You are on page 1of 19

European Heart Journal (2023) 44, 3040–3058 SPECIAL ARTICLE

https://doi.org/10.1093/eurheartj/ehad388 Thrombosis and antithrombotic treatment

Management of patients on antithrombotic


therapy with severe infections: a joint clinical
consensus statement of the ESC Working
Group on Thrombosis, the ESC Working Group

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


on Atherosclerosis and Vascular Biology,
and the International Society on Thrombosis
and Haemostasis
Bruna Gigante 1*, Jerrold H. Levy 2, Eric van Gorp 3,
Alessandro Bartoloni 4, Marie-Luce Bochaton-Piallat 5, Magnus Bäck 6,7
,
Hugo ten Cate 8,9, Christina Christersson 10, José Luis Ferreiro 11,
Tobias Geisler 12, Esther Lutgens 13,14, Sam Schulman 15,16,
Robert F. Storey 17, Jecko Thachil 18, Gemma Vilahur 19,
Patricia C. Liaw 20†, and Bianca Rocca 21*†
1
Division of Cardiovascular Medicine, Department of Medicine, Karolinska Institutet, Solnavägen 30. 17164 and Department of Cardiology, Danderyds Hospital, Entrévägen 2, 182 88,
Stockholm, Sweden; 2Departments of Anesthesiology, Critical Care, and Surgery (Cardiothoracic), Duke University School of Medicine, Durham, United States; 2301 Erwin Road, Durham,
NC 27710, USA; 3Department of Viroscience, Erasmus MC, Rotterdam, PO box 2040 [Room Ee1726], 3000 CA Rotterdam, The Netherlands; 4Department of Experimental and Clinical
Medicine, Infectious Diseases Unit, University of Florence, Largo Brambilla 3, 50100 Florence, Italy; 5Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1
rue Michel-Servet 1, CH-1211 Genève, Switzerland; 6Department of Translational Cardiology, Karolinska Institutet and Karolinska University Hospital, CMM L8:01, 171 76 Stockholm,
Sweden; 7INSERM U1116, University of Lorraine, Nancy University Hospital, 2 rue Jean Lamour, 54505 Vandoeuvre les Nancy Cedex, France; 8Department of Internal medicine,
Thrombosis Expertise Center, Maastricht University Medical Center and CARIM school for cardiovascular diseases, Universiteitsingel 50, PO Box 616, 6200 MD Maastricht, The
Netherlands; 9Center for Thrombosis and Haemostasis, Gutenberg University Medical Center, Langenbeckstr. 1, Bldg. 403, 55131 Mainz, Germany; 10Department of Medical Sciences,
Cardiology, Uppsala University, Akademiska Sjukhuset, 75185, Uppsala, Sweden; 11Department of Cardiology and Bio-Heart Cardiovascular Diseases Research Group; Bellvitge University
Hospital – Bellvitge Biomedical Research Institute (IDIBELL); CIBERCV; L’Hospitalet de Llobregat, Hospital Duran i Reynals - Edifici Terapèutic - 2a planta Gran Via de l’Hospitalet, 199,
08908 Hospitalet de Llobregat Barcelona – Spain; 12Department of Cardiology and Angiology, University Hospital Tübingen, Otfried-Müller-Straße 10, 72076 Tübingen, Germany;
13
Cardiovascular Medicine, Experimental CardioVascular Immunology Laboratory, Mayo Clinic, 200 First St SW, 55905, Rochester, MN, USA; 14Institute for Cardiovascular Prevention
(IPEK), Ludwig-Maximilians Universität, München, Germany & German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Pettenkoferstrasse 9, 80336,
Munich, Germany; 15Department of Medicine and Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, ON, Canada HHS - General Hospital 237, Barton
Street East, Hamilton, ON, L8L 2X2, Canada; 16Department of Obstetrics and Gynecology, I.M. Sechenov First Moscow State Medical University, Bol’shaya Pirogovskaya Ulitsa, 2, стр. 4,
Moscow 119435, Russia; 17Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Pegasus House, 463a Glossop Road, Sheffield, S10 2QD, UK;
18
Department of Haematology, Manchester University Hospitals, Oxford road, Manchester, M13 9WL, UK; 19Institut de Recerca Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau,
CIBERCV, Avda. Sant Antoni Maria Claret 167, 08025 Barcelona, Spain; 20Department of Medicine, Thrombosis & Atherosclerosis Research Institute (TaARI), McMaster University, 237
Barton Street East Hamilton, Ontario L8L 2X2, Canada; and 21Department of Safety and Bioethics, Section on Pharmacology, Catholic University School of Medicine, Largo F. Vito 1, 00168
Rome, Italy

Received 6 October 2022; revised 1 May 2023; accepted 29 May 2023; online publish-ahead-of-print 13 July 2023

* Corresponding authors. Tel/Fax: +39 06 30154253. E-mails: bianca.rocca@unicatt.it; b.rocca@tiscali.it (B.R.); Tel/Fax: +46-(0)8-52487082. E-mail: bruna.gigante@ki.se (B.G.)

P.C.L. and B.R. are last co-authors.
© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
Severe infections and antithrombotic therapy 3041

Graphical Abstract

Sepsis
Induced
Coagulopathy
Score

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


Anticoagulants
SAPT DAPT
0 (OAC, Heparins)

Platelets
No change No change No change
149-100x109/L

Platelets
No change No change OAC
<100-30x109/L

Platelets
No change Heparins
29-20x109/L

Platelets
6 <20x109/L

Bacteria and viruses are a common cause of severe infections requiring hospitalization and different degrees of organ support. Patients on single or
combined antithrombotic therapy at high and very high cardiovascular risk are more prone to severe infections and related complications in the
short and long term than the general population. With increasing severity, infections associate with coagulopathy and this imposes modulation
of antithrombotic therapy according to the underlying cardiovascular diseases, indication for treatment, clinical conditions, and patient’s prognosis.
When platelet count is below 100 × 109/L, in patients already on oral anticoagulation (OAC), heparins should be used as described in Tables 3 and 4
and should be stopped when platelet count is below 30 × 109/L. Patients with ongoing dual antiplatelet therapy (DAPT) should be shifted to single
antiplatelet therapy (SAPT) with a P2Y12 inhibitor or low-dose acetylsalicylic acid (ASA). Single antiplatelet therapy with ASA may be more favour­
able in these patients when platelet count is below 30 × 109/L. When platelet count is below 20 × 109/L, antithrombotic therapy should be discon­
tinued, with the possible exception of patients with very recent acute coronary syndrome, i.e. <3 months, in whom low-dose ASA may be
considered, as shown in Tables 3 and 4.

Abstract

Patients with severe infections and a pre-existing indication for antithrombotic therapy, i.e. antiplatelet agents, anticoagulant drugs, or their combi­
nations, require integrated clinical counselling among coagulation, infectious disease, and cardiology specialists, due to sepsis-induced coagulopathy
that frequently occurs. Bacterial and viral pathogens constitute an increasing threat to global public health, especially for patients with ongoing an­
tithrombotic treatment who have a high risk of thrombotic recurrences and high susceptibility to severe infections with increased morbidity and
mortality. Similarly, sepsis survivors are at increased risk for major vascular events. Coagulopathy, which often complicates severe infections, is as­
sociated with a high mortality and obligates clinicians to adjust antithrombotic drug type and dosing to avoid bleeding while preventing thrombotic
complications. This clinical consensus statement reviews the best available evidence to provide expert opinion and statements on the management
of patients hospitalized for severe bacterial or viral infections with a pre-existing indication for antithrombotic therapy (single or combined), in whom
sepsis-induced coagulopathy is often observed. Balancing the risk of thrombosis and bleeding in these patients and preventing infections with vac­
cines, if available, are crucial to prevent events or improve outcomes and prognosis.
.............................................................................................................................................................................................
Keywords Antiplatelet drugs • Anticoagulant drugs • Sepsis • Infections • Sepsis-induced coagulopathy
3042 Gigante et al.

the current ESC Scientific Documents policy as detailed in


Introduction Supplementary data online, Figure S1.
Severe bacterial and viral infections are often associated with a coagulo­ Patients on mechanical circulatory support were not included.
pathy that can be defined as any alteration or disturbance of haemostasis Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infec­
resulting in either bleeding, clotting, or both.1 More specifically, a dysre­ tion was also excluded since it has recently been addressed by dedi­
gulated balance of thrombosis and fibrinolysis can result in excessive clot­ cated ESC and ISTH guidelines.18,19
ting, bleeding, or both, depending on the host’s immunological and
inflammatory responses that change over time.2,3 Antithrombotic drugs
are increasingly used worldwide, with a predicted annual growth rate of Immunothrombosis or
nearly 8% between 2022 and 20294 and an estimated prevalence of pre­ thromboinflammation in severe
scriptions in the European population of ∼15%–20% across different
countries.5,6 Thus, the management of antithrombotic treatment in pa­ infections?
tients suffering from severe infections represents a frequent clinical prob­ To describe the prothrombotic and coagulopathic responses induced

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


lem and a challenge. Clinical decisions should balance thrombotic and by the immune and inflammatory reactions to infections, in the last
bleeding risks associated with severe infections as well as the underlying two decades, the literature interchangeably uses the words thromboin­
history of arterial and/or venous thrombotic disease requiring antithrom­ flammation and immunothrombosis to describe the link between in­
botic therapy. flammation and thrombosis.2,3,20 Both terms were first reported by
The scope of this clinical consensus statement is to review the cur­ Tanguay et al. in 2004.21
rent evidence on the management of patients with ongoing antithrom­ The term thromboinflammation is derived from thrombosis asso­
botic therapy (single or combined antiplatelet and/or anticoagulant ciated with inflammation and is used to describe pathophysiologic
agents) who are hospitalized because of a severe infection (Graphical perturbations due to vascular endothelial injury and/or loss of antith­
Abstract). This document focuses on severe infections of bacterial and rombotic and antiinflammatory functions.21 As a result, both cellular
viral aetiology and refers to the sepsis-induced coagulopathy (SIC) cri­ and humoral inflammatory mechanisms of immune surveillance are
teria, defined and scored according to the International Society on activated. In acute infections, thromboinflammation may culminate
Thrombosis and Haemostasis (ISTH) (Table 1).1 in microvascular thrombosis, which is the hallmark of the disease,
as has been reported in post-mortem studies of patients with acute
respiratory distress syndrome due to pathogens invading the re­
spiratory tract and provoking an inflammatory response associated
Methodology and definitions with acute lung injury.22 This thromboinflammatory response be­
The panel of co-authors has been selected because of their came increasingly recognized currently due to coronavirus disease
complementary expertise in the fields of cardiovascular pharmacol­ 2019 (COVID-19).23 This response to sepsis also occurs following
ogy, infectious disease, clinical cardiology, coagulation, and athero­ ischaemia–reperfusion injury,24 trauma, organ transplant rejection,
sclerosis pathophysiology, as detailed in the Supplementary material and extracorporeal circulation and can progress to DIC.2 In these
online. thromboinflammatory states, loss of endothelial cell function and
We performed a systematic review of the literature (Supplementary systemic inflammatory responses lead to humoral and cellular injury,
data online, Table S1) and used the current European Society of as noted.
Cardiology (ESC) classification of cardiovascular risk9 (see Immunothrombosis was again described in 2013 by Engelmann and
Supplementary data online, Table S2). Severe infections were defined Massberg25 to refer to an intrinsic effector pathway of innate (host)
according to the Third International Consensus Definitions for Sepsis immunity that activates systemic inflammation and haemostasis,
and Septic Shock (see Supplementary data online, Table S3).10 The with thrombin and fibrin generation to localize and immobilize infec­
SIC score was used to grade the severity of the coagulopathy. This tions. As part of any inflammatory response to attenuate microbial in­
score has been validated in sepsis patients with coagulopathy and shows vasion, the microcirculatory thrombosis also produces multiorgan
higher specificity compared with disseminated intravascular coagulopa­ injury.26,27 These important host defence mechanisms have been
thy (DIC) (see Supplementary data online, Table S4).11–14 Furthermore, long known, but with the ongoing pandemic and massive numbers
anticoagulant therapy has been reported to benefit patients who met of COVID-19 patients without pre-existing immunity who manifested
criteria for SIC.14 Finally, since DIC can also be of non-infectious eti­ lung or multiorgan dysfunction, the concept of immunothrombosis
ology,11 we agreed to use SIC and not to refer to DIC.1,8 was increasingly reported. This term describes the microvascular
We agreed to refer to the Sequential Organ Failure Assessment thrombotic response that facilitates microbe containment and
(SOFA) score (see Supplementary data online, Table S3),15 since ele­ elimination, a critical component of innate immunity.2,25 Similar to
ments of the SOFA score are included in the SIC criteria (Table 1) to thromboinflammation, this response requires humoral and cellular ac­
grade the severity of sepsis. Notably, SIC development in patients hos­ tivation, due to contact activation pathways through factor XII and
pitalized for sepsis increases mortality from 25.4% to 56.1%.16 complement activation, and subsequent generation of additional
We agreed to address the pathophysiology of haemostasis and the pro-inflammatory mediators that include neutrophil extracellular
management of patients with ongoing antiplatelet and anticoagulant traps.26,28 Thus, contact activation pathways likely represent a physio­
therapy during severe infections and the benefits and risks of vaccin­ logical mechanism of innate immunity and are also crucial in SIC and
ation as preventive strategy. Consensus statements for each section acute infections.3,29,30
were reached using the Delphi methodology17 as detailed in the In summary, thromboinflammation and immunothrombosis have
supplementary material online. In clinical consensus statement, advices many similarities, but should not be used interchangeably, even if
for clinical management were classified in four categories, according to they have been used synonymously in the past. Although
Severe infections and antithrombotic therapy 3043

prothrombin time (PT) and progressive thrombocytopenia; however,


Table 1 Scoring for sepsis-induced coagulopathy (SIC)a
thrombocytopenia can also be multifactorial in intensive care unit
Points (ICU) patients. Conversely, patients with SIC often have acute lung in­
................................... jury and/or vasoplaegia/shock. Acrocyanosis and ischaemic limbs that
0 1 2 are not related to vasopressor use occur in association with SIC and
....................................................................................
Prothrombin time (PT-INR) >1.2 1.2–1.4 >1.4 shock liver.40
9
Critical to managing these patients is early intervention with
Platelet count (×10 /L) >150 100–149 <100 antibiotics, source control, and cardiopulmonary support, as septic
b
Total SOFA (respiratory, 0 1 ≥2 shock can also ensue. The initial site of infection, especially in critical or­
cardiovascular, hepatic, and renal) gans, may also be relevant for personalized decisions on antithrombotic
drugs.
SOFA, Sequential Organ Failure Assessment; INR, international normalized ratio.
Modified from Iba et al.7
Moreover, the causative bacterial trigger may not be isolated, and of­
a
Total SIC score ≥4 corresponds to DIC ≥ 5.8 ten patients are treated with empiric broad-spectrum or multiple anti­

