Sei sulla pagina 1di 8

Thrombosis Research 133 (2014) 131–138

Contents lists available at ScienceDirect

Thrombosis Research
journal homepage: www.elsevier.com/locate/thromres

Mini Review

Use of antiplatelet agents in sepsis: A glimpse into the future


Karolina Akinosoglou a,⁎, Dimitrios Alexopoulos b
a
Department of Internal Medicine, University General Hospital of Patras, 26504, Rio, Greece
b
Department of Cardiology, University General Hospital of Patras, 26504, Rio, Greece

a r t i c l e i n f o a b s t r a c t

Article history: As mechanisms of sepsis pathophysiology have been elucidated with time, sepsis may be considered nowadays,
Received 7 June 2013 as an uncontrolled inflammatory and pro-coagulant response to a pathogen. In this cascade of events, platelets
Received in revised form 4 July 2013 play a key role, via interaction with endothelial cells and modulation of both innate and adaptive immune
Accepted 8 July 2013
system. In that manner, inhibition of platelet function could represent a useful tool for attenuating inflammatory
Available online 6 October 2013
response and improving outcomes. Data on current antiplatelet agents, including acetylsalicylic acid, P2Y12
Keywords:
inhibitors and GPIIb/IIIa antagonists, in animal models are promising. Clinical data in patients hospitalized for
Antiplatelet agents pneumonia, at risk for acute lung injury, and/or critically ill revealed an association between antiplatelet therapy
Platelets and reduction in both short-term mortality and prevalence of acute lung injury, as well as, the need for intensive
P2Y12 inhibitors care unit admission, without a concomitant increased bleeding risk. In need of innovative approach in the treat-
Sepsis ment of sepsis, further prospective, interventional, randomized trials are pivotal to establish potential use of
GPIIb/IIIa antagonists antiplatelet agents in this context.
© 2013 Elsevier Ltd. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 131
Role of Platelets in Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132
Antiplatelet Agents in Sepsis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132
Acetylsalicylic Acid (ASA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 132
P2Y12 Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
GPIIb/IIIa Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Issues to be Considered . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Conflict of Interest Statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136

Introduction mortality rate [1,2]. As a result, an estimated economic burden of


nearly 17 billion dollars annually in the USA, has currently been at-
Sepsis is a prevalent syndrome that carries significant morbidity tributed to sepsis [1].
and mortality. Almost 24% of patients with an admission diagnosis Activation of blood platelets is a typical finding in patients with sys-
of sepsis will progress to severe sepsis with an associated 35–50% temic inflammation and sepsis, while thrombocytopenia in critically ill

Abbreviations: ADAMTS-13, A Disintegrin And Metalloproteinase with a ThromboSpondin type 1 motif, member 13; ADP, Adenosine Diphosphate; ATP, Adenosine Triphosphate; ALI,
Acute Lung Injury; ARDS, Acute Respiratory Distress Syndrome; ASA, AcetylSaliculic Acid; ATL, 15-epi-lipoxin A4; CABG, Coronary Artery Bypass Grafting; CAP, Community Acquired
Pneumonia; COX, CyclOXygenase; CRP, C-Reactive Protein; C4b, complement component 4b; DIC, Disseminated Intravascular Coagulation; eNOs, endogenous nitric oxide synthase;
Erk1, extra cellular signal kinase 1; GP, GlycoProtein; ICU, Intensive Care Unit; IL-#, InterLeukine #; IL-10r, InterLeukine 10 receptor; iNOs, inducible nitric oxide synthase; LPS,
LipoPolySacccharide; Ltb4r, LeukoTriene b4 Receptor; MIP-1a, Macrophage inflammatory protein-1alpha; MOF, Multiple Organ Failure; NFκB, nuclear factor kappa-light-chain-enhancer
of activated B cells; NET, Neutrophil Extracellular Trap; NO, Nitrogen Oxide; PAF, Platelet Activating Factor; PLATO, PLATelet inhibition and clinical Outcomes; PMN, PolyMorphoNuclear
cells; PSGL-1, P-Selectin Glycoprotein Ligand-1; RANTES, Regulated on Activation, Normal T cell Expressed and Secreted; ROS, Reactive Oxygen Species; SIRS, Systemic Inflammatory Re-
sponse Syndrome; TCR, T-Cell Receptor; TF, Tissue Factor; TLR, Toll-Like Receptor; TNF alpha, Tumor Necrosis Factor alpha; vWF, von Willebrand Factor.
⁎ Corresponding author at: Dept of Internal Medicine, University Hospital of Patras, 26504, Rio, Greece. Tel.: +30 2610999583.
E-mail address: akin@upatras.gr (K. Akinosoglou).

0049-3848/$ – see front matter © 2013 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.thromres.2013.07.002
132 K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138

