You are on page 1of 11

PERSpECTIVES

hypoxaemia (low oxygen levels in the


Endothelial dysfunction and blood) and dyspnoea (shortness of breath),
potentially evolving towards ARDS14. This

immunothrombosis as key stage of the disease is characterized by high


levels of pro-inflammatory cytokines, such

pathogenic mechanisms in COVID-19 as interleukin-6 (IL-6), IL-1β, IL-18 and


granulocyte–macrophage colony-stimulating
factor (GM–CSF)2,15–17. This event has been
Aldo Bonaventura   , Alessandra Vecchié, Lorenzo Dagna, Kimberly Martinod   , referred to as a cytokine storm18,19, although
recent evidence suggests that cytokine
Dave L. Dixon, Benjamin W. Van Tassell, Francesco Dentali, levels in patients with COVID-19 are
Fabrizio Montecucco   , Steffen Massberg   , Marcel Levi and Antonio Abbate lower than those reported in patients with
non-COVID-19 ARDS, sepsis, influenza
Abstract | Coronavirus disease 2019 (COVID-19) is a clinical syndrome caused by
virus infection and chimeric antigen
infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). receptor (CAR) T cell-induced cytokine
Patients with severe disease show hyperactivation of the immune system, which release syndrome20,21. SARS-CoV-2 has been
can affect multiple organs besides the lungs. Here, we propose that SARS-CoV-2 shown to trigger the IL-1/IL-6 pathway to
infection induces a process known as immunothrombosis, in which activated a larger extent than other coronaviruses22.
neutrophils and monocytes interact with platelets and the coagulation cascade, Through the analysis of lungs of patients
who died from COVID-19-related ARDS,
leading to intravascular clot formation in small and larger vessels. Microthrombotic SARS-CoV-2 was found to activate the
complications may contribute to acute respiratory distress syndrome (ARDS) and NLRP3 inflammasome23,24. In vitro studies also
other organ dysfunctions. Therapeutic strategies aimed at reducing showed NLRP3 inflammasome activation
immunothrombosis may therefore be useful. Several antithrombotic and by SARS-CoV-2 infection of primary
immunomodulating drugs have been proposed as candidates to treat patients with human monocytes. In addition, NLRP3
inflammasome products, such as IL-1β and
SARS-CoV-2 infection. The growing understanding of SARS-CoV-2 infection
IL-18, are increased in patients with severe
pathogenesis and how it contributes to critical illness and its complications may COVID-19 and positively correlated with
help to improve risk stratification and develop targeted therapies to reduce the adverse clinical outcomes24. These findings
acute and long-term consequences of this disease. support a potential relationship between
SARS-CoV-2 infection and inflammatory
Coronavirus disease 2019 (COVID-19) systems cooperate to block pathogens and features, such as the dysregulated cytokine
is caused by the severe acute respiratory limit their spread10. In this Perspective, we response.
syndrome coronavirus 2 (SARS-CoV-2) and describe how SARS-CoV-2 might induce Increased levels of ferritin have also
can present with a range of manifestations, a prothrombotic state and discuss how been observed among patients with
including pneumonia and acute respiratory currently available and developing therapies COVID-19, similar to what is observed
distress syndrome (ARDS)1. Although may address COVID-19-associated in macrophage activation syndrome16,19,25.
many individuals develop asymptomatic or immunothrombosis. Even though ferritin levels observed in
paucisymptomatic SARS-CoV-2 infection, macrophage activation syndrome are
those with severe-to-critical disease show Immunothrombosis in COVID-19 markedly higher than those currently
a dysregulated hyperactivation of the COVID-19-associated hyperinflammation. observed in COVID-19 (≥10,000–
immune system that can cause an abnormal The clinical spectrum of COVID-19 ranges 100,000 ng ml–1 versus 500–3,000 ng ml–1),
cytokine immune response and is likely to from asymptomatic or paucisymptomatic ferritin levels remain predictive of poor
be responsible for most of the organ damage (in up to 45% of individuals) to severe outcomes in COVID-19 (refs26,27).
occurring in the late phase of the disease2. disease requiring admission of patients to With regard to inflammatory cells, large
It is now apparent that SARS-CoV-2 the intensive care unit (ICU)11. Generally, numbers of CD68+ macrophages were
infection induces a prothrombotic 3–10% of subjects requires hospitalization, observed in the alveoli of patients who died
state manifesting especially with with up to 20% of them experiencing severe from COVID-19-associated pneumonia,
microthrombosis3,4, as observed in patients disease and a high mortality rate12,13. The and SARS-CoV-2 proteins were detected in
with COVID-19 and in post-mortem initial phase of COVID-19 is characterized CD68+ alveolar macrophages28,29. Analysis
samples from individuals who died after by damage directly driven by the virus. of the bronchoalveolar fluid of patients
SARS-CoV-2 infection5–9. This thrombotic After approximately 7–10 days of symptoms with severe or critical COVID-19 showed
state is reminiscent of a process known (including fever, cough, fatigue, anorexia a higher percentage of macrophages and
as immunothrombosis (Box 1; Fig. 1), and myalgias), a subgroup of patients neutrophils and a lower proportion of
in which the immune and coagulation progresses to severe disease and develops myeloid dendritic cells, plasmacytoid