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


b
SOFA scoring included in SIC grading does not include the mental status since it is biotic agents that may elicit cytochrome P450 (CYP)-based, clinically
tailored to clinical aspects relevant in sepsis.
relevant drug–drug interactions with some antithrombotic agents [clo­
pidogrel, ticagrelor, dabigatran, and vitamin K antagonist (VKA)], as
shown in the Supplementary data online, Table S6.
immunothrombosis per se is a protective, antimicrobial mechanism
aimed to locally contain pathogens including bacteria and fungi, its dys­
regulation and systemic exacerbation, as observed in thromboinflam­ Viral infections
mation, may ultimately be harmful to the host. The incidence of viral sepsis is unknown as often the viral aetiology is
difficult to ascertain.41,42 Depending on the season, sepsis may compli­
cate 25%–40% of viral infections. During ICU admittance, secondary in­
Haemostasis during severe fections, which were not the reason for admittance, and bacterial
infections in severe, primarily airborne viral infections (e.g. influenza
infections: pathophysiology and and SARS-CoV-2) can also occur.43 Viral sepsis represents ∼30% of
clinical aspects sepsis in South East Asia.44 Viral infectious diseases transmitted by ani­
mal vectors pose potential global threats,45 as viral haemorrhagic fevers
Bacterial infections such as dengue have the potential to become pandemic.46 Viral infec­
In recent decades, the incidence of bacterial sepsis has increased: in the tions trigger coagulation disorders mostly through inflammatory path­
USA, the estimated incidence was 240 cases/100 000 population in ways, similar to bacterial agents.47,48 However, unlike most bacterial
2015,31 while in 2019, a nationwide study in France estimated an inci­ infections treatable with antibiotics, specific antiviral drugs are often
dence of 403/100 000 population.32 Global, climate, and demographic not available and/or have limited efficacy.
changes affect infectious disease features and epidemiology.33 Severe Many viruses continue their thromboinflammatory effects until the
infections caused by gram-positive bacteria34,35 have increased, almost host immune system can engage, often including elements of the coagu­
reaching the incidence observed for gram-negative bacteria. The lation and fibrinolytic cascades depending on the infection severity.49
thromboinflammatory cascade triggered by infections is summarized Many enveloped viruses, such as SARS-CoV-2, herpes simplex virus
in Figure 1. The mechanisms underlying the coagulopathy39 observed type 1, and influenza, acquire host-derived constituents such as tissue
during the most common severe bacterial infections in hospitalized pa­ factor, which enhance viral infectivity and promote activation of coagu­
tients are represented in Figure 2 and summarized in Supplementary lation and inflammation.36 In acute infections, macro- and microthrom­
data online, Table S5. boinflammation triggered by endothelial injury, leukocyte subset
Coagulopathy and vascular endothelial injury occur following severe activation and migration, and complement activation contributes
bacterial infections due to inflammatory and thrombotic responses that ultimately to organ failure (Figure 1).1,49 Viral infections, with their
include both humoral and cellular components of haemostasis, namely different clinical phenotypes (mild, moderate, and severe) and acute
the coagulation cascade, adhesive proteins, platelets, and inflammatory/ or chronic evolution, may present different risk profiles with
immune cells (e.g. neutrophils, lymphocytes, and monocytes).1,2 regard to thrombotic complications.47 Another important distinction
Multiple pathways, including enhanced tissue factor expression, neutro­ with regard to the clinical phenotype is prothrombotic virus types, in­
phil activation, and release of multiple cellular constituents, such as cluding influenza, SARS-CoV-2, and human immunodeficiency virus, as
DNA, histones, and damage-associated molecular patterns, are in­ compared with haemorrhagic viruses such as dengue, Ebola, and
volved, overwhelming the physiological vascular-protective mechan­ Hanta.47
isms (Figure 1). Endothelial dysfunction is associated with loss of the Severe viral infection may initially be complicated by more localized
endogenous vascular-protective mechanisms and contributes to micro­ microthrombi, bleeding, and/or multiorgan failure.1,47 Critically ill pa­
thrombi.2 Endothelial cells also enhance the synthesis of tissue plas­ tients requiring mechanical ventilation, extracorporeal life support,
minogen activator and plasminogen activator inhibitor-1, with and prolonged ICU stays may also develop secondary bacterial infec­
consequent derangement of the coagulation and fibrinolytic systems, tions, such as ventilator-associated pneumonia, and additional haemo­
contributing to SIC1,16 (Table 1), a life-threatening complication of se­ static dysfunction. Understanding and classifying the thromboembolic
vere infections characterized by consumption of clotting factors and risk period are important to balance the risks of bleeding vs. thrombosis
platelets, co-existence of thrombosis, hypofibrinolysis and bleeding, at a given time point with regard to prevention of venous thrombo­
and possible progression to DIC and multiorgan failure.1,7,40 The two embolism (VTE) and/or continuation of ongoing antithrombotic
major biomarkers of SIC diagnosis (Table 1) are prolonged therapy.
3044 Gigante et al.

Bacteria. Lipopolysaccharide (LPS) Viruses. Enveloped viruses such as


(gram-negative) and peptidoglycan SARS-CoV-2 and HSV-1 acquire
(gram-positive) bind to Toll-like receptors endothelial- and monocyte-derived TF
(TLRs)/CD14 on monocytes. This which enhances viral infectivity and also
induces the release of inflammatory activates coagulation. Viruses can also
cytokines and upregulates tissue factor activate platelets (e.g. via SARS-CoV-
(TF) expression. Bacteria can also 2/ACE2 interactions) which results in
activate platelets (e.g. via binding of LPS platelet aggregation and the release of
with platelet TLRs). inflammatory cytokines.

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


Thrombin

Figure 1 Common mechanisms of infection-induced thromboinflammation. Bacteria and viruses induce the expression of tissue factor via distinct
mechanisms (described above in boxes). Tissue factor initiates coagulation which results in the generation of thrombin, an enzyme that not only forms
the fibrin clot but also activates platelets and the complement cascade. Inflammatory cytokines such as tumour necrosis factor α, interleukin-6,
interleukin-1, and prostaglandins promote endothelial cell injury. Injured endothelial cells exhibit a prothrombotic phenotype by shedding natural antic­
oagulants such as heparan sulfate and thrombomodulin, releasing plasminogen activator inhibitor-1 which inhibits fibrinolysis, and releasing ultra-large
von Willebrand factor and P-selectin from Weibel–Palade bodies. Ultra-large von Willebrand factor and P-selectin mediate platelet and neutrophil ad­
hesion, respectively. Activated platelets induce the release of neutrophil extracellular traps which provide a scaffold that traps platelets and red blood
cells. Neutrophil extracellular trap components such as extracellular DNA, histones, and damage-associated molecular patterns trigger coagulation
activate platelets and impair fibrinolysis. The net result of severe bacterial and viral infections is vascular occlusion characterized by thromboinflamma­
tion.7,36–38 Created with Biorender.com. TF, tissue factor; HS, heparan sulfate; TM, thrombomodulin; PAI, plasminogen activator inhibitor; ULVWF,
ultra-large von Willebrand factor; NETs, neutrophil extracellular traps; DAMPs, damage-associated molecular patterns.

Patients with severe infections on ASA may be considered in those patients with documented significant
coronary or peripheral artery disease on imaging, or diabetes mellitus
antiplatelet therapy (DM) with organ damage and/or longstanding and/or other multiple
risk factors.9,50,51 Moreover, high coronary artery calcium score (mostly
Patients with multiple cardiovascular risk ≥100 Agatston)52 has been suggested as cardiovascular risk enhancer,
factors and enhancers useful to re-assess and stratify cardiovascular risk and identify patients
Recent ESC guidelines have classified cardiovascular risk as high or very who may derive a net benefit from low-dose ASA prophylaxis.52,53
high for patients without symptomatic atherosclerotic cardiovascular Cardiovascular risk factors amplify prothrombotic diathesis, pro­
disease (ASCVD), according to the presence of multiple risk factors, mote atherosclerotic plaque progression,54 and may impair the im­
high lifetime risk for ASCVD, and documented significant atherosclerotic mune response during severe infections. Due to immune dysfunction
plaque burden (see Supplementary data online, Table S2).9 Patients with and high prevalence of antibiotic resistance, patients with type 2 DM
history of coronary revascularization without myocardial infarction (MI) have a high risk for infections and two to six times higher incidence
are referred to in the Patients on single antiplatelet therapy section. of sepsis vs. matched non-type 2 DM subjects.55,56
Although acetylsalicylic acid (ASA) should not be given routinely to pa­ Obesity is also characterized by a prothrombotic and low-grade in­
tients without history of symptomatic ASCVD,9 guidelines indicate that flammatory milieu.57 Whether obesity is an independent risk factor for
Severe infections and antithrombotic therapy 3045

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


Figure 2 Modulation of haemostasis by Staphylococcus aureus and Pseudomonas species infections. Severe bacterial infections can modulate haemo­
stasis via multiple mechanisms. Long-chain polyphosphates (≥500 phosphate units) produced by bacteria promote FXIIa-mediated activation of blood
coagulation. Polyphosphates also incorporate into fibrin clots and render the clots resistant to fibrinolysis. Staphylococcus aureus secretes coagulase and
von Willebrand binding protein which activate prothrombin to thrombin in a non-enzymatic manner. Staphylococcus aureus also secretes fibronectin-
binding protein A and clumping factor A which activate platelets. With respect to NETosis, Staphylococcus aureus secretes pore-forming toxins (such as
Panton–Valentine leucocidin) which induces a unique and rapid (5–60 min) form of NETosis that occurs in an oxidant-independent mechanism.
Elastase, a major virulence factor in Pseudomonas species, activates FXII in the coagulation cascade. Pseudomonas species also secrete protease IV which
inhibits fibrinolysis via the degradation of plasminogen and fibrinogen. The result of severe infections of Staphylococcus aureus and Pseudomonas species is
immunothrombosis, whose dysregulation may lead to vascular occlusion and thromboinflammation. Created with Biorender.com. vWbp, von
Willebrand binding protein; FnbpA, fibronectin-binding protein A; ClfA, clumping factor A; NETs, neutrophil extracellular traps.

sepsis,58 sepsis-associated mortality, and severe thromboses in sepsis, all patients with severe infections that are bedridden or have SIC, to re­
including SARS-CoV-2,59–61 remains controversial.58,62–64 duce the risk of VTE.70 Some settings with a high thrombosis burden,
The safety and efficacy of continuing low-dose ASA in patients with including SARS-CoV-2, may require higher than prophylactic doses
high-degree thrombocytopenia and severe infections have not been (intermediate or therapeutic).19 In each case, the use of heparins at
studied in randomized clinical trials (RCTs), since thrombocytopenia is prophylactic or higher doses should be weighed against the bleeding
an exclusion criterion in all RCTs on antithrombotic drugs. However, risk in the individual patient (Tables 2–4).
no increased bleeding risk was observed outside the ICU setting in can­
cer patients with ASCVD with platelet count <100 × 109/L who re­
ceived ASA (median platelet count in thrombocytopenic patients 32 ×
109/L), suggesting a favourable risk/benefit profile of ASA even in patients
with low-to-moderate degree thrombocytopenia.65,66 Interestingly, ASA Consensus statements Strength of advice
....................................................................................
exerts its antiinflammatory effects at doses higher than those used for
We advise that in patients who are already on
cardiovascular prevention,67 although some data may suggest some indir­
low-dose ASA because of high and very
ect antiinflammatory effect at lower doses.68 However, ASA started high cardiovascular risk due to multiple risk
during hospitalization for viral SARS-CoV-2 infections did not reduce factors, with no history of symptomatic
mortality vs. placebo despite significantly shortening hospitalization,69 ASCVD, low-dose ASA should be
even though some observational studies and meta-analyses suggested maintained, but may require adjustment
a survival benefit of antiplatelet treatment, mostly ASA, started during according to the course of the severe
hospitalization specifically in patients with severe infections or sepsis infection and degree of SIC and
(see Supplementary data online, Table S7). thrombocytopenia (Tables 2–4).19,71,72
Additional prophylactic anticoagulation, with low-molecular-weight Continued
(LMWH) or unfractionated (UFH) heparins, should be considered in
3046 Gigante et al.