patients is associated with poor outcome [3–5]. It has been well Furthermore, platelets express toll-like receptors (TLRs), pathogen
established that platelets significantly contribute to organ failure via recognition receptors involved in activation of innate immunity, in-
their haemostatic and thrombotic potential, resulting in thrombotic cluding TLR2 and TLR4 that recognize the common bacterial mole-
microangiopathy and disseminated intravascular coagulation. What is cules peptidoglycan and lipopolysaccharide (LPS), respectively [11].
less known however, is their involvement in the adaptive immune Interactions between platelet TLR4 and LPS may contribute in throm-
response [6] as well as, their interaction with microorganisms [7]. A bocytopenia in sepsis and pulmonary fibrin deposition [11]. Activated
number of agents including corticosteroids, tumour necrosis factor platelets, particularly in the context of LPS stimulation, trigger the re-
(TNF)-alpha antagonists and interleukin 1 (IL-1) receptor antagonists, lease of extracellular DNA traps (NETs) with proteolytic activity from
have been suggested to potentially block the inflammatory cascade, neutrophils, serving to capture and degrade microbes [11]. Thus,
prevent severe sepsis and ensure better outcomes, [8]. Although data immune-mediated effects of platelets may be important for the host
from phase II clinical trials showed promise, larger studies have not defence.
demonstrated any significant improvement in clinical outcomes [8]. At the same time, platelet activation exacerbates the release of more
Antiplatelet drugs including acetylsalicylic acid (ASA), and P2Y12 than 35 distinct, active compounds that are capable of modulating not
inhibitors are widely used in patients with cardiovascular disease for only their own function, but also that of cells around them [4,6,11].
the secondary prevention of atherothrombotic events [9]. Several stud- Platelet alpha granules contain chemokines and other soluble media-
ies have shown that ASA and clopidogrel not only diminish the risk of tors of inflammation such as IL-1b, regulated on activation normal T
atherothrombotic events, but also reduce markers of systemic inflam- cell expressed and secreted protein (RANTES), macrophage inflamma-
mation, including C-reactive protein (CRP), soluble CD62P (P-selectin) tory protein (MIP-1 alpha), monocyte chemo-attractant protein, thy-
and CD54, pro-inflammatory cytokines, and platelet-leukocyte conju- mus and activation-regulated chemokine, and antimicrobial defensins
gates [10]. It is assumed that the anti-inflammatory effects of antiplatelet (thrombocidins) involved in coagulation pathways or stimulating
drugs are mediated by an inhibition of platelet activation [10]. Thus, further platelet recruitment and attracting neutrophils and leukocytes,
derived by the need to extend current pharmacological treatment which are key players in mediating ongoing inflammatory responses
options, the question arises, whether drugs that inhibit platelet activa- such as sepsis [4,6,11]. Among them, the potent serine protease
tion, such as ASA or P2Y12 inhibitors, may have a benefit in critically granzyme B is expressed, resulting in cytotoxic platelets mediating
ill patients. apoptosis in lung and spleen tissue [17]. Fig. 1 summarizes current
This review will highlight current data on potential use of antiplatelet examples of activated platelets mediating immune responses.
agents in managing sepsis. However, the effects of certain bacterial products on platelet func-
tion have not been found to be consistent and may vary according to
species, timing of the study, location of platelets in the circulation, and
Role of Platelets in Sepsis pathogenesis of sepsis [18–20]. For example, LPS has been shown to
increase platelet aggregation in various animal models [21–23], yet bac-
There is substantial evidence that blood platelets play an important terial products seem to decrease human platelet aggregation in vitro
role in the development of multiple organ failure (MOF) in septic [24,25]. In a small group of 74 patients with sepsis and severe sepsis
patients [4,5]. During infection and systemic inflammation, platelets Mavromattis et al. [26] showed that platelet aggregation increased
become activated, as reflected by an increase in the number of CD62P- somewhat in patients with an uneventful course but decreased over
positive platelets and platelet-leukocyte conjugates [3,5]. Different mech- time in patients who developed severe sepsis. Similarly, in a series of
anisms have been associated with platelet activation, including imbalance 26 patients Boldt et al. noted decreased platelet aggregation in critically
between plasma level of high molecular weight von-Willebrand factor ill patients, as later confirmed by others at a molecular level [18,27].
and - its cleavage protease - a disintegrin and metalloproteinase with a However, Gawaz et al. [19] described increased adhesion and activation
thrombospondin type 1 motif, member 13 (ADAMTS-13), and binding of platelets, even in patients in an intensive care unit (ICU).
of endotoxins to specific receptors at the platelet surface [5,11]. Once
activated, the platelet alters from its normal disc-like shape to a more Antiplatelet Agents in Sepsis
spherical appearance with arm-like extensions, that facilitate adhesion
to the endothelium and to other cells [11]. Platelet/endothelium interac- Acetylsalicylic Acid (ASA)
tion plays a central role in inflammatory mechanisms within the vessel
wall [12]. It has been reported that activated platelets - as occurs in Endothelial dysfunction is one of the early signs of a systemic in-
endotoxemia - aggregate and can bind to endothelial cells, despite flammatory reaction and is believed to be one of the triggers of organ
the presence of intact endothelium [11,13,14]. Platelet adhesion to failure in sepsis [15]. ASA stimulates the synthesis of 15-epi-lipoxin
endothelium enhances the procoagulatory activity of endothelial cells, A4 (ATL), which in turn increases nitric oxide synthesis through endo-
subsequently impairing microcirculation and, thus, leading to hypoper- thelial and inducible nitric oxide synthase [28]. Nitric oxide inhibits
fusion and organ dysfunction [11,15]. the interactions between leucocytes and endothelial cells, resulting in
Platelet activation also causes the expression of cell surface receptors decreased polymorph neutrophil recruitment [28] (Fig. 2). Contrary to
and release of molecules that can amplify the immune response [11]. findings in models of endotoxemia [29,30], low doses of aspirin seem
Namely, the initial interactions are driven by P-selectin (CD62P) on to inhibit platelet activation and have anti-inflammatory effects in
the surface of activated platelets, which is recognised by P-selectin gly- patients with coronary heart disease. The latter include decrease in
coprotein ligand-1 (PSGL-1) on leukocytes [3,5,11,16]. Functional cross the expression of pro-inflammatory mediators including TNF, IL-6, TF,
talk between the P-selectin-PSGL-1 pair and the leukocyte integrin or up-regulation of NFkB, a central transcription factor in inflammation
αMβ2 (Mac-1) orchestrates the molecular events necessary for leuko- and cell death regulation.[10] (Fig. 2).
cyte recruitment, via platelet activation and the subsequent vascular Several studies evaluating aspirin for the treatment of sepsis in
response to injury [6,11,16]. Adhesion of platelets to monocytes results animal models, have suggested a potential benefit [31,32]. Recently,
in signalling dependent transcriptional regulation, which leads to Derhaschnig et al. [33] evaluated the effects of aspirin and a
expression of important mediators of inflammation and pro-coagulant nitroderivative of aspirin (NCX-4016) on platelet function in healthy
tissue factor (TF) [11]. Platelet surface expression of CD40L (CD154) volunteers after infusion of endotoxin. NCX-4016 acts through COX in-
and its subsequent binding to the CD40 receptor may also be involved hibition and nitric oxide release, thus, reducing thrombin induced
in immune cell recruitment, chemokine production, IgM to IgG isotype platelet activation more effectively than ASA [34,35]. Interestingly,
switching, and dendritic cell maturation [11]. while NCX-4016 had virtually no effect on platelet function, aspirin
K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138 133

TCR

Platelets promote the activation of MHC


PAF
thrombin Gram (-) monocytes & dendritic cells through
T cell
bacteria CD40–CD154 interactions
DC
ADAMTS-13
vWF αiib/β3
DC activation increases antigen
presentation to T cells

Activated
LPS Platelets produce cytokines
platelet
& chemokines & promote
Circulating
neutrophil activation
platelets TLR4
PAF
RANTES
thrombocidins
CD154
IL-1b
TL4 expressing platelets PMN
present LPS to neutrophils
CD40
and promote their activation MIP-1a

P
Platelets mediate WBC rolling,
adhesion, arrest in the flowing
blood, & emigration
Platelets promote neutophil Platelet expressed
formation of NETs which selectins promote
trap free bacteria ROS
neutrophil tethering