NATuRE REVIEWS | Immunology

0123456789();:
Perspectives

dendritic cells and T cells than that of These features are more consistent with proposed the occurrence of two or more
patients with moderate disease30. They a state of hypercoagulability coupled with a of the following four criteria in cases of
also had higher levels of pro-inflammatory severe inflammatory state rather than with proven COVID-19: a decrease in platelet
cytokines (such as IL-6, IL-8 and IL-1β) and classical disseminated intravascular coagulation count (<150 × 109 l–1); an increase in D-dimer
chemokines (such as CCL2, CCL3, CCL4 (DIC)36. Indeed, DIC has been reported levels (more than two times the upper
and CCL7) in the bronchoalveolar fluid30. in a few patients, usually in the advanced limit of normal); prothrombin time >1 s or
These data suggest that lung macrophages stages of the disease33,37,38. Recently, low international normalized ratio >1.2; and the
contribute to inflammatory responses in antithrombin levels were linked to poor presence of thrombosis (macrothrombosis
SARS-CoV-2 infection, favouring excessive outcomes in COVID-19, especially and/or microthrombosis)45. The risk of
inflammation in severe pneumonia. Similar among patients with obesity39. Hence, COVID-19-associated coagulopathy is
to other respiratory infections, leukocyte it is possible that antithrombin activity defined by one of the aforementioned four
recruitment to the lungs is finely tuned changes as SARS-CoV-2 infection worsens criteria and one of the following ones:
by specific molecules31, the uncontrolled and a putative mechanism might be the increased fibrinogen levels; increased
activity of which may result in complications cytokine-driven imbalance in endogenous VWF levels (more than two times the
observed in patients with COVID-19 in both anticoagulant levels40. In macrophage upper normal limit); or the presence of
the lungs and other organs5,32. activation syndrome, liver function lupus anticoagulant and/or high-titre
impairment is a typical feature and may antiphospholipid antibodies45. Importantly,
COVID-19-associated coagulopathy. contribute to coagulopathy, yet liver COVID-19-associated coagulopathy and
An important pathophysiological feature impairment in COVID-19 is usually mild risk of COVID-19-associated coagulopathy
of COVID-19 is the development of a and transient, without overt acute liver may further progress and/or predispose to
prothrombotic state. The occurrence of injury41,42. DIC as COVID-19 severity increases.
a COVID-19-specific coagulopathy is Apart from a state of hypercoagulability
suggested by elevated levels of fibrinogen, in the lungs of patients with ARDS, a COVID-19-associated endothelial
von Willebrand factor (VWF) and the fibrin hypofibrinolytic state in the alveolar space dysfunction. Although COVID-19 should
degradation product D-dimer in the blood, was observed in COVID-19-related ARDS be regarded as a systemic disease, it acutely
whereas patients generally show minor due to increased levels of fibrinolytic causes a lung-centred injury primarily
or no changes in prothrombin time inhibitors43. Accordingly, high levels of affecting the vascular endothelium. COVID-
(a measure of time to clot), activated plasminogen activator inhibitor 1 (PAI1) 19-associated pneumonia is characterized by
partial thromboplastin time (coagulation were described in patients with SARS-CoV44. infiltration of the lungs with macrophages
time), antithrombin levels, activated In order to properly identify COVID- and neutrophils that cause diffuse
protein C levels and platelet count33–36. 19-associated coagulopathy, Iba et al. lung alveolar damage, the histological
equivalent of ARDS46–48. Similar findings
were previously reported in patients with
Box 1 | Immunothrombosis: a conserved mechanism of host defence SARS-CoV49. However, the striking finding
The term immunothrombosis was originally described by Engelmann and Massberg to refer to an in COVID-19 compared with SARS-CoV
intrinsic effector pathway of innate immunity triggered by pathogens and injured cells to reduce is the more extensive viral infection within
the spread and survival of the invading pathogens10,157. By contrast, the term ‘thromboinflammation’ the lungs, which results in more diffuse
refers to a process in which inflammation and thrombosis coexist within microvessels in response to inflammation involving the pulmonary
harmful stimuli (pathogens, injured cells or other irritants)158. Immunothrombosis is mainly triggered vessels50. This extensive infection reflects
by neutrophils and monocytes and is supported by the formation of microthrombi in small vessels, in the particular tropism of SARS-CoV-2 for
which endothelial cells exposed to microorganisms adopt a pro-adhesive phenotype159. angiotensin-converting enzyme 2 (ACE2) —
During immunothrombosis, neutrophils and monocytes release tissue factor and extracellular
the cellular receptor for SARS-CoV-2 —
nucleosomes and degrade endogenous anticoagulants, thereby facilitating activation of coagulation
induced by inflammation160. Importantly, tissue factor decorates neutrophil extracellular traps
expressed by type II pneumocytes51, which
(NETs), as found in sepsis161 and, more recently, in coronavirus disease 2019 (COVID-19)-related are anatomically close to the lung vascular
immunothrombosis86. NETosis by neutrophils also enhances the activity of the coagulation system network and are typically described to be
by increasing fibrin deposition162,163. Histones present in NETs, especially histones H3 and H4, can hyperplasic in samples from patients with
dose-dependently enhance thrombin generation not only by reducing thrombomodulin-mediated COVID-19 (refs52–54). This hyperplasia of
protein C activation164 but also by directly activating platelets141. Finally, NETs can bind plasma endothelial cells is likely to be caused by
proteins needed for platelet adhesion and thrombus development, such as fibronectin and von lung tissue ischaemia due to congestion of
Willebrand factor (VWF)141. In turn, platelets participate in this process in several ways: they increase small vessels by inflammatory cells (that
the accumulation of immune cells that express tissue factor10,165; they bind directly to neutrophils is, immunothrombosis) and thrombosis in
and NETs162,166,167; they release damage-associated molecular patterns that promote tissue factor
large vessels55.
expression within thrombi168; and they directly bind to microorganisms for presentation to innate
immune cells169,170.
Endothelial dysfunction was suggested
Microorganisms have evolved mechanisms to escape immunothrombosis, such as the expression to be an important pathophysiological event
of streptokinase to dissolve fibrin171 or nucleases to degrade NETs172,173. Some bacteria use in infections by other coronaviruses, which
immunothrombosis to avoid their killing, such as Staphylococcus aureus, whose staphylocoagulase and can directly infect endothelial cells56–59.
VWF-binding protein trigger fibrin generation to form a biofilm and an abscess pseudocapsule174,175. SARS-CoV-2 directly infects vascular
In sum, immunothrombosis can be considered a beneficial mechanism of intravascular immunity. endothelial cells and leads to cellular
However, when immunothrombosis is uncontrolled, it causes dysregulated activation of the damage and apoptosis, thus decreasing
coagulation cascade, leading to microthrombus formation and inflammation, which in turn the antithrombotic activity of the normal
enhance each other and may, ultimately, develop into thrombosis (thromboinflammation) and endothelium7,55. Alveolar damage, vessel wall
disseminated intravascular coagulation.
oedema, hyaline thrombi, microhaemorrhage