Table 2 Management of antithrombotic therapy in patients with multiple cardiovascular risk factors and enhancers and
no history of symptomatic ASCVD

Severe infections where only Severe infections and Severe infections and Severe infections and platelet
preventive antithrombotic platelet count ≥20 × platelet count <20 × 109/L count <20 × 109/L in the
treatment is indicated 109/L presence of multiorgan failure
......................................................................................................................................................................................
During No routine antiplatelet therapy, SAPT No routine antiplatelet No antiplatelet therapy No antithrombotic treatment
as pre-existing indication therapy, SAPT as
pre-existing indication
Prophylactic anticoagulationa Prophylactic anticoagulation Prophylactic anticoagulation in
patients at high
thromboembolic risk

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


After SAPT as pre-existing indication SAPT as pre-existing SAPT as pre-existing indication SAPT as pre-existing indication
indication
No routine prophylactic No routine prophylactic No routine prophylactic No routine prophylactic
anticoagulationb anticoagulationb anticoagulationb anticoagulationb

ASCVD, atherosclerotic cardiovascular disease; SAPT, single antiplatelet therapy, usually low-dose aspirin.
a
Therapeutic dose anticoagulation may be considered in patients without high risk of bleeding.
b
Thromboprophylaxis may be considered in high-risk patients (persistent immobility, history of venous thromboembolism, advanced age, obesity, cancer, thrombophilia, increased
D-dimer concentrations, and high inflammatory activity) and low risk of bleeding.

no significant increases in bleeding up to 90 days (Table 5). Thus, with


Continued
the limitations of observational studies, current evidence does not
Consensus statements Strength of advice show harm and supports continuing SAPT during new-onset sepsis
.................................................................................... or severe infections (Tables 5 and S7).
Additional parenteral anticoagulation at Sepsis-induced coagulopathy is associated with high mortality and may
prophylactic or at higher doses depending benefit from prophylactic LMWH (Tables 3 and 4).40 Prophylactic
on the individual bleeding risk may be LMWH does not contraindicate SAPT in patients with previous
appropriate in patients with high and very
ASCVD. However, bleeding increases with the degree of thrombocyto­
high cardiovascular risk as needed
penia, especially for platelet counts <25 × 109/L, with bleeding rates of
according to SIC severity (Tables 3
∼15%/year in thrombocytopenic patients89 vs. ∼0.07%/year in healthy
and 4).73–76
subjects.90 Evidence of efficacy and safety of SAPT in thrombocytopenic
Temporary switching to parental cardiovascular patients is limited since these patients are excluded from
anticoagulation may be appropriate in cardiovascular trials. Acute ischaemic and bleeding events in patients
some situations where combination is not with thrombocytopenia of various origin are associated with four- to
justifiable (e.g. SIC with low platelet count five-fold increased mortality vs. non-thrombocytopenic subjects.89,91
and/or high bleeding risk). Acetylsalicylic acid used for secondary prevention did not significantly in­
crease major bleeding with platelet counts >20 × 109/L92 while with
≤20 × 10/9/L platelets, data do not support continuing SAPT (Tables 3
and 4). However, data on SAPT in severe thrombocytopenia and sepsis
Patients on single antiplatelet therapy are not available. Gastrointestinal bleeding during antithrombotic treat­
According to current guidelines, single antiplatelet therapy (SAPT), ment is preventable by gastroprotectant drugs including proton pump
mainly with low-dose ASA, is used for prevalent stable symptomatic inhibitors (PPIs),93,94 which should be used in sepsis, SIC, thrombocyto­
ASCVD or previous revascularization in the absence of history of penia, and/or prophylactic LMWH while on SAPT.66 Moreover, multiple
MI.9,51 ASCVD patients have a high susceptibility to viral77 and bacterial drugs (vasoactive, antibiotics, antivirals, and corticosteroids) are
infections78 and worse sepsis outcomes.79 Moreover, sepsis survivors co-administered during sepsis, potentially generating clinically relevant
appear at increased risk for future major vascular events.80 drug interactions.95 Since ASA is not biotransformed by the CYP450 en­
Observational studies on sepsis report that SAPT is associated with a zymes,96 no interactions are reported or can be anticipated, while the
better outcome (Table 5). Acetylsalicylic acid started before sepsis/sep­ P2Y12 inhibitor clopidogrel, a pro-drug with multiple CYP-dependent
tic shock has been associated with a reduction of mortality rates by 7% bioactivation steps and low bioavailability, is subjected to clinically rele­
[95% confidence interval (CI): 2%–12%, P = 0.005] in large observation­ vant interactions that may modify its antiplatelet effect (see
al studies85 while a prospective study reported reduced acute respira­ Supplementary data online, Table S6).97,98
tory distress syndrome with no mortality reduction.88 Expectedly, Recent ESC guidelines9,51 recommend PPIs in patients on SAPT (IA)
pre-sepsis ASA usage was largely associated with previous to reduce gastrointestinal bleeding.93,99–101 Some studies have sug­
ASCVD.85,88 A meta-analysis, including 10 large studies totalling 689 gested that in ICU patients, gastric acid suppression may further in­
897 patients hospitalized for sepsis,86 showed reduced mortality with crease the risk of penumonia.102 However, in the case of
SAPT (mostly ASA) started before sepsis. Available safety data report coagulopathy, as for SIC, the benefit of reducing major gastrointestinal
Severe infections and antithrombotic therapy 3047

Table 3 Management of antithrombotic drugs in patients with ongoing antithrombotic treatment, SOFA score of 1a,
and increasing SICa score

Patients on SAPT Patients on DAPT Patients on Patients on OAC


long-term OAC for recentb
thromboembolic
event
......................................................................................................................................................................................
SIC score = 2
Platelet count 100– No change <3 m after PCI or ACS: no No change No change
149 × 109/L change
or 3–6 m after PCI/ACS: a
PT ratio 1.2–1.4 P2Y12 inhibitor or ASA

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


≥6 m after PCI/ACS: ASA
or a P2Y12 inhibitor
SIC score = 3
Plateleat count 100– Platelet count ≥20 × <3 m after PCI or ACS: no Platelet count 100– Platelet count 100–
149 × 109/L and PT 109/L: no change change or consider P2Y12 149 × 109/L and PT 149 × 109/L and
ratio 1.2–1.4 inhibitor or ASA ratio 1.2–1.4: no PT ratio 1.2–1.4:
change no change
or Platelet count <20 × Consider ASA if platelet count Platelet count <100 × Platelet count 50–
Platelet count 109/L: stop SAPT <20 × 109/L 109/L 100 × 109/L
<100 × 109/L 3–6 m after PCI/ACS: P2Y12 Change VKA/ Change VKA/
or inhibitor or ASA DOAC to heparinc DOAC to
PT ratio >1.4 Consider ASA if platelet at prophylactic or therapeutic dosed
count <20 × 109/L intermediate dose heparinc, split into
≥6 m after PCI/ACS: ASA two daily doses
or clopidogrel
Consider no SAPT if platelet
count <20 × 109/L
SIC score = 5
Platelet count <100 × Platelet count ≥20 × <1 m after PCI or ACS: no Platelet count ≥30 × Platelet count ≥30 ×
109/L and PT ratio 109/L: no change change if platelet count ≥20 × 109/L: no change 109/L: no change
>1.4 Platelet count <20 × 109/L or consider P2Y12 Platelet count <30 × Platelet count 30–
109/L: stop SAPT inhibitor (clopidogrel) or ASA 109/L: stop heparinc 50 × 109/L:
Consider SAPT-ASA prophylactic dose
monotherapy if platelets heparinc
<20 × 109/L Platelet count <30 ×
1–3 m after PCI/ACS: P2Y12 109/L: stop heparinc
inhibitor (clopidogrel) or ASA
Consider SAPT–ASA
monotherapy if platelets
<20 × 109/L
≥3 m after PCI/ACS: ASA or
clopidogrel
Consider no APT if platelet
count <20 × 109/L

These treatment proposals should be considered in light of individual patient characteristics and may not be appropriate if the risk of life-threatening stent thrombosis is high or other
patient characteristics suggest that bleeding risk of DAPT or a more effective P2Y12 inhibitor (ticagrelor or prasugrel) outweighs the thrombotic risk.
ACS, acute coronary syndrome; APT, antiplatelet therapy; DAPT, dual antiplatelet therapy; OAC, oral anticoagulant; PT, prothrombin time; SAPT, single antiplatelet therapy; VKA, vitamin
K antagonist.
a
SIC and SOFA definitions and scores are shown in Table 1 and Supplementary data online, Table S3, respectively.
b
Recent thromboembolism refers to an event within the previous 3 months.
c
Heparins: low-molecular weight heparin (LMWH) at indicated dose for creatinine clearance >30 mL/min, with dose adjustment for creatinine clearance 15–30 mL/min; unfractionated
heparin (UFH) if creatinine clearance <15 mL/min.
d
Therapeutic dose LMWH should be reduced when the calculated creatinine clearance is <30–40 mL/min according to product monograph, or changed to UFH. If creatinine clearance
<15 mL/min, use only UFH.
3048 Gigante et al.

Table 4 Management of antithrombotic drugs in patients with ongoing antithrombotic treatment, SOFAa score of at
least 2, and increasing SICa score

Patients on SAPT Patients on DAPT Patients on Patients on OAC


long-term OAC due to recentb
thromboembolic
event
......................................................................................................................................................................................
SIC score = 3
Platelet count 100– No change <3 m after PCI or ACS: no No change No change
149 × 109/L change
or 3–6 m after PCI/ACS: consider
PT ratio 1.2–1.4 P2Y12 inhibitor or ASA (SAPT)

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


≥6 m after PCI/ACS: SAPT
SIC score = 4
Platelet count 100– Platelet count ≥20 × <1 m after PCI/ACS: no Platelet count 100– Platelet count 100–
149 × 109/L and PT 109/L: no change change or consider P2Y12 149 × 109/L and PT 149 × 109/L and PT
ratio 1.2–1.4 Platelet count <20 × inhibitor (clopidogrel) or ASA ratio 1.2–1.4: no ratio 1.2–1.4: no
or 109/L: stop SAPT (SAPT) change change
Platelet count Consider SAPT with ASA if Platelet count 50– Platelet count 50–
<100 × 109/L platelet count <20 × 109/L 100 × 109/L: change 100 × 109/L: change
or 1–3 m after PCI or ACS: no VKA/DOAC to VKA/DOAC to
PT ratio >1.4 change or P2Y12 inhibitor or therapeutic dosec therapeutic dosec
ASA heparind, split into two heparind, split into
Consider SAPT with ASA if daily doses two daily doses
platelet count <20 × 109/L
3–6 m after PCI/ACS: P2Y12
inhibitor or ASA
Consider SAPT with ASA if
platelets <20 × 109/L
≥6 m after PCI/ACS: SAPT
Consider no SAPT if platelet
count <20 × 109/L
SIC score = 6
Platelet count Platelet count ≥20 × <1 m after PCI or ACS: no Platelet count 30– Platelet count 30–
<100 × 109/L and PT 109/L: no change change if platelets ≥20 × 109/ 50 × 109/L: 50 × 109/L:
ratio >1.4 Platelet count <20 × L; consider P2Y12 inhibitor prophylactic dose prophylactic dose
109/L: stop SAPT (clopidogrel) or ASA (SAPT) if heparind; platelet heparind; platelet
platelets >20 × 109/L count <30 × 109/L: count <30 × 109/L:
Consider SAPT with ASA if stop heparin stop heparin
platelets <20 × 109/L
1–3 m after PCI/ACS: P2Y12
inhibitor (clopidogrel) or ASA
Consider SAPT with ASA if
platelet count <20 × 109/L
≥3 m after PCI/ACS: SAPT
Consider no SAPT if platelet
count <20 × 109/L

These treatment proposals should be considered in light of individual patient characteristics and may not be appropriate if the risk of life-threatening stent thrombosis is high or other
patient characteristics suggest that bleeding risk of DAPT or a more effective P2Y12 inhibitor (ticagrelor or prasugrel) outweighs the thrombotic risk.
ACS, acute coronary syndrome; APT, antiplatelet therapy; DAPT, dual antiplatelet therapy; OAC, oral anticoagulant; PT, prothrombin time; SAPT, single antiplatelet therapy; VKA, vitamin
K antagonist.
a
SIC and SOFA definitions and scores are provided in Table 1 and Supplementary data online, Table S3, respectively.
b
Recent thromboembolism refers to an event within the previous 3 months.
c
Therapeutic dose LMWH should be reduced when the calculated creatinine clearance is <30–40 mL/min according to product monograph, or changed to UFH. If creatinine clearance
<15 mL/min, use only UFH.
d
Heparins: low-molecular weight heparin (LMWH) at indicated dose for creatinine clearance >30 mL/min, with dose adjustment for creatinine clearance 15–30 mL/min; unfractionated
heparin (UFH) if creatinine clearance <15 mL/min.
Severe infections and antithrombotic therapy 3049

Table 5 Studies in patients on single or dual antiplatelet therapy before hospitalization for severe infections or sepsis

Study (year) Design Population Comparison and Effectiveness Safety Other remarks
type of antiplatelet
agent(s)
......................................................................................................................................................................................
Erlich et al. Multicentre 161 hospitalized Pre-hospitalization ASA users: lower NA
(2011)81 observational, patients at risk for SAPT–ASA users: acute limb ischaemia
prospective study acute lung injury, 79 OR 0.37 (95% CI:
∼30% due to sepsis 0.16–0.84) P = 0.02
or infection No difference in
mortality or length
of hospitalization.

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


Losche et al. Retrospective Cohort 1: 224 patients Cohort 1, n (%): Cohort 1: ICU Cohort 2: no increase Similar benefit also
(2012)82 analysis of two hospitalized for SAPT–ASA: 38 (17); admission (%) 9.1 vs. of bleeding after correcting
cohorts pneumonia SAPT–clopidogrel: 9 26.3% associated with for SOFA score,
Cohort 2: 615 patients (4) Length of stay 13.9 previous APT use age, gender, and
admitted to the ICU Cohort 2, n (%): ± 6.2 vs. 18.2 ± 10.2 C-reactive
for sepsis SAPT–ASA: 129 (21); days in APT users vs. protein levels
SAPT or DAPT non-users
with clopidogrel: 25 (4) Cohort 2: OR for
total mortality 0.19
(95% CI: 0.12–0.33)
for APT use vs.
non-use; OR 0.20
(95% CI: 0.12–0.33)
for ASA users vs.
non users
Valerio-Rojas Retrospective 651 ICU patients, 272 Comparison of prior Mortality: (a)OR 0.76 Red blood cell Platelet count
et al. (42.8%) on APT antiplatelet vs. no (95% CI: 0.52–1.10) transfused units: not significantly
(2013)83 SOFA score day 1: antiplatelet ARDS: (a)OR 0.50 different among higher in the APT
no APT median SAPT–ASA: n = 241 (95% CI: 0.35–0.71) groups group
(IQR) 6 (4–10) and SAPT–clopidogrel: Mechanical Platelet-transfused PT and INR
APT 6 (4–9), P = ns n=4 ventilation (a)OR patients higher in significantly
DAPT with 0.62 (95% CI: 0.45– the no APT group lower in the APT
clopidogrel: n = 27 0.87) (P = 0.01) group
Tsai et al. Nationwide 683 421 patients Use of antiplatelet Mortality: (a)OR 0.82, NA Benefit higher in
(2015)84 population-based hospitalized for agents before (95% CI: 0.81–0.83, current users
cohort and nested sepsis admission P = 0.001) (aOR 0.78, 95%
case–control Antiplatelets: aspirin CI 0.76–0.79);
study (n = 51 340), non-significant in
clopidogrel or past users (aOR
ticlopidine (13 368), 1.00, 95% CI
prescribed within 0.98–1.02)
1 year before the
index date
Trauer et al. Meta-analysis of 11 n = 6823 ICU patients Comparison of prior Mortality: ASA users: NA
(2017)85 observational hospitalized for aspirin or APT usage significant reduction
studies sepsis vs. no usage by 7% (95% CI: 2%–
Antiplatelet 12%), P = 0.005
included SAPT– APT users:
ASA, SAPT significantly reduced
clopidogrel, or by 6% (95%CI: 0%–
DAPT aspirin plus 12%), P = 0.004
clopidogrel
Ouyang et al. Meta-analysis of 10 689 897 patients SAPT–ASA only used Mortality OR: 0.78 NA A subgroup on
(2019)86 studies hospitalized for in 5 out of 10 studies (95% CI: 0.77–0.80) timing of APT
sepsis SAPT–ASA: OR showed benefit
121 147 on APT for APT
Continued
3050 Gigante et al.