Activated neutrophil PSGL-1


generate ROS that damage
the endothelium
Mac-1 Selectin
Sel

GPIba
Endothelial cell Damaged endothelium
Fibrin

Fig. 1. Activated platelets can mediate cell–cell interactions and affect innate immune responses. Platelet activation by thrombin, collagen, ADP, platelet-activating factor (PAF) and
other agonists including LPS leads to platelet–platelet aggregation. A variety of factors with inflammatory and adaptive immune functions, are released and/or surface displayed,
promoting among others heterotypic aggregates. Examples of how activated platelets can mediate cell–cell interactions and affect innate immune responses include a) Following
binding to LPS, platelets may directly kill the bacteria by producing thrombocidins or by aggregating around them and directing neutrophils to extrude their DNA. Neutrophil
extracellular traps (NETs) are formed, that stretch downstream and finally eliminate bacteria, b) activated platelets promote neutrophil tethering and activation through the
expression of selectins, CD154 (also known as CD40L), secretion of inflammatory cytokines and chemokines, c) platelets can also promote the activation of monocytes and dendritic
cells (DCs), particularly through CD40–CD154 interactions. This leads to increased antigen presentation to T cells and enhances adaptive immune responses. ADAMTS-13: A
Disintegrin And Metalloproteinase with a ThromboSpondin type 1 motif, member 13; PAF:Platelet Activating Factor; RANTES: Regulated on Activation, Normal T cell Expressed
and Secreted; MIP-1a: Macrophage inflammatory protein-1a; TCR:T-Cell Receptor; PMN:PolyMorphoNuclear cells; ROS:Reactive Oxygen Species; vWF:von Willebrand Factor;
PSGL-1: P-Selectin Glycoprotein Ligand-1; TLR: Toll-Like Receptor.

appeared to have a protective effect against the formation of endotoxin- syndrome (ARDS) [44–46] even though, in many studies no convincing
induced platelet plugs, as compared with placebo. Observational studies evidence has been reported regarding the impact of platelet inhibition
evaluating the effects of chronic aspirin use on critically ill patients have on ICU or hospital mortality or ICU- or hospital-free days [41,42,47]. In
also reported encouraging results (Table 1). Favourable differences other reports, aspirin use alone had no statistically significant associa-
were noted in patients treated with antiplatelet agents (aspirin or a tion with death, diagnosis of ALI/ARDS, or diagnosis of severe sepsis.
thienopyridine) prior to critical illness, including an apparent reduction However, prehospital use of aspirin co-administered with statins was
in organ dysfunction [36]. This finding comes in line with previous data associated with favourable outcomes [48]. Of note, ASA administration
exhibiting that prior treatment with antiplatelet agents, including aspi- was not associated with increased bleeding events or increased need
rin, was associated with shorter length of stay and fewer ICU admissions for transfusion even in patients with increased bleeding risk e.g. neuro-
[37]. Even following ICU admission, it appears that patients who were surgery patients [36,38,42].
administered low-dose ASA at the time of the development of systemic
inflammatory response syndrome (SIRS) and sepsis, showed a signifi-
cant difference in mortality compared to those not using ASA in this P2Y12 Inhibitors
period [38–40]. Furthermore, in critically ill patients, antiplatelet thera-
py (95% with ASA) before hospitalization significantly reduced develop- The thienopyridines target the P2Y12 subclass of adenosine di-
ment of acute lung injury (ALI) (12.7% vs 28.0%) [41,42] and need for phosphate (ADP) receptors and thereby blunt platelet activation and
mechanical ventilation [42], as well as, mortality associated with septic aggregation stimulated by ADP [49]. Stimulation of P2Y12 receptors
shock (by ~ 45%) [43]. This finding comes in agreement with preclinical promotes further release of the platelet agonist ADP from dense gran-
studies, suggesting that, platelet activation and sequestration play a ules, is essential for ADP-mediated complete activation of glycoprotein
critical role in the pathogenesis of ALI/acute respiratory distress IIb/IIIa and Ia/IIa, and further stabilizes platelet aggregates[50]. The
134 K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138

prasugrel
clopidogrel
ticagrelor
cangrelor 2

P2Y12
receptor

1 IL13, IL10r, C4b,


Activated Ltb4r, Erk1genes
Abciximab ADP 5
Eptifibatide Platelet
Tirofiban δ-granules
cox
NFκB
αiib/β3 TxA2 gene
TxA2
receptor
7

Fibrinogen IL-6
3 TNF-a
4 Selectin
Activated Platelet
Lung & Spleen Cells
6 Leukocyte CD40

CD154

8
ROS

NO 9
granzyme B
10
eNOs Endothelial cell
iNOs

Fig. 2. Different antiplatelet agents interfere with a variety of pathways mediating sepsis. (1,2) Amplification of platelet activation response is hampered by ASA and P2Y12 inhib-
itors through blockade of thromboxane (TxA2) production and ADP binding to P2Y12 receptors respectively. (3,4) Platelet aggregation, hence microcirculation impairment during
sepsis, is attenuated following blockade of ADP mediated GPIIb/IIIa (αiib/β3) receptor maturation by P2Y12 inhibitors and prevention of aggregate formation by GPIIb/IIIa antag-
onist administration. (5,6) ASA and clopidogrel down regulate expression of pro inflammatory mediators i.e. cytokines, complement factors, etc probably through modulation of
certain transcription factors e.g. NFκB. (7) ASA and P2Y12 inhibitors alter platelet-leukocyte interactions through decreased expression of cell ligands e.g. P-selectin, CD40,
CD154. (8) P2Y12 inhibitors attenuate endothelial damage caused by neutrophil ROS production. (9) ASA promotes anti-adhesive NO production preventing leukocyte tethering,
adhesion and emigration. (10) GPIIb/IIIa antagonists have been found to decrease platelet mediated cytotoxicity caused by granzyme-B secretion. ASA: acetylsalicylic acid; COX:
CyclOXygenase; IL-13: InterLeukine 13; IL-10r: InterLeukine 10 receptor; C4b: complement component 4b; Erk1: extra cellular signal kinase 1; NFκB: nuclear factor
kappa-light-chain-enhancer of activated B cells; ROS:Reactive Oxygen Species; NO: Nitrogen Oxide; IL-6: InterLeukine 6; TNF-a: Tumour Necrosis Factor a; iNOs: inducible nitric
oxide synthase; eNOs: endogenous nitric oxide synthase; Ltb4r: LeukoTriene b4 Receptor.

