated 115:E1550–E1559
receptor 4 (PAR4): a promising target for antiplatelet ther- 93 Du F, Jiang P, He S, et al.: Antiplatelet therapy for critically
apy. Int J Mol Sci, 2018; 19:573 ill patients: a pairwise and bayesian network meta-analysis.
76 Xiang R-W, Han RB, Yang JY, et al.: Comparative efficacy Shock, 2018; 49:616–624
and safety of multiple antiplatelet therapies for secondary 94 Trauer J, Muhi S, McBryde ES, et al.: Quantifying the effects
prevention of ischemic stroke or transient ischemic attack: a of prior Acetyl-Salicylic acid on sepsis-related deaths: an
network meta-analysis. J Neurol Sci, 2019; 399:199–206 individual patient data meta-analysis using propensity
77 Fanaroff AC, Hasselblad V, Roe MT, et al.: Antithrombotic matching. Crit Care Med, 2017; 45:1871–1879
agents for secondary prevention after acute coronary syn- 95 Zapata JC, Cox D, Salvato MS: The role of platelets in the
dromes: a systematic review and network meta-analysis. Int pathogenesis of viral hemorrhagic fevers. PLoS Negl Trop
J Cardiol, 2017; 241:87–96 Dis, 2014; 8:e2858
78 McFadyen JD, Schaff M, Peter K: Current and future anti- 96 Alonso AL, Cox D: Platelet interactions with viruses and
platelet therapies: emphasis on preserving haemostasis. Nat parasites. Platelets, 2015; 26:317–323
Rev Cardiol, 2018; 15:181 97 Fitzgerald JR, Foster TJ, Cox D: The interaction of bacterial
79 Hanjaya-Putra D, Haller C, Wang X, , et al.: Platelet-tar- pathogens with platelets. Nat Rev Microbiol, 2006; 4:445–
geted dual pathway antithrombotic inhibits thrombosis with 457
preserved hemostasis. JCI Insight, 2018; 3:e99329. https://d 98 O’Brien L, Kerrigan SW, Kaw G, et al.: Multiple mechanisms
oi.org/10.1172/jci.insight.99329 for the activation of human platelet aggregation by Staphy-
80 Plow EF, Wang Y, Simon DI: The search for new antithrom- lococcus aureus: roles for the clumping factors ClfA and
botic mechanisms and therapies that may spare hemostasis. ClfB, the serine-aspartate repeat protein SdrE and protein A.
Blood, 2018; 131:1899–1902 Mol Microbiol, 2002; 44:1033–1044

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11
Future of anti-platelet agents 11

99 Kerrigan SW, Douglas I, Wray A, et al.: A role for glycopro- proinflammatory responses. Ann Rheum Dis, 2019; 78:228–
tein Ib in Streptococcus sanguis-induced platelet aggrega- 237
tion. Blood, 2002; 100:509–516 104 Pietersz GA, Mottram PL, van de Velde NC, et al.: Inhibition
100 Kerrigan SW, Jakubovics NS, Keane C, et al.: Role of Strep- of destructive autoimmune arthritis in FccRIIa transgenic
tococcus gordonii surface proteins SspA/SspB and Hsa in mice by small chemical entities. Immunol Cell Biol, 2009;
platelet function. Infect Immun, 2007; 75:5740–5747 87:3–12
101 Moriarty RD, Cox A, McCall M, et al.: Escherichia coli 105 Tomo S, Mohan S, Ramachandrappa VS, et al.: Dynamic
induces platelet aggregation in an FcgammaRIIa-dependent modulation of DC-SIGN and FcΥR2A receptors expression
manner. J Thromb Haemost, 2016; 14:797–806 on platelets in dengue. PLoS ONE, 2018; 13:e0206346
102 Byrne MF, Kerrigan SW, Corcoran PA, et al.: Helicobacter 106 Sung P-S, Huang T-F, Hsieh S-L: Extracellular vesicles from
pylori binds von Willebrand factor and interacts with GPIb CLEC2-activated platelets enhance dengue virus-induced
to induce platelet aggregation. Gastroenterology, 2003; lethality via CLEC5A/TLR2. Nat Commun, 2019; 10:2402
124:1846–1854 107 Zheng SL, Roddick AJ: Association of aspirin use for pri-
103 Chen B, Vousden KA, Naiman B, et al.: Humanised effector- mary prevention with cardiovascular events and bleeding
null FccRIIA antibody inhibits immune complex-mediated events. JAMA, 2019; 321:277–287

© 2019 International Society of Blood Transfusion, ISBT Science Series (2019) 0, 1–11

You might also like