Table 5 Continued

Study (year) Design Population Comparison and Effectiveness Safety Other remarks
type of antiplatelet
agent(s)
......................................................................................................................................................................................
(SAPT–ASA, SAPT– 0.60 (95% CI: 0.53– administered
clopidogrel, DAPT 0.68) also after
clopidogrel, or hospitalization
prasugrel)
Chow et al. Observational, 1:2 17 347 patients Pre-hospital In-hospital mortality Epistaxis (%): 0.9 vs. 0.4 Blood transfusion
(2021)87 propensity score hospitalized for antiplatelet therapy (%): 18.9 with vs. in users vs. no users. higher in
matched for COVID-19: 6781 on vs. no antiplatelet 21.5 with and No difference in GI non-users (4%

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


demographics and pre-hospital therapy without or IC bleeding vs. 2.8%, P <
comorbidities antiplatelet drugs SAPT–ASA: 83.9% pre-hospital APT 0.001)
hospital mortality PE Mean platelet count: SAPT–clopidogrel (HR 0.81; 95% CI: Subgroup
210 (160–275) 8.2% 0.76–0.87) analysis
×103/µL DAPT: 7.4% PE: 2.2% vs. 3% in underpowered
users vs. non-users to detect
(P = 0.002) differences
Subgroup analysis: between SAPT–
lower mortality in ASA vs. SAPT–
SAPT–ASA only clopidogrel vs.
No mortality DAPT
reduction for
SAPT–clopidogrel
or DAPT

APT, antiplatelets; ASA, acetylsalicylic acid; ARDS, acute respiratory distress syndrome; CI, confidence interval; DAPT, dual antiplatelet therapy; HR, hazard ratio; GI, gastrointestinal; IC,
intracerebral; ICU, intensive care unit; INR, international normalized ratio; IQR, interquartile range; (a)OR, (adjusted) odds ratio; NA, not available; PE, pulmonary embolism; PT,
prothrombin time; SAPT, single antiplatelet therapy.

bleeding seems higher than the risk of pneumonia, and PPIs seemed Patients on dual antiplatelet therapy
more effective than histamine H2 receptor antagonists, although When a patient develops severe infection while on dual antiplatelet
more randomized trials are needed.94,103 If a PPI is administered, espe­ therapy (DAPT) consisting of ASA and a P2Y12 inhibitor, large observa­
cially in severely thrombocytopenic patients on clopidogrel, omepra­ tional studies support the safety of continuing DAPT in this setting
zole and esomeprazole should be avoided due to potential and
(Table 5), but evidence from randomized studies about the balance of
clinically relevant drug–drug interactions (see Supplementary data
efficacy and safety, especially as SIC progresses, is lacking.87,109,110
online, Table S6).104–107
Few observational and post hoc analyses suggested that DAPT with clo­
pidogrel may be associated with an increased risk of infection vs. ASA
alone after coronary artery bypass surgery,111 and that DAPT with tica­
grelor may be associated with a lower risk of bacterial lung infec­
tion.112–115 A weak off-target clopidogrel effect with a minor decrease
Consensus statements Strength of advice
.................................................................................... on leucocyte count has been described, but without any impact on rates
We advise to continue SAPT in patients with a of neutropenia.116,117 On the other hand, platelet P2Y12 inhibition has
clear, pre-existing indication in secondary been inconsistently reported to reduce systemic inflammatory response
cardiovascular prevention during new-onset in animal and human models of endotoxemia,118–121 but the clinical rele­
sepsis or severe infections, unless severe SIC
vance remains unknown. Furthermore, SAPT with either ticagrelor or clo­
with platelet count ≤20 × 109/L occurs.71,86,88
pidogrel, in addition to standard anticoagulant therapy, failed to improve
We advise to check for clinically relevant drug clinical outcomes in non-critically ill hospitalized COVID-19 patients.122
interactions with antimicrobials when For most patients treated with ASA and clopidogrel, ASA or clopido­
clopidogrel or ticagrelor is used and with grel123,124 may be stopped, and SAPT was used starting from 3 to 6
PPIs when clopidogrel is used.107,108 months after percutaneous coronary intervention (PCI) and/or acute
coronary syndrome (ACS), without significant stent thrombosis risk
We advise to treat patients with high degree of
SIC, including thrombocytopenia, with (Tables 3 and 4).123–126
gastroprotective agents, preferably For DAPT-treated patients who develop an indication for full-dose
PPIs.19,93,99,102,103 anticoagulant therapy during sepsis, triple antithrombotic therapy
(DAPT plus an anticoagulant drug) carries a high bleeding risk, while
dual antithrombotic therapy with clopidogrel and one anticoagulant
Severe infections and antithrombotic therapy 3051

Table 6 Studies in patients with anticoagulant therapy during severe infections or sepsis

Study (year) Study type Population Comparison or Efficacy outcome Safety Other remarks
intervention outcome
......................................................................................................................................................................................
Umemura Meta-analysis of 14 767 patients Anticoagulants vs. Mortality overall, RR 0.97 Bleeding Only three of the
et al. 24 RCTs with sepsis, control (95% CI: 0.92–1.02) complications studies used
(2016)137 treated with any Subgroup with DIC, overall, RR heparins
anticoagulant, of RR 0.72 (95% CI: 0.62– 1.33 (95% CI:
those 3206 had 0.85) 1.12–1.57)
DIC Subgroup with
DIC, RR 1.26
(95% CI: 0.86–

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


1.85)
Wang et al. Meta-analysis of Patients with sepsis UFH or LMWH vs. 28-day mortality, OR Bleeding A variety of heparin
(2014)138 nine controlled (592) or severe control 0.66 (95% CI: 0.56– complications, dose regimens
trials sepsis (3011) 0.77) OR 1.06 (95% were used
CI: 0.83–1.36)
Li et al. Meta-analysis of 684 adult patients LMWH at mainly 28-day mortality, RR 0.52 Bleeding All studies were small
(2021)139 10 RCTs with sepsis intermediate dose (95% CI: 0.38–0.70) complications, with 32–159
vs. control APACHE II score RR 1.29 (95% patients
mean difference −4.42 CI: 0.76–2.17)
(95% CI: −5.50 to
−3.33
Søgaard Nationwide 3030 patients with Warfarin at the time 90-day all-cause Bleeding No information on
et al. healthcare AF on warfarin of sepsis diagnosis mortality, HR 0.64 complications, how
(2017)140 database. vs. 55 721 vs. no warfarin. (95% CI: 0.58–0.69); 1.19 (95% CI: anticoagulation
Inverse patients without Warfarin effect thromboembolism, 1.00–1.41) was managed
probability of AF and warfarin, will remain 3–5 1.25 (95% CI: 0.89– during
treatment all admitted for days after stopping 1.76) hospitalization. No
weighting sepsis data on sepsis
severity

APT, antiplatelet drugs; ASA, acetylsalicylic acid; ARDS, acute respiratory distress syndrome; CI, confidence interval; DAPT, dual antiplatelet therapy; DIC, disseminated intravascular
coagulation; HR, hazard ratio; GI, gastrointestinal; IC, intracerebral; ICU, intensive care unit; INR, international normalized ratio; IQR, interquartile range; LMWH,
low-molecular-weight heparin; NA, not available; (a)OR, (adjusted) odds ratio; PE, pulmonary embolism; PT, prothrombin time; RCT, randomized clinical trial; RR, relative risk;
SAPT, single antiplatelet therapy; UFH, unfractionated heparin.

drug may offer a safer strategy, even early after PCI and/or ACS,127 pro­ secondary haemostasis (see Supplementary data online, Figure S2).
vided a normal platelet count or a mild degree of thrombocytopenia, Consequently, antithrombotic therapy should be carefully tailored in
although this strategy has never been tested in severe infection devel­ DAPT-treated patients with severe infection who require therapeutic
oping SIC. Moreover, the safety vs. efficacy of a dual antithrombotic anticoagulation or develop coagulopathy.
therapy with ASA, as compared with clopidogrel or ticagrelor, has Current ESC guidelines9,51 recommend PPIs94 in patients on DAPT to
not been tested in a contemporary setting nor in severe infections reduce the risk of gastrointestinal bleeding19,99; however, caution should
and sepsis with or without SIC. Thus, in patients with sepsis or severe be used in critically ill patients,102 as detailed in the previous section.
infections with an indication for DAPT, who need full anticoagulant
doses, it is safe to stop DAPT and continue with one antiplatelet agent,
either clopidogrel or aspirin depending on the severity of SIC,
thrombocytopenia, and co-medication, also earlier after PCI or ACS Consensus statements Strength of advice
(Table 5), although this strategy has never been tested in sepsis and/ ....................................................................................
or in severely thrombocytopenic patients (Tables 3 and 4). We advise to continue DAPT following recent (i.e.
Importantly, in patients with severe sepsis on ticagrelor or clopidogrel, up to 6 months) ACS with or without PCI, in
potential clinically relevant drug–drug interactions with antimicrobial patients with platelet count >50 × 109/L.81–87
agents should always be considered due to their CYP450-dependent bio­
transformation (see Supplementary data online, Table S7).128 Moreover,
Continuing DAPT if platelet count is 30–50 ×
∼25% of patients on ticagrelor develop drug-related dyspnoea,129,130
109/L may be appropriate unless coagulation is
which may require differential diagnosis.130 Caution is required with clopi­
severely impaired, or there is a high bleeding
dogrel if hepatic dysfunction develops during severe infection as this can risk. In the latter cases, we advise to switch
reduce its antiplatelet effect.107 Ticagrelor is also contraindicated in severe to SAPT with clopidogrel or low-dose
hepatic impairment due to risk of accumulation and bleeding.108 ASA.69,81–87,133–136
Since thrombin is also a potent platelet activator, anticoagulants
Continued
may also diminish platelet activation,131,132 on top of impairing
3052 Gigante et al.

There is a paucity of data reporting management of patients with se­


Continued
vere infections on long-term VKA. Further, with the high mortality,
Consensus statements Strength of advice morbidity, and coagulopathy associated with sepsis and severe infec­
.................................................................................... tions, interpretation of the few reports based on single-centre observa­
Continuing SAPT with ASA may be appropriate tional experience is challenging. A systematic review identified eight
in patients with more severe cases of SARS-CoV-2–related prosthetic heart valve thrombosis, with­
thrombocytopenia and at higher risk of major out evidence for non-adherence to the VKA regimen.71 Intensified
vascular event recurrence, on a case-by-case
monitoring of INR or switch to intravenous heparin during hospitaliza­
basis.66,81–87
tion might reduce this risk (Tables 3 and 4).
Continue with one antiplatelet agent (ASA or a There is a lack of studies on patients with recent VTE and sepsis.
P2Y12 inhibitor, preferably clopidogrel) may be Generally, the risk of recurrent VTE decreases with time. For those
appropriate when patients receive therapeutic with VTE within 1 month, especially with pulmonary embolism, the bene­
dose of anticoagulation or develop higher fit of full-dose anticoagulation probably outweighs the bleeding risk. By

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


degree of SIC (>3) with PT >1.4. splitting the therapeutic dose of LMWH into two daily doses, high
We advise to check for relevant drug–drug peak concentrations are avoided. With longer intervals from the VTE,
interactions for ticagrelor and clopidogrel and intermediate-dose LMWH is probably sufficient (Tables 3 and 4).
ongoing antiviral or antibiotic agents.107,108 Clinically relevant drug–drug interaction should be considered with each
OAC, either VKA or DOAC, based on P-glycoprotein- and/or
CYP450-related biotransformation (see Supplementary data online,
We advise to treat patients with gastroprotective Table S7). Drugs that compete as substrates, inhibitors, or enhancers of
agents, preferably PPIs.19,93,99,102,103 their activity may affect warfarin efficacy and/or safety; thus, INR monitoring
should be more frequent to adjust dosing (Graphical Abstract). Dabigatran
is contraindicated with strong P-glycoprotein inhibitors including azole anti­
mycotics, some antivirals,142 while caution should be exerted with clarithro­
mycin.142 Edoxaban dosing should be reduced with strong P-glycoprotein
inhibitors,143 and rivaroxaban144 and apixaban145 are not recommended
with drugs that are strong inhibitors of both P-glycoprotein and CYP3A4
(azole antimycotics and HIV protease inhibitors).
Patients on oral anticoagulants Current ESC guidelines9,51 recommend the use of PPIs94 in patients
Anticoagulants, including antithrombin and soluble thrombomodulin treated with OAC (IA)146,147; however, caution should be used in crit­
(see Supplementary data online, Figure S3), may improve outcomes in ically ill patients102 as stated above.
SIC by reducing organ dysfunction, especially with pre-existing coagulo­
pathy.1 In a meta-analysis of 24 trials (14 767 patients) addressing vari­
ous anticoagulants in sepsis, reduced mortality was only observed in
those with sepsis-associated DIC.137 However, there was an increased
Consensus statements Strength of advice
risk for bleeding, and only five studies used heparin as anticoagulant. A ....................................................................................
meta-analysis of nine studies with prophylactic dose UFH or LMWH in For patients on oral anticoagulation for stroke
sepsis demonstrated a significant reduction in mortality without a sig­ prophylaxis in AF in case of severe infection,
nificant increase in bleeding risk.138 Furthermore, in a meta-analysis we advise to switch to low- or intermediate-
of 10 trials of specifically LMWH, in 8 of which at intermediate dose dose LMWH when platelets drop between
(684 patients), reduced 28-day mortality was again observed without 100 and 30 × 109/L (only if INR is <2.0 for
increased bleeding (Table 6).139 those on a VKA).138,139
Outcomes for patients with atrial fibrillation (AF) receiving warfarin and We advise to interrupt anticoagulant treatment
diagnosed with sepsis were analysed from a Danish national database, using for platelet count <30 × 109/L. For patients
non-anticoagulated patients without AF as controls.140 Bleeding complica­ with VTE, the dose of heparin should be
tions were marginally higher, and mortality was lower in the AF group, but based on the severity of the infection and
interpretation is hampered by the 90-day observation period, thus well be­ whether the indication is a recent
yond the phase of critical care. A retrospective cohort study addressed the thromboembolic event.66
anticoagulant management of 115 septic patients with AF in the ICU, of
For patients with VTE within 1 month, it may be
whom 34 continued warfarin or heparin during hospitalization.141 Time appropriate to remain on full anticoagulation,
in therapeutic range with warfarin was <50% (Table 6). preferably with LMWH, split into two daily
Anticoagulated patients had similar survival rates and more bleeding doses.
complications. Current knowledge of risk/benefit of direct oral anticoa­
gulants (DOAC) in AF and SIC is limited. Oral anticoagulations are gen­ We advise to check for clinically relevant drug–
erally unsuitable for patients in ICU due to vomiting, gastroparesis, drug interactions in patients treated with
unpredictable absorption, and need for frequent cessation due to inva­ OAC based on ongoing antimicrobial
sive procedures. International normalized ratio (INR) values are specif­ treatment and on the specific anticoagulant
ically intended for VKA monitoring and are not informative for other drug in use.148
anticoagulants. The PT ratio thresholds in the SOFA score are not help­
Continued
ful for decision-making when OACs are used.
Severe infections and antithrombotic therapy 3053

Continued

Consensus statements Strength of advice Consensus statements Strength of advice


.................................................................................... ....................................................................................
We advise to treat patients with high degree of We advise to vaccinate against influenza
SIC with gastroprotective agents, preferably patients at high and very high risk of ASCVD
PPIs.19,93,99,102,103 or with previous ASCVD.51,160,171

In patients with a long-term VKA indication, we It may be appropriate to reinforce the use of
advise to intensify INR monitoring or switch vaccinations for patients at high and very high
to heparin during hospitalization.71,141 risk of ASCVD or with previous ASCVD to
reduce the risk of severe infections and the
consequent possible associated SIC.