thienopyridines ticlopidine, clopidogrel and prasugrel irreversibly experiments, a tendency of a higher survival rate in the clopidogrel-
inhibit the P2Y12 receptor and are rather specific for platelets [49]. treated animals was noted [37]. Clopidogrel appears to modulate gene
Clopidogrel, the most widely used thienopyridine, may attenuate expression in peripheral blood cells reflecting the transcriptional activity
platelet expression of inflammatory and immune markers, receptors of granulocytes, lymphocytes and monocytes [55]. In Winning’s model of
and the release of inflammatory cytokines including IL- 1a, 2, 6, 13, experimental endotoxinaemia, 25 annotated transcripts were found to
10, TNF alpha, and TNF beta [10]. Recent evidence has come to show be down-regulated in mice pre-treated with clopidogrel, compared to
that P2Y12 blockade may counteract platelet contribution to inflamma- mice without such pre-treatment [52]. The gene products are involved
tion in vivo and in vitro [37,51–53]. Inhibition of thrombin-induced in various networks of inflammation and cellular stress response such
P-selectin expression by P2Y12 blockade in human platelets in vitro, sig- as cytokines and their receptors (IL13, IL10r), complement factors and
nificantly curbs platelet-leukocyte interaction and subsequent platelet- immune globulins (C4b), leukotriene system, growth factors and recep-
induced reactive oxygen species (ROS) production in polymorphonu- tors, protein kinases and others [52] (Fig. 2). Thus, one could speculate
clear neutrophils and tissue factor production in monocytes [51] that clopidogrel, similarly to ASA, suppresses some proinflammatory
(Fig. 2). Moreover, a reduction of LPS-mediated thrombocytopenia, pathways.
fibrin deposition in the lungs, and inflammatory mediator upregulation Recent studies investigating the effects of R-138727, prasugrel’s
occurs in mice pretreated with clopidogrel [37,53,54]. Clopidogrel active metabolite, showed that low-micromolar concentrations of
attenuated the fall in platelet count that was observed 6 hours follow- this P2Y12 antagonist inhibited ADP-induced platelet P-selectin ex-
ing LPS administration, with no effect on survival and haemodynamics pression, as well as, the formation of platelet-leukocyte conjugates
during the first 24–48 hours. Nonetheless, in a later phase of the [56] (Fig. 2). Totani et al. [57] confirmed and extended these previous
K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138 135

Table 1
Clinical data on the use of antiplatelet agents and sepsis.

LITERATURE AIM STUDY DESIGN ANTIPLATELET AGENT PATIENTS-METHODS OUTCOME

Winning et al. Effect of antiplatelet agents Retrospective ASA, clopidogrel 615 admitted to an ICU Significant reduction in mortality
[36] on outcome of ICU patients cohort within 24 hrs after in patients with normal or high
hospitalization; 25% received bleeding risk
antiplatelet drugs
Winning et al. Effect of antiplatelet agents Retrospective ASA, clopidogrel 224 patients admitted for Significant reduction in need of
[37] on outcome in patients at cohort ticlopidin CAP; 20% received intensive care treatment and length
risk for organ failure anti-platelet drugs of hospital stay
Eisen et al. Association of ASA Retrospective ASA 5523 with SIRS; 2082 ASA administration was associated
[38] administration at the time of cohort receiving ASA. with a lower mortality in patients
development of SIRS with with SIRS/sepsis.
reduced mortality.
Sossdorf et al. Impact of low-dose Retrospective ASA, clopidogrel 979 patients with severe Decreased mortality following ASA
[39] ASA/NSAIDs on hospital sepsis/septic shock admitted administration; benefit of ASA abolished
mortality to a surgical ICU when NSAIDs co administered; NSAIDs,
clopidogrel, statins without significant
effects;
Otto et al. [40] Benefit of ASA in critically ill Retrospective ASA,clopidogrel 886 patients admitted to Significantly lower mortality in
patients & possible interference surgical ICU patients treated with ASA (100 mg/dl)
with atherosclerotic vascular during ICU stay. Evidence that
disease clopidogrel may have similar effect.
Benefit abolished when combined
use of ASA and clopidogrel
Erlich et al. Association between Retrospective ASA-containing 161 patients admitted to a Reduced incidence of ALI/ARDS
[41] pre-hospitalization antiplatelet cohort medication, clopidogrel medical ICU; 49% receiving
therapy & ALI/ARDS in ICU bisulfate, ticlopidine antiplatelet therapy
patients at high risk for ALI. hydrochloride, cilostazol,
dipyramidole
Valerio-Rojas Outcome of critically ill patients Retrospective ASA, clopidogrel 651 patients admitted to No association of chronic antiplatelet
et al. [42] with/without antiplatelet cohort ICU; 42.8% on antiplatelet therapy with decreased hospital/ICU
therapy before development of therapy (88.6% ASA,1.5% mortality. Potentially protective
severe sepsis/septic shock clopidogrel, 9.9% against development of ALI/ARDS &
ASA + clopidogrel) reduces needs of mechanical
ventilation
Losche et al. Effect of ASA administration on Observational ASA 834 patients admitted to ICU Reduction in ICU mortality
[43] outcome of ICU patients with with severe sepsis or septic
severe sepsis/septic shock shock; 20% received
low-dose ASA
Kor et al. Association between Secondary analysis ASA 3855 adult non-surgical Reduced incidence of ALI in those
[47] pre-hospitalization ASA use & ALI of prospective patients at-risk for ALI; 25.3% receiving ASA in univariate analysis;
in a cohort of at-risk patients multicenter receiving ASA during not statistically significant following
international cohort hospitalization propensity adjustment.
investigation
O'Neal et al. Association of prehospital statin Cross-sectional ASA 575 patients admitted to the No independent association of
[48] & combined ASA and statin use analysis of medical or surgical ICU; 26% pre-hospital aspirin use with
with risk of sepsis, ALI/ARDS, and prospective cohort on statin therapy prior to ALI/ARDS, sepsis, hospital mortality
mortality in critically ill patients. hospitalization in multivariable analysis. Lower
rates of ALI/ARDS, severe sepsis &
hospital mortality in combined ASA
and statin use in univariable analysis
Gross et al. Effects of clopidogrel on Retrospective clopidogrel 417,648 patients; 14,947 CAP more common among patients
[58] incidence & severity of CAP cohort (3.6%) receiving N6 taking clopidogrel. Near-significant
consecutive clopidogrel trends in need for mechanical
prescriptions ventilation, risk of ARDS/ALI, &
mortality favouring clopidogrel

ASA:acetylsalicylic acid; CAP: community acquired pneumonia, ALI: acute lung injury, ARDS: acute respiratory distress syndrome; ICU:intensive care unit; NSAID: non-steroidal
anti-inflammatory drug.