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


Vaccination in patients at high and Conclusions and future directions
very high cardiovascular risk The clinical course of coagulopathy associated with severe infections
should guide the management of patients with a previous clear indi­
Vaccinations decrease the risk of ASCVD, though with different cation for single or combined antithrombotic agents due to previous
effectiveness. Influenza vaccine, regardless of formulation and obser­ thrombotic disorders or at high thrombotic risk (Tables 3 and 4
vation period, reduced the risk of hospitalization for stroke, heart fail­ and Graphical Abstract). Antithrombotic therapy should be main­
ure,149 ACS, peripheral artery disease,122,150 all-cause, and tained throughout the hospitalization, taking into account the pro­
cardiovascular mortality149,151,152 in high-risk patients. The relative gression toward SIC and the degree of thrombocytopenia
effectiveness of high vs. standard dose of quadrivalent influenza vac­ (Graphical Abstract).
cine in preventing cardiovascular and pulmonary diseases at a popu­ Some limitations need to be acknowledged. This document does not
lation level is under investigation in a randomized, registry-based consider patients on mechanical circulatory devices, a common condi­
trial in elderly Finnish citizens.153 In patients with recent ACS, influ­ tion in sepsis patients in ICU, and patients requiring acute revasculari­
enza vaccine reduced the risk for all-cause and cardiovascular mortal­ zation. It does not include management of antithrombotic therapy
ity,154,155 regardless of vaccine formulation.150 An early vaccination during severe fungal and parasite infections.
with double-dose influenza vaccination does not seem to add benefits However, in patients on antithrombotic therapy who develop severe
in terms of prognosis in ACS patients156 and, in a small clinical trial, infections or sepsis, mortality remains high despite multiple considera­
the risk of hospitalization for ACS, heart failure, and stroke.157 In pa­ tions of clinical management due to the complex inflammatory and
tients with chronic coronary syndrome, a trend towards a reduction other immunomodulatory effects of sepsis.
in cardiovascular mortality and incidence of cardiovascular complica­ Beyond early therapy of the underlying causative infection, the un­
tions was observed.154,158,159 The ESC guidelines recommend influ­ met need is the task of improving therapeutic modalities, specifically fo­
enza vaccine in elderly patients with chronic coronary syndromes cusing on novel methods for attenuating thromboinflammation in this
(IB).51 In a recent meta-analysis, influenza vaccine reduced the risk critically ill patient population.
of ASCVD, cardiovascular, and all-cause mortality in patients with
ASCVD.160 The addition of pneumococcal polysaccharide vaccine
to influenza vaccine reduced the incidence of ACS, ischaemic stroke,
and heart failure hospitalization.161 However, in observational stud­
Supplementary data
ies, no consistent benefits of pneumococcal polysaccharide vaccine Supplementary data are available at European Heart Journal online.
alone have been observed,133,162,163 with a possible beneficial effect
on the risk for ACS, but not ischaemic stroke.164 No clinical trial or
large longitudinal study has assessed the effect of influenza vaccine Declarations
on the risk of incident VTE or AF. Recently, SARS-CoV-2 vaccines
have been given worldwide and long follow-up may reveal long-term Disclosure of Interest
cardiovascular effects. Vaccine-induced immune thrombotic A.B.: consultant fee from MSD Italy, investigator-initiated grant to the
thrombocytopenia is a rare (1:50 000–100 000)165 but often fatal institution from GSK Spa, research grant from Nordic Pharma Srl,
autoimmune adverse reactions to SARS-CoV-2 vaccinations and travel grant from Pfizer srl.
(ChAdOx1 and Ad26.COV2.S) characterized by coagulopathy, acute C.C.: speaker fees from Astra Zeneca, Boehringer Ingelheim, Bayer,
arterial and venous thrombosis, and bleeding.166–169 Notably, differ­ Bristol Myers Squibb, and Orion Pharma outside the submitted work.
ent mechanisms underlie vaccine-induced immune thrombotic J.L.F.: speaker fees from Eli Lilly Co, Daiichi Sankyo, Inc., AstraZeneca,
thrombocytopenia vs. other thromboembolic complications during Roche Diagnostics, Pfizer, Abbott, Ferrer, Rovi, Boehringer Ingelheim,
SARS-CoV-2 infection.170 and Bristol-Myers Squibb; consulting fees from AstraZeneca, Eli Lilly
3054 Gigante et al.

Co., Ferrer, Boston Scientific, Pfizer, Boehringer Ingelheim, Daiichi 9. Visseren FLJ, Mach F, Smulders YM, Carballo D, Koskinas KC, Bäck M, et al. 2021 ESC
Sankyo, and Bristol-Myers Squibb; and research grant from AstraZeneca. guidelines on cardiovascular disease prevention in clinical practice. Eur Heart J 2021;42:
3227–3337. https://doi.org/10.1093/eurheartj/ehab484
T.G.: personal fees from AstraZeneca, Boehringer Ingelheim, Pfizer, 10. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al.
Boston Scientific, and Abbott; grants and personal fees from Bayer The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3).
Healthcare, Bristol Myers Squibb, Daiichi Sankyo, Eli Lilly, and JAMA 2016;315:801–810. https://doi.org/10.1001/jama.2016.0287
Medtronic outside of the submitted work; supported by the German 11. Iba T, Arakawa M, Di Nisio M, Gando S, Anan H, Sato K, et al. Newly proposed
sepsis-induced coagulopathy precedes international society on thrombosis and
Research Foundation (DFG) KFO-274-Project number 190538538, haemostasis overt-disseminated intravascular coagulation and predicts high mortality.
by the German Research Foundation (DFG)-Project number J Intensive Care Med 2020;35:643–649. https://doi.org/10.1177/0885066618773679
374031971-TRR 240, and funded by the German Research 12. Iba T, Arakawa M, Levy JH, Yamakawa K, Koami H, Hifumi T, et al. Sepsis-induced coa­
Foundation (DFG)-Project number 335549539-GRK2381. gulopathy and Japanese association for acute medicine DIC in coagulopathic patients
with decreased antithrombin and treated by antithrombin. Clin Appl Thromb Hemost
J.H.L.: steering or advisory committees for Octapharma, Merck, and
2018;24:1020–1026. https://doi.org/10.1177/1076029618770273
Werfen. 13. Yamakawa K, Yoshimura J, Ito T, Hayakawa M, Hamasaki T, Fujimi S. External validation
E.L.: speaker’s fees from Novartis and Novo Nordisk. of the two newly proposed criteria for assessing coagulopathy in sepsis. Thromb

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


B.R.: speaker fee from Sobi (Sweden), consultant fee from Aboca Haemost 2019;119:203–212. https://doi.org/10.1055/s-0038-1676610
14. Ding R, Wang Z, Lin Y, Liu B, Zhang Z, Ma X, et al. Comparison of a new criteria for
(Italy), and investigator-initiated grant to the institution from Bayer
sepsis-induced coagulopathy and International Society on Thrombosis and
AG for a study on rivaroxaban. Haemostasis disseminated intravascular coagulation score in critically ill patients
S.S.: research grant from Octapharma and honoraria from Bayer, with sepsis 3.0: a retrospective study. Blood Coagul Fibrinolysis 2018;29:551–558.
Boehringer Ingelheim, Bristol-Myers Squibb, Daiichi-Sankyo, and Sanofi. https://doi.org/10.1097/mbc.0000000000000755
R.F.S.: research grants and personal fees from AstraZeneca, 15. Vincent JL, Moreno R, Takala J, Willatts S, De Mendonça A, Bruining H, et al. The SOFA
(Sepsis-related Organ Failure Assessment) score to describe organ dysfunction/failure.
Cytosorbents, GlyCardial Diagnostics, and Thromboserin and personal On behalf of the Working Group on Sepsis-Related Problems of the European Society
fees from Alnylam, Bayer, Bristol Myers Squibb/Pfizer, Chiesi, CSL of Intensive Care Medicine. Intensive Care Med 1996;22:707–710. https://doi.org/10.
Behring, Daiichi Sankyo, HengRui, Idorsia, Intas Pharmaceuticals, 1007/bf01709751
Medscape, Novartis, PhaseBio, Portola, and Sanofi Aventis. 16. Lyons PG, Micek ST, Hampton N, Kollef MH. Sepsis-associated coagulopathy severity
predicts hospital mortality. Crit Care Med 2018;46:736–742. https://doi.org/10.1097/
J.T.: speaker honoraria from Bayer, BMS Pfizer, Boehringer, and
CCM.0000000000002997
Daichii Sankyo. 17. Rowe G TWG. The Delphi technique as a forecasting tool: issues and analysis. Int J
H.t.C.: research support from Bayer; consultancy fees from Forecasting 1999;15:353–375. https://doi.org/10.1016/S0169-2070(99)00018-7
AstraZeneca, Leo, Galapagos, Novostia, and Alveron; shareholder 18. Task Force for the management of COVID-19 of the European Society of Cardiology.
ESC guidance for the diagnosis and management of cardiovascular disease during the
Coagulation Profile; and all revenues deposited at the CARIM School
COVID-19 pandemic: part 2—care pathways, treatment, and follow-up. Eur Heart J
for Cardiovascular Diseases, Maastricht University. 2021;43:1059–1103. https://doi.org/10.1093/eurheartj/ehab697
B.G., M.-L.B.-P., M.B., G.V., E.v.G., and P.C.L. have no conflict of inter­ 19. Schulman S, Sholzberg M, Spyropoulos AC, Zarychanski R, Resnick HE, Bradbury CA,
est to report. et al. ISTH guidelines for antithrombotic treatment in COVID-19. J Thromb Haemost
2022;20:2214–2225. https://doi.org/10.1111/jth.15808
20. Martinod K CD. Immunothrombosis and thromboinflammation in host defense and
Data Availability disease. Platelets 2021;32:314–324. https://doi.org/10.1080/09537104.2020.1817360
21. Tanguay JF, Geoffroy P, Sirois MG, Libersan D, Kumar A, Schaub R, et al. Prevention of
No new data were generated or analysed in support of this research.
in-stent restenosis via reduction of thrombo-inflammatory reactions with recombin­
ant P-selectin glycoprotein ligand-1. Thromb Haemost 2004;91:1186–1193. https://
Funding doi.org/10.1160/th03-11-0701
22. Tomashefski JF Jr, Davies P, Boggis C, Greene R, Zapol WM, Reid L. The pulmonary
All authors declare no funding for this contribution. vascular lesions of the adult respiratory distress syndrome. Am J Pathol 1983;112:
112–126.
23. Levy JH, Iba T, Olson LB, Corey KM, Ghadimi K, Connors JM. COVID-19: thrombosis,
References thromboinflammation, and anticoagulation considerations. Int J Lab Hematol 2021;43:
1. Iba T, Levi M, Levy JH. Sepsis-induced coagulopathy and disseminated intravascular co­ 29–35. https://doi.org/10.1111/ijlh.13500
agulation. Semin Thromb Hemost 2020;46:89–95. https://doi.org/10.1055/s-0039- 24. Stark K, Massberg S. Interplay between inflammation and thrombosis in cardiovascular
1694995 pathology. Nat Rev Cardiol 2021;18:666–682. https://doi.org/10.1038/s41569-021-
2. Jackson SP, Darbousset R, Schoenwaelder SM. Thromboinflammation: challenges of 00552-1
therapeutically targeting coagulation and other host defense mechanisms. Blood 25. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity.
2019;133:906–918. https://doi.org/10.1182/blood-2018-11-882993 Nat Rev Immunol 2013;13:34–45. https://doi.org/10.1038/nri3345
3. Delabranche X, Helms J, Meziani F. Immunohaemostasis: a new view on haemostasis 26. Meyers S, Crescente M, Verhamme P, Martinod K. Staphylococcus aureus and neutro­
during sepsis. Ann Intensive Care 2017;7:117. https://doi.org/10.1186/s13613-017-
phil extracellular traps: the master manipulator meets its match in immunothrombosis.
0339-5
Arterioscler Thromb Vasc Biol 2022;42:261–276. https://doi.org/10.1161/atvbaha.121.
4. Data Bridge Market Research. Global Antithrombotic Drugs Market—industry trends
316930
and forecast to 2029. https://www.databridgemarketresearch.com/reports/global-
27. Iba T, Levy JH. Inflammation and thrombosis: roles of neutrophils, platelets and endo­
antithrombotic-drugs-market.
thelial cells and their interactions in thrombus formation during sepsis. J Thromb
5. Adelborg K, Grove EL, Sundbøll J, Laursen M, Schmidt M. Sixteen-year nationwide
Haemost 2018;16:231–241. https://doi.org/10.1111/jth.13911
trends in antithrombotic drug use in Denmark and its correlation with landmark stud­
28. Zhu Y, Chen X, Liu X. NETosis and neutrophil extracellular traps in COVID-19: im­
ies. Heart 2016;102:1883–1889. https://doi.org/10.1136/heartjnl-2016-309402
6. García Rodríguez LA, Cea Soriano L, de Abajo FJ, Valent F, Hallas J, Gil M, et al. Trends munothrombosis and beyond. Front Immunol 2022;13:838011. https://doi.org/10.
in the use of oral anticoagulants, antiplatelets and statins in four European countries: a 3389/fimmu.2022.838011
population-based study. Eur J Clin Pharmacol 2022;78:497–504. https://doi.org/10. 29. Wuillemin WA, Fijnvandraat K, Derkx BH, Peters M, Vreede W, ten Cate H, et al.
1007/s00228-021-03250-6 Activation of the intrinsic pathway of coagulation in children with meningococcal sep­
7. Iba T, Umemura Y, Wada H, Levy JH. Roles of coagulation abnormalities and micro­ tic shock. Thromb Haemost 1995;74:1436–1441. https://doi.org/10.1055/s-0038-
thrombosis in sepsis: pathophysiology, diagnosis, and treatment. Arch Med Res 2021; 1649961
52:788–797. https://doi.org/10.1016/j.arcmed.2021.07.003 30. Jansen PM, Pixley RA, Brouwer M, de Jong IW, Chang AC, Hack CE, et al. Inhibition of
8. Iba T, Levy JH, Warkentin TE, Thachil J, van der Poll T, Levi M. Diagnosis and manage­ factor XII in septic baboons attenuates the activation of complement and fibrinolytic
ment of sepsis-induced coagulopathy and disseminated intravascular coagulation. J systems and reduces the release of interleukin-6 and neutrophil elastase. Blood 1996;
Thromb Haemost 2019;17:1989–1994. https://doi.org/10.1111/jth.14578 87:2337–2344. https://doi.org/10.1182/blood.V87.6.2337.bloodjournal8762337
Severe infections and antithrombotic therapy 3055

31. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of sepsis in the United 57. Vilahur G, Ben-Aicha S, Badimon L. New insights into the role of adipose tissue in
States from 1979 through 2000. N Engl J Med 2003;348:1546–1554. https://doi.org/10. thrombosis. Cardiovasc Res 2017;113:1046–1054. https://doi.org/10.1093/cvr/cvx086
1056/NEJMoa022139 58. Wang S, Liu X, Chen Q, Liu C, Huang C, Fang X. The role of increased body mass index
32. Pandolfi F, Guillemot D, Watier L, Brun-Buisson C. Trends in bacterial sepsis incidence in outcomes of sepsis: a systematic review and meta-analysis. BMC Anesthesiol 2017;17:
and mortality in France between 2015 and 2019 based on National Health Data 118. https://doi.org/10.1186/s12871-017-0405-4
System (Systeme National des donnees de Sante (SNDS)): a retrospective observa­ 59. Friedman AN, Guirguis J, Kapoor R, Gupta S, Leaf DE, Timsina LR, et al. Obesity, in­
tional study. BMJ Open 2022;12:e058205. https://doi.org/10.1136/bmjopen-2021- flammatory and thrombotic markers, and major clinical outcomes in critically ill pa­
058205 tients with COVID-19 in the US. Obesity (Silver Spring) 2021;29:1719–1730. https://
33. Baker RE, Mahmud AS, Miller IF, Rajeev M, Rasambainarivo F, Rice BL, et al. Infectious doi.org/10.1002/oby.23245
disease in an era of global change. Nat Rev Microbiol 2022;20:193–205. https://doi.org/ 60. Aghili SMM, Ebrahimpur M, Arjmand B, Shadman Z, Pejman Sani M, Qorbani M, et al.
10.1038/s41579-021-00639-z Obesity in COVID-19 era, implications for mechanisms, comorbidities, and prognosis:
34. Mayr FB, Yende S, Angus DC. Epidemiology of severe sepsis. Virulence 2014;5:4–11. a review and meta-analysis. Int J Obes (Lond) 2021;45:998–1016. https://doi.org/10.
https://doi.org/10.4161/viru.27372 1038/s41366-021-00776-8
35. Vincent JL, Rello J, Marshall J, Silva E, Anzueto A, Martin CD, et al. International study of 61. Hendren NS, de Lemos JA, Ayers C, Das SR, Rao A, Carter S, et al. Association of body
the prevalence and outcomes of infection in intensive care units. JAMA 2009;302: mass index and age with morbidity and mortality in patients hospitalized with
2323–2329. https://doi.org/10.1001/jama.2009.1754 COVID-19: results from the American Heart Association COVID-19

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


36. Pryzdial ELG, Sutherland MR, Lin BH, Horwitz M. Antiviral anticoagulation. Res Pract Cardiovascular Disease Registry. Circulation 2021;143:135–144. https://doi.org/10.
Thromb Haemost 2020;4:774–788. https://doi.org/10.1002/rth2.12406 1161/CIRCULATIONAHA.120.051936
37. Li C, Li J, Ni H. Crosstalk between platelets and microbial pathogens. Front Immunol 62. El-Solh A, Sikka P, Bozkanat E, Jaafar W, Davies J. Morbid obesity in the medical ICU.
2020;11:1962. https://doi.org/10.3389/fimmu.2020.01962 Chest 2001;120:1989–1997. https://doi.org/10.1378/chest.120.6.1989
38. Zhang S, Liu Y, Wang X, Yang L, Li H, Wang Y, et al. SARS-CoV-2 binds platelet ACE2 63. Jagan N, Morrow LE, Walters RW, Plambeck RW, Wallen TJ, Patel TM, et al. Sepsis and
to enhance thrombosis in COVID-19. J Hematol Oncol 2020;13:120. https://doi.org/10. the obesity paradox: size matters in more than one way. Crit Care Med 2020;48:
1186/s13045-020-00954-7 e776–e782. https://doi.org/10.1097/ccm.0000000000004459
39. Ramachandran G. Gram-positive and gram-negative bacterial toxins in sepsis: a brief 64. Pepper DJ, Sun J, Welsh J, Cui X, Suffredini AF, Eichacker PQ. Increased body mass
review. Virulence 2014;5:213–218. https://doi.org/10.4161/viru.27024 index and adjusted mortality in ICU patients with sepsis or septic shock: a systematic
40. Iba T, Levy JH, Raj A, Warkentin TE. Advance in the management of sepsis-induced review and meta-analysis. Crit Care 2016;20:181. https://doi.org/10.1186/s13054-016-
coagulopathy and disseminated intravascular coagulation. J Clin Med 2019;8:728. 1360-z
https://doi.org/10.3390/jcm8050728 65. Sarkiss MG, Yusuf SW, Warneke CL, Botz G, Lakkis N, Hirch-Ginsburg C, et al. Impact
41. Lin G-L, McGinley JP, Drysdale SB, Pollard AJ. Epidemiology and immune pathogenesis
of aspirin therapy in cancer patients with thrombocytopenia and acute coronary syn­
of viral sepsis. Front Immunol 2018;9:2147. https://doi.org/10.3389/fimmu.2018.02147
dromes. Cancer 2007;109:621–627. https://doi.org/10.1002/cncr.22434
42. Anesi GL. COVID-19: epidemiology, clinical features, and prognosis of the critically ill
66. Falanga A, Leader A, Ambaglio C, Bagoly Z, Castaman G, Elalamy I, et al. EHA guidelines
adult. In. Up To Date. accessed 06222022; 2022.
on management of antithrombotic treatments in thrombocytopenic patients with can­
43. Timsit JF, Ruppé E, Barbier F, Tabah A, Bassetti M. Bloodstream infections in critically ill
cer. Hemasphere 2022;6:e750. https://doi.org/10.1097/HS9.0000000000000750
patients: an expert statement. Intensive Care Med 2020;46:266–284. https://doi.org/10.
67. Patrono C, Rocca B. Nonsteroidal antiinflammatory drugs: past, present and future.
1007/s00134-020-05950-6
Pharmacol Res 2009;59:285–289. https://doi.org/10.1016/j.phrs.2009.01.011
44. Southeast Asia Infectious Disease Clinical Research Network. Causes and outcomes of
68. Chiang N, Bermudez EA, Ridker PM, Hurwitz S, Serhan CN. Aspirin triggers antiinflam­
sepsis in Southeast Asia: a multinational multicentre cross-sectional study. Lancet Glob
matory 15-epi-lipoxin A4 and inhibits thromboxane in a randomized human trial. Proc
Health 2017;5:e157–e167. https://doi.org/10.1016/S2214-109X(17)30007-4
Natl Acad Sci U S A 2004;101:15178–15183. https://doi.org/10.1073/pnas.0405445101
45. FitzGerald GA, Patrono C. The coxibs, selective inhibitors of cyclooxygenase-2. N Engl
69. RECOVERY Collaborative Group. Aspirin in patients admitted to hospital with
J Med 2001;345:433–442. https://doi.org/10.1056/NEJM200108093450607
COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial.
46. World Health Organization. Integrating Neglected Tropical Diseases Into Global Health
Lancet 2022;399:143–151. https://doi.org/10.1016/s0140-6736(21)01825-0
and Development: Fourth WHO Report on Neglected Tropical Diseases: World Health
70. Kahn SR, Lim W, Dunn AS, Cushman M, Dentali F, Akl EA, et al. Prevention of VTE in
Organization. https://apps.who.int/iris/handle/10665/255011. License: CC BY-NC-SA
nonsurgical patients: antithrombotic therapy and prevention of thrombosis, 9th ed:
3.0 IGO. 2017.
American College of Chest Physicians Evidence-Based Clinical Practice Guidelines.
47. Goeijenbier M, van Wissen M, van de Weg C, Jong E, Gerdes VEA, Meijers JCM, et al.
Chest 2012;141:e195S–e226S. https://doi.org/10.1378/chest.11-2296
Review: viral infections and mechanisms of thrombosis and bleeding. J Med Virol 2012;
71. Trieu TK, Birkeland K, Kimchi A, Kedan I. Comprehensive collection of COVID-19 re­
84:1680–1696. https://doi.org/10.1002/jmv.23354
48. Raadsen M, Du Toit J, Langerak T, van Bussel B, van Gorp E, Goeijenbier M, et al. lated prosthetic valve failure: a systematic review. J Thromb Thrombolysis 2022:55:
Thrombocytopenia in virus infections. J Clin Med 2021;10:877. https://doi.org/10. 474–489. https://doi.org/10.1007/s11239-022-02746-x
3390/jcm10040877 72. Su W, Miao H, Guo Z, Chen Q, Huang T, Ding R. Associations between the use of
49. Goeijenbier M, van Gorp EC, Van den Brand JM, Stittelaar K, Bakhtiari K, Roelofs JJTH, aspirin or other antiplatelet drugs and all-cause mortality among patients with
et al. Activation of coagulation and tissue fibrin deposition in experimental influenza in COVID-19: a meta-analysis. Front Pharmacol 2022;13:989903. https://doi.org/10.
ferrets. BMC Microbiol 2014;14:134. https://doi.org/10.1186/1471-2180-14-134 3389/fphar.2022.989903
50. Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A, Delgado V, et al. 2019 ESC 73. Rentsch CT, Beckman JA, Tomlinson L, Gellad WF, Alcorn C, Kidwai-Khan F, et al.
guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collab­ Early initiation of prophylactic anticoagulation for prevention of coronavirus disease
oration with the EASD. Eur Heart J 2020;41:255–323. https://doi.org/10.1093/ 2019 mortality in patients admitted to hospital in the United States: cohort study.
eurheartj/ehz486 BMJ 2021;372:n311. https://doi.org/10.1136/bmj.n311
51. Knuuti J, Wijns W, Saraste A, Capodanno D, Barbato E, Funck-Brentano C, et al. 2019 74. Poulakou G, Dimakakos E, Kollias A, Kyriakoulis KG, Rapti V, Trontzas I, et al. Beneficial
ESC guidelines for the diagnosis and management of chronic coronary syndromes. Eur effects of intermediate dosage of anticoagulation treatment on the prognosis of hos­
Heart J 2020;41:407–477. https://doi.org/10.1093/eurheartj/ehz425 pitalized COVID-19 patients: the ETHRA study. In Vivo 2021;35:653–661. https://doi.
52. Akintoye E, Afonso L, Bengaluru Jayanna M, Bao W, Briasoulis A, Robinson J. org/10.21873/invivo.12305
Prognostic utility of risk enhancers and coronary artery calcium score recommended 75. Nadkarni GN, Lala A, Bagiella E, Chang HL, Moreno PR, Pujadas E, et al.
in the 2018 ACC/AHA multisociety cholesterol treatment guidelines over the pooled Anticoagulation, bleeding, mortality, and pathology in hospitalized patients with
cohort equation: insights from 3 large prospective cohorts. J Am Heart Assoc 2021;10: COVID-19. J Am Coll Cardiol 2020;76:1815–1826. https://doi.org/10.1016/j.jacc.2020.
e019589. https://doi.org/10.1161/JAHA.120.019589 08.041
53. Golub I S, Termeie Orly G, Kristo S, Schroeder LP, Lakshmanan S, Shafter AM, et al. 76. Tang N, Bai H, Chen X, Gong J, Li D, Sun Z. Anticoagulant treatment is associated with
Major global coronary artery calcium guidelines. JACC Cardiovasc Imaging 2023;16: decreased mortality in severe coronavirus disease 2019 patients with coagulopathy. J
98–117. https://doi.org/10.1016/j.jcmg.2022.06.018 Thromb Haemost 2020;18:1094–1099. https://doi.org/10.1111/jth.14817
54. Oikonomou E, Leopoulou M, Theofilis P, Antonopoulos AS, Siasos G, Latsios G, et al. 77. Zhang Y, Wang M, Zhang X, Liu T, Libby P, Shi G-P. COVID-19, the pandemic of the
A link between inflammation and thrombosis in atherosclerotic cardiovascular dis­ century and its impact on cardiovascular diseases. Cardiol Discov 2021;1:233–258.
eases: clinical and therapeutic implications. Atherosclerosis 2020;309:16–26. https:// https://doi.org/10.1097/CD9.0000000000000038
doi.org/10.1016/j.atherosclerosis.2020.07.027 78. Wang Y, Ouyang Y, Liu B, Ma X, Ding R. Platelet activation and antiplatelet therapy in
55. Schuetz P, Castro P, Shapiro NI. Diabetes and sepsis: preclinical findings and clinical sepsis: a narrative review. Thromb Res 2018;166:28–36. https://doi.org/10.1016/j.
relevance. Diabetes Care 2011;34:771–778. https://doi.org/10.2337/dc10-1185 thromres.2018.04.007
56. Shah BR, Hux JE. Quantifying the risk of infectious diseases for people with diabetes. 79. Merx MW, Weber C. Sepsis and the heart. Circulation 2007;116:793–802. https://doi.
Diabetes Care 2003;26:510–513. https://doi.org/10.2337/diacare.26.2.510 org/10.1161/circulationaha.106.678359
3056 Gigante et al.