observations in vitro, showing that treatment with prasugrel seems to of antiplatelet drug therapy including clopidogrel with favourable out-
inhibit platelet activation and platelet-leukocyte interaction peptide come, [36,41,42] these results were mainly driven by the large number
in whole blood from mice subjected to a systemic inflammatory reac- of patients receiving ASA. Thus, due to the low number of patients receiv-
tion. Moreover, analysis of plasma markers of platelet activation and ing P2Y12 inhibitors in these studies no subgroup analysis is available.
inflammation, namely TXB2, TNF-alpha revealed that treatment No clinical data on the cyclopentyl-triazolo-pyrimidine ticagrelor
with prasugrel curbed inflammatory reactions, in vivo [57]. and the ATP analogue cangrelor currently exist. However, interest-
Clinical studies also suggest that P2Y12 inhibitors may be associ- ingly the mortality reduction observed with ticagrelor versus clopidogrel
ated with better outcomes in patients with pneumonia and critical following coronary artery bypass grafting (CABG) in the PLATelet inhibi-
illness [36,37] (Table 1). Prior treatment with antiplatelet agents, in- tion and patient Outcomes (PLATO) trial, was associated among others
cluding P2Y12 inhibitors, was associated with favourable length of with fewer deaths attributed to infection complication [59,60]. This find-
stay and fewer ICU admissions even though, community acquired ing comes in line with recent data indicating that ticagrelor appears to
pneumonia (CAP) appeared to be more common among patients taking reduce neutrophil recruitment and lung damage in abdominal sepsis in
clopidogrel [37,58]. However, later clinical studies reported an association pre treated mice [61]. The latter may reflect one of ticagrelor’s many
136 K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138

off-target pleiotropic effects attributed to its adenosine mediated mode described above, indicating similar contradictory results.[18,19] Thus,
of action [62], however, what the exact mechanism behind its apparently the question arises of whether hypercoagulability observed in mice
strong anti-inflammatory action is, remains to be seen. and rat models during endotoxinemia and/or sepsis is not actual, or
whether hypercoagulability quickly turns to hypocoagulability, owing
GPIIb/IIIa Antagonists to consumption of hemostatic factors depending on the stage of sepsis.
And if so, could sepsis trigger adaptive high platelet reactivity, hence
The platelet GPIIb/IIIa (αιιb/β3) receptor has been identified as the inadequate response to commonly used antiplatelet regimens, by pro-
pivotal mediator of platelet aggregation. GPIIb/IIIa antagonists block voking increased platelet aggregation at least in its initial stages? Inter-
fibrinogen binding to the GPIIb/IIIa receptor on activated platelets estingly, prasugrel’s inhibitory effects on a broad spectrum of platelet
and exert a strong antiplatelet effect by inhibiting the final common aggregation pathways was blunted in a recent model of systemic inflam-
step of platelet aggregation [63]. mation, in otherwise healthy volunteers following endotoxin infusion
There are only few reports on GPIIb/IIIa inhibitors in animal model of compared to placebo [73]. This is particularly important, as use of anti-
endotoxin-induced inflammatory responses and/or organ failure. In a platelet drugs is associated with increased bleeding events resulting in
rabbit model of endotoxic shock, treatment with AZ-1, a murine mono- their discontinuation in critically ill patients, even though, recent data
clonal antibody against GPIIb/IIIa that inhibits platelet aggregation, showed clear benefit even in patients with an increased bleeding risk
resulted in a decreased monocyte tissue factor expression and coagula- [36].
tion changes, restoration of endothelium-derived vascular relaxation In similar sense, there is conflicting evidence to support prophylactic
and significant decrease in endothelial cell injury, as well as, decreased antiplatelet administration in patients at risk for infection. In contrast to
mortality rate [64]. Moreover, in a previous study, Taylor et al. demon- pre-clinical results, suggesting a potential benefit of antiplatelet treat-
strated that a murine monoclonal antibody to the human platelet glyco- ment in infection [71], a retrospective study has suggested an increased
protein (GP)-IIb/IIIa receptor protects against disseminated intravascular risk of infection in clopidogrel-treated patients undergoing CABG com-
coagulation and ischemic organ damage observed in baboons treated pared with those treated with aspirin alone [74]. The mechanisms by
with C4b binding protein and sublethal infusion of Escherichia coli [65]. which clopidogrel may increase the risk of infection-related mortality
In the same manner, GP IIb/IIIa inhibition, using abciximab, protects are not known, even though, clopidogrel treatment has been associated
against endothelial dysfunction and an increase in leukocyte adherence with neutropenia [75]. Findings that use of clopidogrel may be associat-
to the vascular wall during experimental endotoxemia [66]. Of note, in- ed with increased risk of CAP and hospitalization, as well as, a trend to-
hibition of platelet aggregation via GPIIb/IIIa blockade with eptifibatide, wards reduced severity indices in hospitalized patients, [37] would be
attenuates platelet granzyme B-mediated apoptosis and results in less consistent with a biphasic role for platelets in lung infection and its se-
severe sepsis and extended survival in a murine model of abdominal quelae. Platelets may be important in the initial clearance of pathogens.
sepsis [67] (Fig. 2). However, whether these results can be extrapolated This is supported by the increased risk of sepsis seen among clopidogrel-
to humans remains unclear. In another study of human low grade treated patients. Once the infection progresses systemically, inhibition of
endotoxemia, GPIIb/IIIa blockade with eptifibatide or tirofiban did not platelet function may have beneficial consequences, especially with
seem to influence TF-induced coagulation activation, hence sepsis path- regards to mechanical ventilation, ALI, ARDS or even DIC according to a
way [68]. It is possible that these compounds may not be effective on cir- previous report [76]. Further investigations may determine the magni-
culating platelets, although, may exert a beneficial effect on platelets that tude of this effect.
are aggregated in the microcirculation [64].
Conclusions
Issues to be Considered
In the light of the above data, the current practice to discontinue
Animal studies and observational clinical studies mentioned above antiplatelet drugs in patients with sepsis should be tested in a prospec-
have provided some evidence that antiplatelet drugs may reduce tive trial. Since no specific therapy for sepsis, beyond source control and
organ failure and mortality in critically ill patients. However, in absence antimicrobial therapy currently exists, prevention or innovative treat-
of interventional and prospective large randomized trials the results ment approach is of utmost importance, especially in an era of stringent
presented here should be regarded as exploratory and hypothesis budget. Given the compelling observational and scientific data in this
generating, considered with vigilance while conclusions are being ex- debilitating syndrome, patients at high risk for the development of
trapolated with an uncertain degree of bias regarding the wide use of sepsis and a low risk for the development of side effects should be
antiplatelet agents in septic patients. considered as candidates of being prescribed, or at least not being
A number of issues that have to be considered further have been discontinued antiplatelet agents until clinical trials or further evidence
identified. First, in the clinical studies described, mostly or exclusively dictates otherwise.
aspirin was used as the antiplatelet drug, whereas clopidogrel or
GPIIb/IIIa inhibitors as rather specific antiplatelet drugs were pre-
Conflict of Interest Statement
dominantly used in animal models [43]. Following pooled analysis
of studies reporting the favourable association of antiplatelet therapy
Dr Alexopoulos reports receiving honoraria for lecturing by Astra
and outcomes in patients hospitalized for pneumonia, at risk for ALI,
Zeneca.
and/or critically ill, [58], the reduction in mortality was mainly driven
by the large study of Eisen et al., which may hamper the ability to
generalize the results since only ASA was utilized in the respective References
study [38]. [1] Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epide-
Moreover, it has previously been reported that sepsis, at least in its miology of severe sepsis in the United States: analysis of incidence, outcome, and
early state, is associated with hypercoagulability, indicated in partic- associated costs of care. Crit Care Med 2001;29:1303–10.
[2] Alberti C, Brun-Buisson C, Chevret S, Antonelli M, Goodman SV, Martin C, et al.
ular by an increase in clot formation rate and maximum clot firmness
Systemic inflammatory response and progression to severe sepsis in critically ill
[69,70]. However, the increase in hemostatic efficacy in these models infected patients. Am J Respir Crit Care Med 2005;171:461–8.
seems to be in contrast to data obtained in other studies, exhibiting a [3] Russwurm S, Vickers J, Meier-Hellmann A, Spangenberg P, Bredle D, Reinhart K,
significant prolongation of clotting time and clot formation rate as et al. Platelet and leukocyte activation correlate with the severity of septic
organ dysfunction. Shock 2002;17:263–8.
well as a decrease in clot formation rate and maximum clot firmness [4] Vincent JL, Yagushi A, Pradier O. Platelet function in sepsis. Crit Care Med 2002;30:
[71,72]. These findings could reflect respective aggregometry data S313–7.
K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138 137