80. Ou SM, Chu H, Chao PW, Lee Y-J, Kuo S-C, Chen T-J, et al. Long-term mortality and 100. Han Y, Liao Z, Li Y, Zhao X, Ma S, Bao D, et al. Magnetically controlled capsule endos­
major adverse cardiovascular events in sepsis survivors. A nationwide population- copy for assessment of antiplatelet therapy-induced gastrointestinal injury. J Am Coll
based study. Am J Respir Crit Care Med 2016;194:209–217. https://doi.org/10.1164/ Cardiol 2022;79:116–128. https://doi.org/10.1016/j.jacc.2021.10.028
rccm.201510-2023OC 101. Moayyedi P, Eikelboom JW, Bosch J, Connolly SJ, Dyal L, Shestakovska O, et al. Safety
81. Erlich JM, Talmor DS, Cartin-Ceba R, Gajic O, Kor DJ. Prehospitalization antiplatelet of proton pump inhibitors based on a large, multi-year, randomized trial of patients
therapy is associated with a reduced incidence of acute lung injury: a population-based receiving rivaroxaban or aspirin. Gastroenterology 2019;157:682–691.e2. https://doi.
cohort study. Chest 2011;139:289–295. https://doi.org/10.1378/chest.10-0891 org/10.1053/j.gastro.2019.05.056
82. Losche W, Boettel J, Kabisch B, Winning J, Claus RA, Bauer M, et al. Do aspirin and 102. MacLaren R, Kassel LE, Kiser TH, Fish DN. Proton pump inhibitors and histamine-2
other antiplatelet drugs reduce the mortality in critically ill patients? Thrombosis receptor antagonists in the intensive care setting: focus on therapeutic and adverse
2012;2012:720254. https://doi.org/10.1155/2012/720254 events. Expert Opin Drug Saf 2015;14:269–280. https://doi.org/10.1517/14740338.
83. Valerio-Rojas JC, Jaffer IJ, Kor DJ, Gajic O, Cartin-Ceba R. Outcomes of severe sepsis 2015.986456
and septic shock patients on chronic antiplatelet treatment: a historical cohort study. 103. Ye Z, Reintam Blaser A, Lytvyn L, Wang Y, Guyatt GH, Mikita JS, et al. Gastrointestinal
Crit Care Res Pract 2013;2013:782573. https://doi.org/10.1155/2013/782573 bleeding prophylaxis for critically ill patients: a clinical practice guideline. BMJ 2020;368:
84. Tsai MJ, Ou SM, Shih CJ, Chao P-W, Wang L-F, Shih Y-N, et al. Association of prior l6722. https://doi.org/10.1136/bmj.l6722
antiplatelet agents with mortality in sepsis patients: a nationwide population-based co­ 104. Frelinger AL III, Lee RD, Mulford DJ, Wu J, Nudurupati S, Nigam A, et al. A randomized,
hort study. Intensive Care Med 2015;41:806–813. https://doi.org/10.1007/s00134-015- 2-period, crossover design study to assess the effects of dexlansoprazole, lansopra­

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


3760-y zole, esomeprazole, and omeprazole on the steady-state pharmacokinetics and
85. Trauer J, Muhi S, McBryde ES, Al Harbi SA, Arabi YM, Boyle AJ, et al. Quantifying the pharmacodynamics of clopidogrel in healthy volunteers. J Am Coll Cardiol 2012;59:
effects of prior acetyl-salicylic acid on sepsis-related deaths: an individual patient data 1304–1311. https://doi.org/10.1016/j.jacc.2011.12.024
meta-analysis using propensity matching. Crit Care Med 2017;45:1871–1879. https:// 105. Ferreiro JL, Ueno M, Tomasello SD, Capodanno D, Desai B, Dharmashankar K, et al.
doi.org/10.1097/CCM.0000000000002654 Pharmacodynamic evaluation of pantoprazole therapy on clopidogrel effects: results of
86. Ouyang Y, Wang Y, Liu B, Ma X, Ding R. Effects of antiplatelet therapy on the mortality a prospective, randomized, crossover study. Circ Cardiovasc Interv 2011;4:273–279.
rate of patients with sepsis: a meta-analysis. J Crit Care 2019;50:162–168. https://doi. https://doi.org/10.1161/circinterventions.110.960997
org/10.1016/j.jcrc.2018.12.004 106. Ferreiro JL, Ueno M, Capodanno D, Desai B, Dharmashankar K, Darlington A, et al.
87. Chow JH, Yin Y, Yamane DP, Davison D, Keneally RJ, Hawkins K, et al. Association of Pharmacodynamic effects of concomitant versus staggered clopidogrel and omepra­
prehospital antiplatelet therapy with survival in patients hospitalized with COVID-19: a zole intake: results of a prospective randomized crossover study. Circ Cardiovasc
propensity score-matched analysis. J Thromb Haemost 2021;19:2814–2824. https://doi.
Interv 2010;3:436–441. https://doi.org/10.1161/circinterventions.110.957829
org/10.1111/jth.15517 107. https://www.ema.europa.eu/en/documents/product-information/plavix-epar-
88. Wiewel MA, de Stoppelaar SF, van Vught LA, Frencken JF, Hoogendijk AJ, Klein
product-information_en.pdf
Klouwenberg PMC, et al. Chronic antiplatelet therapy is not associated with altera­
108. https://www.ema.europa.eu/en/documents/product-information/brilique-epar-
tions in the presentation, outcome, or host response biomarkers during sepsis: a
product-information_en.pdf
propensity-matched analysis. Intensive Care Med 2016;42:352–360. https://doi.org/
109. Du F, Jiang P, He S, Song D, Xu F. Antiplatelet therapy for critically ill patients: a pair­
10.1007/s00134-015-4171-9
wise and Bayesian network meta-analysis. Shock 2018;49:616–624. https://doi.org/10.
89. Adelborg K, Corraini P, Darvalics B, Frederiksen H, Ording A, Horváth-Puhó E, et al.
1097/SHK.0000000000001057
Risk of thromboembolic and bleeding outcomes following hematological cancers: a
110. Santoro F, Nunez-Gil IJ, Vitale E, Viana-Llamas MC, Reche-Martinez B, Romero-Pareja
Danish population-based cohort study. J Thromb Haemost 2019;17:1305–1318.
R, et al. Antiplatelet therapy and outcome in COVID-19: the health outcome predict­
https://doi.org/10.1111/jth.14475
ive evaluation registry. Heart 2022;108:130–136. https://doi.org/10.1136/heartjnl-
90. Antithrombotic Trialists C, Baigent C, Blackwell L, Collins R, Emberson J, Godwin J,
2021-319552
et al. Aspirin in the primary and secondary prevention of vascular disease: collaborative
111. Blasco-Colmenares E, Perl TM, Guallar E, Baumgartner WA, Conte JV, Alejo D, et al.
meta-analysis of individual participant data from randomised trials. Lancet 2009;373:
Aspirin plus clopidogrel and risk of infection after coronary artery bypass surgery. Arch
1849–1860. https://doi.org/10.1016/S0140-6736(09)60503-1
Intern Med 2009;169:788–796. https://doi.org/10.1001/archinternmed.2009.42
91. Hakim DA, Dangas GD, Caixeta A, Nikolsky E, Lansky AJ, Moses JW, et al. Impact of
112. Lupu L, Shepshelovich D, Banai S, Hershkoviz R, Isakov O. Effect of ticagrelor on re­
baseline thrombocytopenia on the early and late outcomes after ST-elevation myocar­
ducing the risk of gram-positive infections in patients with acute coronary syndrome.
dial infarction treated with primary angioplasty: analysis from the Harmonizing
Am J Cardiol 2020;130:56–63. https://doi.org/10.1016/j.amjcard.2020.06.016
Outcomes with Revascularization and Stents in Acute Myocardial Infarction
113. Storey RF, James SK, Siegbahn A, Varenhorst C, Held C, Ycas J, et al. Lower mortality
(HORIZONS-AMI) trial. Am Heart J 2011;161:391–396. https://doi.org/10.1016/j.ahj.
following pulmonary adverse events and sepsis with ticagrelor compared to clopido­
2010.11.001
92. Feher A, Kampaktsis PN, Parameswaran R, Stein EM, Steingart R, Gupta D, et al. grel in the PLATO study. Platelets 2014;25:517–525. https://doi.org/10.3109/
Aspirin is associated with improved survival in severely thrombocytopenic cancer pa­ 09537104.2013.842965
tients with acute myocardial infarction. Oncologist 2017;22:213–221. https://doi.org/ 114. Varenhorst C, Alström U, Braun O, Storey RF, Mahaffey KW, Bertilsson M, et al.
10.1634/theoncologist.2016-0110 Causes of mortality with ticagrelor compared with clopidogrel in acute coronary syn­
93. Moayyedi P, Eikelboom JW, Bosch J, Connolly SJ, Dyal L, Shestakovska O, et al. dromes. Heart 2014;100:1762–1769. https://doi.org/10.1136/heartjnl-2014-305619
Pantoprazole to prevent gastroduodenal events in patients receiving rivaroxaban 115. Varenhorst C, Alstrom U, Scirica BM, Hogue CW, Åsenblad N, Storey RF, et al. Factors
and/or aspirin in a randomized, double-blind, placebo-controlled trial. contributing to the lower mortality with ticagrelor compared with clopidogrel in pa­
Gastroenterology 2019;157:403–412 e405. https://doi.org/10.1053/j.gastro.2019.04. tients undergoing coronary artery bypass surgery. J Am Coll Cardiol 2012;60:
041 1623–1630. https://doi.org/10.1016/j.jacc.2012.07.021
94. Scally B, Emberson JR, Spata E, Reith C, Davies K, Halls H, et al. Effects of gastropro­ 116. Nelson TA, Parker WAE, Ghukasyan Lakic T, Westerbergh J, James SK, Siegbahn A,
tectant drugs for the prevention and treatment of peptic ulcer disease and its compli­ et al. Differential effect of clopidogrel and ticagrelor on leukocyte count in relation
cations: a meta-analysis of randomised trials. Lancet Gastroenterol Hepatol 2018;3: to patient characteristics, biomarkers and genotype: a PLATO substudy. Platelets
231–241. https://doi.org/10.1016/S2468-1253(18)30037-2 2022;33:425–431. https://doi.org/10.1080/09537104.2021.1934667
95. Bakker T, Dongelmans DA, Nabovati E, Eslami S, Keizer NF, Abu-Hanna A, et al. 117. CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin
Heterogeneity in the identification of potential drug-drug interactions in the intensive in patients at risk of ischaemic events (CAPRIE). Lancet 1996;348:1329–1339. https://
care unit: a systematic review, critical appraisal, and reporting recommendations. J Clin doi.org/10.1016/s0140-6736(96)09457-3
Pharmacol 2022;62:706–720. https://doi.org/10.1002/jcph.2020 118. Kiers D, van der Heijden WA, van Ede L, Gerretsen J, de Mast Q, van der Ven AJ, et al.
96. Patrono C, Rocca B. Aspirin and other COX-1 inhibitors. Handb Exp Pharmacol A randomised trial on the effect of anti-platelet therapy on the systemic inflammatory
2012:210:137–164. https://doi.org/10.1007/978-3-642-29423-5_6 response in human endotoxaemia. Thromb Haemost 2017;117:1798–1807. https://doi.
97. Rocca B, Petrucci G. Personalized medicine, pharmacogenetics, and clopidogrel: org/10.1160/TH16-10-0799
unraveling variability of response. Mol Interv 2010;10:12–19. https://doi.org/10.1124/ 119. Rabouel Y, Magnenat S, Delabranche X, Gachet C, Hechler B. Platelet P2Y 12 receptor
mi.10.1.4 deletion or pharmacological inhibition does not protect mice from sepsis or septic
98. Varma MVS, Bi YA, Lazzaro S, West M. Clopidogrel as a perpetrator of drug-drug in­ shock. TH Open 2021;5:e343–e352. https://doi.org/10.1055/s-0041-1733857
teractions: a challenge for quantitative predictions? Clin Pharmacol Ther 2019;105: 120. Thomas MR, Outteridge SN, Ajjan RA, Phoenix F, Sangha GK, Faulkner RE, et al.
1295–1299. https://doi.org/10.1002/cpt.1398 Platelet P2Y12 inhibitors reduce systemic inflammation and its prothrombotic effects
99. Hallas J, Dall M, Andries A, Andersen BS, Aalykke C, Hansen JM, et al. Use of single and in an experimental human model. Arterioscler Thromb Vasc Biol 2015;35:2562–2570.
combined antithrombotic therapy and risk of serious upper gastrointestinal bleeding: https://doi.org/10.1161/ATVBAHA.115.306528
population based case-control study. BMJ 2006;333:726. https://doi.org/10.1136/bmj. 121. Totani L, Dell’Elba G, Martelli N, Di Santo A, Piccoli A, Amore C, et al. Prasugrel in­
38947.697558.AE hibits platelet-leukocyte interaction and reduces inflammatory markers in a model
Severe infections and antithrombotic therapy 3057

of endotoxic shock in the mouse. Thromb Haemost 2012;107:1130–1140. https://doi. 145. https://www.ema.europa.eu/en/documents/product-information/eliquis-epar-product-
org/10.1160/TH11-12-0867 information_en.pdf.
122. Berger JS, Kornblith LZ, Gong MN, Reynolds HR, Cushman M, Cheng Y, et al. Effect of 146. Ray WA, Chung CP, Murray KT, Smalley WE, Daugherty JR, Dupont WD, et al.
P2Y12 inhibitors on survival free of organ support among non-critically ill hospitalized Association of proton pump inhibitors with reduced risk of warfarin-related serious
patients with COVID-19: a randomized clinical trial. JAMA 2022;327:227–236. https:// upper gastrointestinal bleeding. Gastroenterology 2016;151:1105–1112.e10. https://
doi.org/10.1001/jama.2021.23605 doi.org/10.1053/j.gastro.2016.08.054
123. Piccolo R, Feres F, Abizaid A, Gilard M, Morice M-C, Hong M-K, et al. Risk of early ad­ 147. Komen J, Pottegård A, Hjemdahl P, Mantel-Teeuwisse AK, Wettermark B, Hellfritzsch
verse events after clopidogrel discontinuation in patients undergoing short-term dual M, et al. Non-vitamin K antagonist oral anticoagulants, proton pump inhibitors and
antiplatelet therapy: an individual participant data analysis. JACC Cardiovasc Interv 2017; gastrointestinal bleeds. Heart 2022;108:613–618. https://doi.org/10.1136/heartjnl-
10:1621–1630. https://doi.org/10.1016/j.jcin.2017.06.001 2021-319332
124. Khan SU, Singh M, Valavoor S, Khan MU, Lone AN, Khan MZ, et al. Dual antiplatelet 148. Mar PL, Gopinathannair R, Gengler BE, Chung MK, Perez A, Dukes J, et al. Drug inter­
therapy after percutaneous coronary intervention and drug-eluting stents: a systemat­ actions affecting oral anticoagulant use. Circ Arrhythm Electrophysiol 2022;15:e007956.
ic review and network meta-analysis. Circulation 2020;142:1425–1436. https://doi.org/ https://doi.org/10.1161/circep.121.007956
10.1161/circulationaha.120.046308 149. Vamos EP, Pape UJ, Curcin V, Harris MJ, Valabhji J, Majeed A, et al. Effectiveness of the
125. Smeeth L, Thomas SL, Hall AJ, Hubbard R, Farrington P, Vallance P, et al. Risk of myo­ influenza vaccine in preventing admission to hospital and death in people with type 2
cardial infarction and stroke after acute infection or vaccination. N Engl J Med 2004; diabetes. CMAJ 2016;188:E342–E351. https://doi.org/10.1503/cmaj.151059