[5] Katz JN, Kolappa KP, Becker RC. Beyond thrombosis: the versatile platelet in crit- [38] Eisen DP, Reid D, McBryde ES. Acetyl salicylic acid usage and mortality in critically
ical illness. Chest 2011;139:658–68. ill patients with the systemic inflammatory response syndrome and sepsis. Crit
[6] Weyrich AS, Zimmerman GA. Platelets: signaling cells in the immune continuum. Care Med 2012;40:1761–7.
Trends Immunol 2004;25:489–95. [39] Sossdorf M, Otto GP, Boettel J, Winning J, Losche W. Benefit of low-dose aspirin and
[7] Kerrigan SW, Cox D. Platelet-bacterial interactions. Cell Mol Life Sci 2010;67:513–23. non-steroidal anti-inflammatory drugs in septic patients. Crit Care 2013;17:402.
[8] Christaki E, Anyfanti P, Opal SM. Immunomodulatory therapy for sepsis: an up- [40] Otto GP, Sossdorf M, Boettel J, Kabisch B, Breuel H, Winning J, et al. Effects of
date. Expert Rev Anti Infect Ther 2011;9:1013–33. low-dose acetylsalicylic acid and atherosclerotic vascular diseases on the out-
[9] Behan MW, Chew DP, Aylward PE. The role of antiplatelet therapy in the second- come in patients with severe sepsis or septic shock. Platelets Sep. 20 2012
ary prevention of coronary artery disease. Curr Opin Cardiol 2010;25:321–8. [Epub ahead of print].
[10] Muhlestein JB. Effect of antiplatelet therapy on inflammatory markers in [41] Erlich JM, Talmor DS, Cartin-Ceba R, Gajic O, Kor DJ. Prehospitalization antiplatelet
atherothrombotic patients. Thromb Haemost 2010;103:71–82. therapy is associated with a reduced incidence of acute lung injury: a population-
[11] Semple JW, Italiano Jr JE, Freedman J. Platelets and the immune continuum. Nat based cohort study. Chest 2011;139:289–95.
Rev Immunol 2011;11:264–74. [42] Valerio-Rojas JC, Jaffer IJ, Kor DJ, Gajic O, Cartin-Ceba R. Outcomes of severe sepsis
[12] Warkentin TE, Aird WC, Rand JH. Platelet-endothelial interactions: sepsis, HIT, and septic shock patients on chronic antiplatelet treatment: a historical cohort
and antiphospholipid syndrome. Hematology Am Soc Hematol Educ Program; study. Crit Care Res Pract 2013;2013:782573.
2003. p. 497–519. [43] Losche W, Boettel J, Kabisch B, Winning J, Claus RA, Bauer M. Do aspirin and other
[13] Reininger AJ, Korndorfer MA, Wurzinger LJ. Adhesion of ADP-activated platelets antiplatelet drugs reduce the mortality in critically ill patients? Thrombosis
to intact endothelium under stagnation point flow in vitro is mediated by the 2012;2012:720254.
integrin alphaIIbeta3. Thromb Haemost 1998;79:998–1003. [44] Goff CD, Corbin RS, Theiss SD, Frierson Jr HF, Cephas GA, Tribble CG, et al.
[14] Bombeli T, Schwartz BR, Harlan JM. Adhesion of activated platelets to endothelial Postinjury thromboxane receptor blockade ameliorates acute lung injury. Ann
cells: evidence for a GPIIbIIIa-dependent bridging mechanism and novel roles for Thorac Surg 1997;64:826–9.
endothelial intercellular adhesion molecule 1 (ICAM-1), alphavbeta3 integrin, [45] Zarbock A, Singbartl K, Ley K. Complete reversal of acid-induced acute lung injury
and GPIbalpha. J Exp Med 1998;187:329–39. by blocking of platelet-neutrophil aggregation. J Clin Invest 2006;116:3211–9.
[15] Czabanka M, Peter C, Martin E, Walther A. Microcirculatory endothelial dysfunc- [46] Looney MR, Nguyen JX, Hu Y, Van Ziffle JA, Lowell CA, Matthay MA. Platelet depletion
tion during endotoxemia–insights into pathophysiology, pathologic mechanisms and aspirin treatment protect mice in a two-event model of transfusion-related
and clinical relevance. Curr Vasc Pharmacol 2007;5:266–75. acute lung injury. J Clin Invest 2009;119:3450–61.
[16] Diacovo TG, Roth SJ, Buccola JM, Bainton DF, Springer TA. Neutrophil rolling, arrest, [47] Kor DJ, Erlich J, Gong MN, Malinchoc M, Carter RE, Gajic O, et al. Association of
and transmigration across activated, surface-adherent platelets via sequential action prehospitalization aspirin therapy and acute lung injury: results of a multicenter in-
of P-selectin and the beta 2-integrin CD11b/CD18. Blood 1996;88:146–57. ternational observational study of at-risk patients. Crit Care Med 2011;39:2393–400.
[17] Freishtat RJ, Natale J, Benton AS, Cohen J, Sharron M, Wiles AA, et al. Sepsis alters the [48] O'Neal Jr HR, Koyama T, Koehler EA, Siew E, Curtis BR, Fremont RD, et al.
megakaryocyte-platelet transcriptional axis resulting in granzyme B-mediated Prehospital statin and aspirin use and the prevalence of severe sepsis and acute
lymphotoxicity. Am J Respir Crit Care Med 2009;179:467–73. lung injury/acute respiratory distress syndrome. Crit Care Med 2011;39:1343–50.
[18] Boldt J, Menges T, Wollbruck M, Sonneborn S, Hempelmann G. Platelet function in [49] Storey RF. Biology and pharmacology of the platelet P2Y12 receptor. Curr Pharm
critically ill patients. Chest 1994;106:899–903. Des 2006;12:1255–9.
[19] Gawaz M, Dickfeld T, Bogner C, Fateh-Moghadam S, Neumann FJ. Platelet function [50] Dorsam RT, Kunapuli SP. Central role of the P2Y12 receptor in platelet activation.
in septic multiple organ dysfunction syndrome. Intensive Care Med 1997;23: J Clin Invest 2004;113:340–5.
379–85. [51] Evangelista V, Manarini S, Dell'Elba G, Martelli N, Napoleone E, Di Santo A, et al.
[20] Johansson D, Shannon O, Rasmussen M. Platelet and neutrophil responses to Gram Clopidogrel inhibits platelet-leukocyte adhesion and platelet-dependent leuko-
positive pathogens in patients with bacteremic infection. PLoS One 2011;6:e26928. cyte activation. Thromb Haemost 2005;94:568–77.
[21] Matera C, Falzarano C, Berrino L, Rossi F. Effects of tetanus toxin, Salmonella [52] Winning J, Baranyai J, Claus R, Eisenhut I, Hamacher J, Reinhart K, et al. Beneficial
typhimurium porin, and bacterial lipopolysaccharide on platelet aggregation. effects of antiplatelet drugs in patients with community-acquired pneumonia and
J Med 1992;23:327–38. in endotoxin shock in mice. Crit Care 2007;11:P27.
[22] Isogai E, Kitagawa H, Isogai H, Matsuzawa T, Shimizu T, Yanagihara Y, et al. Effects [53] Evangelista V, dell’Elba G, Martelli N, Amore C, Pecce R, Piccoli A, et al.
of leptospiral lipopolysaccharide on rabbit platelets. Zentralbl Bakteriol 1989;271: Anti-inflammatory effects of clopidogrel in the mouse. J Thromb Haemost
186–96. 2007;5 [P-M-283].
[23] Itoh H, Cicala C, Douglas GJ, Page CP. Platelet accumulation induced by bacterial [54] Aslam R, Speck ER, Kim M, Crow AR, Bang KW, Nestel FP, et al. Platelet Toll-like re-
endotoxin in rats. Thromb Res 1996;83:405–19. ceptor expression modulates lipopolysaccharide-induced thrombocytopenia and
[24] Saba HI, Saba SR, Morelli G, Hartmann RC. Endotoxin-mediated inhibition of tumor necrosis factor-alpha production in vivo. Blood 2006;107:637–41.
human platelet aggregation. Thromb Res 1984;34:19–33. [55] Setzer F, Oberle V, Blass M, Moller E, Russwurm S, Deigner HP, et al. Platelet-derived
[25] Sheu JR, Hsiao G, Lee C, Chang W, Lee LW, Su CH, et al. Antiplatelet activity of microvesicles induce differential gene expression in monocytic cells: a DNA microar-
Staphylococcus aureus lipoteichoic acid is mediated through a cyclic AMP path- ray study. Platelets 2006;17:571–6.
way. Thromb Res 2000;99:249–58. [56] Frelinger III AL, Jakubowski JA, Li Y, Barnard MR, Fox ML, Linden MD, et al. The
[26] Mavrommatis AC, Theodoridis T, Orfanidou A, Roussos C, Christopoulou-Kokkinou V, active metabolite of prasugrel inhibits ADP-stimulated thrombo-inflammatory
Zakynthinos S. Coagulation system and platelets are fully activated in uncomplicated markers of platelet activation: Influence of other blood cells, calcium, and aspirin.
sepsis. Crit Care Med 2000;28:451–7. Thromb Haemost 2007;98:192–200.
[27] Yaguchi A, Pradier O, Lobo S, Vincent JL. Platelet aggregation is impaired at the [57] Totani L, Dell'Elba G, Martelli N, Di Santo A, Piccoli A, Amore C, et al. Prasugrel in-
level of cyclooxygenase and thromboxane synthetase in severe sepsis. Intensive hibits platelet-leukocyte interaction and reduces inflammatory markers in a
Care Med 2001;27:S166. model of endotoxic shock in the mouse. Thromb Haemost 2012;107:1130–40.
[28] Morris T, Stables M, Hobbs A, de Souza P, Colville-Nash P, Warner T, et al. Effects of [58] Gross AK, Dunn SP, Feola DJ, Martin CA, Charnigo R, Li Z, et al. Clopidogrel treat-
low-dose aspirin on acute inflammatory responses in humans. J Immunol 2009;183: ment and the incidence and severity of community acquired pneumonia in a co-
2089–96. hort study and meta-analysis of antiplatelet therapy in pneumonia and critical
[29] Jilma B, Blann A, Pernerstorfer T, Stohlawetz P, Eichler HG, Vondrovec B, et al. Reg- illness. J Thromb Thrombolysis 2013;35:147–54.
ulation of adhesion molecules during human endotoxemia. No acute effects of as- [59] Varenhorst C, Alstrom U, Scirica BM, Hogue CW, Asenblad N, Storey RF, et al. Factors
pirin. Am J Respir Crit Care Med 1999;159:857–63. contributing to the lower mortality with ticagrelor compared with clopidogrel in pa-
[30] Pernerstorfer T, Schmid R, Bieglmayer C, Eichler HG, Kapiotis S, Jilma B. Acetamin- tients undergoing coronary artery bypass surgery. J Am Coll Cardiol 2012;60:1623–30.
ophen has greater antipyretic efficacy than aspirin in endotoxemia: a random- [60] Storey R, Steg P, James S, Husted S, Cannon C, Becker R, et al. Reduction in pulmo-
ized, double-blind, placebo-controlled trial. Clin Pharmacol Ther 1999;66:51–7. nary adverse events and associated sepsis and mortality in acute coronary
[31] Hinshaw LB, Solomon LA, Erdos EG, Reins DA, Gunter BJ. Effects of acetylsalicylic acid syndrome patients treated with ticagrelor compared to clopidogrel: a post hoc
on the canine response to endotoxin. J Pharmacol Exp Ther 1967;157:665–71. analysis of the plato study. J Am Coll Cardiol 2012;59:E482.
[32] Halushka PV, Wise WC, Cook JA. Protective effects of aspirin in endotoxic shock. [61] Braun O, Rahman M, Gustafsson D, Thorlacius H. Ticagrelor Reduces Neutrophil
J Pharmacol Exp Ther 1981;218:464–9. Recruitment and Lung Damage in Abdominal Sepsis. J Am Coll Cardiol 2013;61:10S.
[33] Derhaschnig U, Schweeger-Exeli I, Marsik C, Cardona F, Minuz P, Jilma B. Effects of [62] Serebruany VL. Mortality benefit in PLATO cannot be explained by antiplatelet
aspirin and NO-aspirin (NCX 4016) on platelet function and coagulation in human properties of ticagrelor. Cardiology 2010;117:231–3.
endotoxemia. Platelets 2010;21:320–8. [63] Coller BS. Blockade of platelet GPIIb/IIIa receptors as an antithrombotic strategy.
[34] del Soldato P, Sorrentino R, Pinto A. NO-aspirins: a class of new anti-inflammatory Circulation 1995;92:2373–80.
and antithrombotic agents. Trends Pharmacol Sci 1999;20:319–23. [64] Pu Q, Wiel E, Corseaux D, Bordet R, Azrin MA, Ezekowitz MD, et al. Beneficial effect
[35] Lechi C, Andrioli G, Gaino S, Tommasoli R, Zuliani V, Ortolani R, et al. The of glycoprotein IIb/IIIa inhibitor (AZ-1) on endothelium in Escherichia coli
antiplatelet effects of a new nitroderivative of acetylsalicylic acid–an in vitro endotoxin-induced shock. Crit Care Med 2001;29:1181–8.
study of inhibition on the early phase of platelet activation and on TXA2 produc- [65] Taylor FB, Coller BS, Chang AC, Peer G, Jordan R, Engellener W, et al. 7E3 F(ab')2, a
tion. Thromb Haemost 1996;76:791–8. monoclonal antibody to the platelet GPIIb/IIIa receptor, protects against
[36] Winning J, Neumann J, Kohl M, Claus RA, Reinhart K, Bauer M, et al. Antiplatelet microangiopathic hemolytic anemia and microvascular thrombotic renal failure
drugs and outcome in mixed admissions to an intensive care unit. Crit Care Med in baboons treated with C4b binding protein and a sublethal infusion of
2010;38:32–7. Escherichia coli. Blood 1997;89:4078–84.
[37] Winning J, Reichel J, Eisenhut Y, Hamacher J, Kohl M, Deigner HP, et al. Anti-platelet [66] Walther A, Czabanka M, Gebhard MM, Martin E. Glycoprotein IIB/IIIA-inhibition
drugs and outcome in severe infection: clinical impact and underlying mechanisms. and microcirculatory alterations during experimental endotoxemia–an intravital
Platelets 2009;20:50–7. microscopic study in the rat. Microcirculation 2004;11:79–88.
138 K. Akinosoglou, D. Alexopoulos / Thrombosis Research 133 (2014) 131–138