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023


351:2611–2618. https://doi.org/10.1056/NEJMoa041747 150. Vardeny O, Kim K, Udell JA, Joseph J, Desai AS, Farkouh ME, et al. Effect of high-dose
126. Capodanno D, Baber U, Bhatt DL, Collet J-P, Dangas G, Franchi F, et al. P2y(12) inhibi­ trivalent vs standard-dose quadrivalent influenza vaccine on mortality or cardiopul­
tor monotherapy in patients undergoing percutaneous coronary intervention. Nat Rev monary hospitalization in patients with high-risk cardiovascular disease: a randomized
Cardiol 2022;19:829–844. https://doi.org/10.1038/s41569-022-00725-6 clinical trial. JAMA 2021;325:39–49. https://doi.org/10.1001/jama.2020.23649
127. Lopes RD, Heizer G, Aronson R, Vora AN, Massaro T, Mehran R, et al. Antithrombotic 151. Modin D, Claggett B, Kober L, Schou M, Jensen JUS, Solomon SD, et al. Influenza vac­
therapy after acute coronary syndrome or PCI in atrial fibrillation. N Engl J Med 2019; cination is associated with reduced cardiovascular mortality in adults with diabetes: a
380:1509–1524. https://doi.org/10.1056/NEJMoa1817083 nationwide cohort study. Diabetes Care 2020;43:2226–2233. https://doi.org/10.2337/
128. Schilling U, Dingemanse J, Ufer M. Pharmacokinetics and pharmacodynamics of ap­ dc20-0229
proved and investigational P2Y12 receptor antagonists. Clin Pharmacokinet 2020;59: 152. Modin D, Claggett B, Jorgensen ME, Køber L, Benfield T, Schou M, et al. Flu vaccine and
545–566. https://doi.org/10.1007/s40262-020-00864-4 mortality in hypertension: a nationwide cohort study. J Am Heart Assoc 2022;11:
129. Storey RF, Bliden KP, Patil SB, Karunakaran A, Ecob R, Butler K, et al. Incidence of dys­ e021715. https://doi.org/10.1161/JAHA.121.021715
pnea and assessment of cardiac and pulmonary function in patients with stable coron­ 153. Hollingsworth R, Palmu A, Pepin S, Dupuy M, Shrestha A, Jokinen J, et al. Effectiveness
ary artery disease receiving ticagrelor, clopidogrel, or placebo in the ONSET/OFFSET of the quadrivalent high-dose influenza vaccine for prevention of cardiovascular and
study. J Am Coll Cardiol 2010;56:185–193. https://doi.org/10.1016/j.jacc.2010.01.062 respiratory events in people aged 65 years and above: rationale and design of a real-
130. Arora S, Shemisa K, Vaduganathan M, Qamar A, Gupta A, Garg SK, et al. Premature world pragmatic randomized clinical trial. Am Heart J 2021;237:54–61. https://doi.
ticagrelor discontinuation in secondary prevention of atherosclerotic CVD: JACC re­ org/10.1016/j.ahj.2021.03.007
view topic of the week. J Am Coll Cardiol 2019;73:2454–2464. https://doi.org/10.1016/j. 154. Gurfinkel EP, de la Fuente RL, Mendiz O, Mautner B. Influenza vaccine pilot study in
jacc.2019.03.470 acute coronary syndromes and planned percutaneous coronary interventions: the
131. Sumaya W, Parker WAE, Fretwell R, Hall I, Barmby D, Richardson J, et al. FLU Vaccination Acute Coronary Syndromes (FLUVACS) study. Circulation 2002;
Pharmacodynamic effects of a 6-hour regimen of enoxaparin in patients undergoing 105:2143–2147. https://doi.org/10.1161/01.cir.0000016182.85461.f4
primary percutaneous coronary intervention (PENNY PCI Study). Thromb Haemost 155. Frobert O, Gotberg M, Erlinge D, Akhtar Z, Christiansen EH, MacIntyre CR, et al.
2018;118:1250–1256. https://doi.org/10.1055/s-0038-1657768 Influenza vaccination after myocardial infarction: a randomized, double-blind, placebo-
132. Shimizu M, Natori T, Tsuda K, Yoshida M, Kamada A, Oi K, et al. Thrombin-induced controlled, multicenter trial. Circulation 2021;144:1476–1484. https://doi.org/10.1161/
platelet aggregation—effect of dabigatran using automated platelet aggregometry−. CIRCULATIONAHA.121.057042
Platelets 2020;31:360–364. https://doi.org/10.1080/09537104.2019.1624707 156. Fonseca HAR, Furtado RHM, Zimerman A, Lemos PA, Franken M, Monfardini F, et al.
133. Lamontagne F, Garant MP, Carvalho JC, Lanthier L, Smieja M, Pilon D, et al. Influenza vaccination strategy in acute coronary syndromes: the VIP-ACS trial. Eur
Pneumococcal vaccination and risk of myocardial infarction. Cmaj 2008;179: Heart J 2022;43:4378–4388. https://doi.org/10.1093/eurheartj/ehac472
773–777. https://doi.org/10.1503/cmaj.070221 157. Phrommintikul A, Kuanprasert S, Wongcharoen W, Kanjanavanit R, Chaiwarith R,
134. Eisen DP, Reid D, McBryde ES. Acetyl salicylic acid usage and mortality in critically ill Sukonthasarn A, et al. Influenza vaccination reduces cardiovascular events in patients
patients with the systemic inflammatory response syndrome and sepsis. Crit Care with acute coronary syndrome. Eur Heart J 2011;32:1730–1735. https://doi.org/10.
Med 2012;40:1761–1767. https://doi.org/10.1097/CCM.0b013e318246b9df 1093/eurheartj/ehr004
135. Meizlish ML, Goshua G, Liu Y, Fine R, Amin K, Chang E, et al. Intermediate-dose antic­ 158. Gurfinkel EP, Leon de la Fuente R, Mendiz O, Mautner B. Flu vaccination in acute cor­
oagulation, aspirin, and in-hospital mortality in COVID-19: a propensity score- onary syndromes and planned percutaneous coronary interventions (FLUVACS)
matched analysis. Am J Hematol 2021;96:471–479. https://doi.org/10.1002/ajh.26102 study. Eur Heart J 2004;25:25–31. https://doi.org/10.1016/j.ehj.2003.10.018
136. Otto GP, Sossdorf M, Boettel J, Kabisch B, Breuel H, Winning J, et al. Effects of low- 159. Ciszewski A, Bilinska ZT, Brydak LB, Brydak LB, Kepka C, Kruk M, et al. Influenza vac­
dose acetylsalicylic acid and atherosclerotic vascular diseases on the outcome in pa­ cination in secondary prevention from coronary ischaemic events in coronary artery
tients with severe sepsis or septic shock. Platelets 2013;24:480–485. https://doi.org/ disease: FLUCAD study. Eur Heart J 2008;29:1350–1358. https://doi.org/10.1093/
10.3109/09537104.2012.724482 eurheartj/ehm581
137. Umemura Y, Yamakawa K, Ogura H, Yuhara H, Fujimi S. Efficacy and safety of anti­ 160. Jaiswal V, Ang SP, Yaqoob S, Yaqoob S, Ishak A, Chia JE, et al. Cardioprotective effects
coagulant therapy in three specific populations with sepsis: a meta-analysis of rando­ of influenza vaccination among patients with established cardiovascular disease or at
mized controlled trials. J Thromb Haemost 2016;14:518–530. https://doi.org/10.1111/ high cardiovascular risk: a systematic review and meta-analysis. Eur J Prev Cardiol
jth.13230 2022;29:1881–1892. https://doi.org/10.1093/eurjpc/zwac152
138. Wang C, Chi C, Guo L, Wang X, Guo L, Sun J, et al. Heparin therapy reduces 28-day 161. Hung IF, Leung AY, Chu DW, Leung D, Cheung T, Chan C-K, et al. Prevention of acute
mortality in adult severe sepsis patients: a systematic review and meta-analysis. Crit myocardial infarction and stroke among elderly persons by dual pneumococcal and in­
Care 2014;18:563. https://doi.org/10.1186/s13054-014-0563-4 fluenza vaccination: a prospective cohort study. Clin Infect Dis 2010;51:1007–1016.
139. Li X, Liu Z, Luo M, Xi Y, Li C, Wang S, et al. Therapeutic effect of low-molecular-weight https://doi.org/10.1086/656587
heparin on adult sepsis: a meta-analysis. Ann Palliat Med 2021;10:3115–3127. https:// 162. Ochoa-Gondar O, Vila-Corcoles A, Rodriguez-Blanco T, de Diego-Cabanes C,
doi.org/10.21037/apm-21-169 Hospital-Guardiola I, Jariod-Pamies M, et al. Evaluating the clinical effectiveness of
140. Søgaard M, Skjøth F, Kjældgaard JN, Lip GYH, Larsen TB. Bleeding complications in an­ pneumococcal vaccination in preventing myocardial infarction: the CAPAMIS study,
ticoagulated patients with atrial fibrillation and sepsis: a propensity-weighted cohort three-year follow-up. Vaccine 2014;32:252–257. https://doi.org/10.1016/j.vaccine.
study. J Am Heart Assoc 2017;6:e007453. https://doi.org/10.1161/jaha.117.007453 2013.11.017
141. Darwish OS, Strube S, Nguyen HM, Tanios MA. Challenges of anticoagulation for atrial 163. Vila-Corcoles A, Ochoa-Gondar O, Rodriguez-Blanco T, de Diego-Cabanes C,
fibrillation in patients with severe sepsis. Ann Pharmacother 2013;47:1266–1271. Satue-Gracia E, Vila-Rovira A, et al. Evaluating clinical effectiveness of pneumococcal
https://doi.org/10.1177/1060028013500938 vaccination in preventing stroke: the CAPAMIS study, 3-year follow-up. J Stroke
142. https://www.ema.europa.eu/en/documents/product-information/pradaxa-epar-product- Cerebrovasc Dis 2014;23:1577–1584. https://doi.org/10.1016/j.jstrokecerebrovasdis.
information_en.pdf. 2013.12.047
143. https://www.ema.europa.eu/en/documents/product-information/lixiana-epar-product- 164. Ren S, Newby D, Li SC, Walkom E, Miller P, Hure A, et al. Effect of the adult pneumo­
information_en.pdf. coccal polysaccharide vaccine on cardiovascular disease: a systematic review and
144. https://www.ema.europa.eu/en/documents/product-information/xarelto-epar-product- meta-analysis. Open Heart 2015;2:e000247. https://doi.org/10.1136/openhrt-2015-
information_en.pdf. 000247
3058 Gigante et al.

165. Marchandot B, Curtiaud A, Trimaille A, Sattler L, Grunebaum L, Morel O. FAPIC score. Eur Heart J 2021;42:4053–4063. https://doi.org/10.1093/eurheartj/
Vaccine-induced immune thrombotic thrombocytopenia: current evidence, potential ehab592
mechanisms, clinical implications, and future directions. Eur Heart J Open 2021;1: 169. See I, Su JR, Lale A, Woo EJ, Guh AY, Shimabukuro TT, et al. US case reports of cerebral
oeab014. https://doi.org/10.1093/ehjopen/oeab014 venous sinus thrombosis with thrombocytopenia after Ad26.COV2.S vaccination, March
166. Pavord S, Scully M, Hunt BJ, Lester W, Bagot C, Craven B, et al. Clinical features of 2 to April 21, 2021. JAMA 2021;325:2448–2456. https://doi.org/10.1001/jama.2021.7517
vaccine-induced immune thrombocytopenia and thrombosis. N Engl J Med 2021; 170. Mandal AKJ, Kho J, Ioannou A, Van den Abbeele K, Missouris CG. COVID-19 and in
385:1680–1689. https://doi.org/10.1056/NEJMoa2109908 situ pulmonary artery thrombosis. Respir Med 2021;176:106176. https://doi.org/10.
167. Greinacher A, Thiele T, Warkentin TE, Weisser K, Kyrle PA, Eichinger S, et al. 1016/j.rmed.2020.106176
Thrombotic thrombocytopenia after ChAdOx1 nCov-19 vaccination. N Engl J Med 171. Udell JA, Zawi R, Bhatt DL, Keshtkar-Jahromi M, Gaughran F, Phrommintikul A, et al.
2021;384:2092–2101. https://doi.org/10.1056/NEJMoa2104840 Association between influenza vaccination and cardiovascular outcomes in high-risk
168. Hwang J, Park SH, Lee SW, Lee SB, Lee MH, Jeong GH, et al. Predictors of mortality patients: a meta-analysis. JAMA 2013;310:1711–1720. https://doi.org/10.1001/jama.
in thrombotic thrombocytopenia after adenoviral COVID-19 vaccination: the 2013.279206

Downloaded from https://academic.oup.com/eurheartj/article/44/32/3040/7223688 by guest on 28 August 2023

You might also like