[67] Sharron M, Hoptay CE, Wiles AA, Garvin LM, Geha M, Benton AS, et al. Platelets [72] Reinhofer M, Brauer M, Franke U, Barz D, Marx G, Losche W. The value of rotation
induce apoptosis during sepsis in a contact-dependent manner that is inhibited thromboelastometry to monitor disturbed perioperative haemostasis and bleeding
by GPIIb/IIIa blockade. PLoS One 2012;7:e41549. risk in patients with cardiopulmonary bypass. Blood Coagul Fibrinolysis 2008;19:
[68] Derhaschnig U, Pachinger C, Schweeger-Exeli I, Marsik C, Jilma B. Blockade of 212–9.
GPIIb/IIIa by eptifibatide and tirofiban does not alter tissue factor induced [73] Spiel AO, Derhaschnig U, Schwameis M, Bartko J, Siller-Matula JM, Jilma B. Effects
thrombin generation in human endotoxemia. Thromb Haemost 2003;90: of prasugrel on platelet inhibition during systemic endotoxaemia: a randomized
1054–60. controlled trial. Clin Sci (Lond) 2012;123:591–600.
[69] Gonano C, Sitzwohl C, Meitner E, Weinstabl C, Kettner SC. Four-day antithrombin [74] Blasco-Colmenares E, Perl TM, Guallar E, Baumgartner WA, Conte JV, Alejo D, et al.
therapy does not seem to attenuate hypercoagulability in patients suffering from Aspirin plus clopidogrel and risk of infection after coronary artery bypass surgery.
sepsis. Crit Care 2006;10:R160. Arch Intern Med 2009;169:788–96.
[70] Luckner G, Mayr VD, Fries DR, Innerhofer P, Jochberger S, Hasibeder WR, et al. [75] Almsherqi ZA, McLachlan CS, Sharef SM. Non-bleeding side effects of clopidogrel:
Uncovering hypercoagulability in sepsis using ROTEMthromboelastometry: a have large multi-center clinical trials underestimated their incidence? Int J Cardiol
case series. Open Crit Care Med 2008;1:1–6. 2007;117:415–7.
[71] Seidel M, Winning J, Claus RA, Bauer M, Losche W. Beneficial effect of [76] Cartin-Ceba R, Valerio-Rojas J, Cabello-Garza J, Kelm D, Gajic O. Antiplatelet
clopidogrel in a mouse model of polymicrobial sepsis. J Thromb Haemost Therapy And Disseminated Intravascular Coagulation In Septic Shock Patients:
2009;7:1030–2. Propensity Score Analysis. Am J Respir Crit Care Med 2011;183:A4698.

Potrebbero piacerti anche