www.nature.com/nri

0123456789();:
Perspectives

P-selectin Pathogen
Endothelial cell
PRR Inactive TF Neutrophil
PSGL1
a Activation releasing NETs
of extrinsic
coagulation
pathway Inactivation
Active TF of natural
anticoagulants
Pathogen
e TFPA
NLRP3 TM
inflammasome Fibrin net C3a
C5a C3a
b Microvesicles Active TF H3
Blood Monocyte VWF NE
vessel Factor f
VWF
c XII H3 MPO g
IL-1β, H3
Activation of H4
IL-18 contact-dependent H4
coagulation pathway PEVs
Platelets
Factor VWF
XIIa
d
DNA

Pathogen isolation
and killing Microvascular
thrombus formation

Fig. 1 | Immunothrombosis is important in promoting immune defence. coagulation through the release of polyphosphates and, along with
Following recognition of a pathogen through pattern recognition receptors endothelial cells, may promote fibrin generation. Platelets can also be acti-
(PRRs), monocytes and monocyte-derived microvesicles present activated vated by C3a and C5a (path f). Activated platelets release large amounts of
tissue factor (TF) on their surfaces and release it at sites of pathogen locali- pro-inflammatory cytokines in platelet extracellular vesicles (PEVs) (path g).
zation, thus activating the extrinsic pathway of coagulation (path a). Through this mechanism, pathogens such as severe acute respiratory syn-
Pathogens also stimulate the NLRP3 inflammasome in monocytes and/or drome coronavirus 2 (SARS-CoV-2) become trapped within the fibrin-based
macrophages, leading to the release of pro-inflammatory cytokines, such NETs and killed. The immunothrombotic process allows pathogen killing
as interleukin-1β (IL-1β) and IL-18 (path b). Neutrophils are recruited and to be restricted to the intravascular compartment, thus limiting injury to
contribute to this process through the release of neutrophil extracellular organs. Although it is clear that immunothrombosis participates in
traps (NETs), which directly activate factor XII and, thus, the contact- SARS-CoV-2 pathogenesis, the exact mechanisms are still under investiga-
dependent pathway of coagulation (path c). NETs also bind von Willebrand tion. These may include the following: direct injury of endothelial cells by
factor (VWF) and help to recruit platelets (path d). Histones, in particular the virus and consequent activation of the coagulation cascade; infiltration
H3 and H4, trigger activation of platelets. In addition, neutrophil elastase of neutrophils that lead to NET formation; induction of hypoxaemia causing
(NE) and myeloperoxidase (MPO) in NETs cleave and inactivate natural anti- upregulation of TF expression by hypoxia-inducible transcription factors
coagulants (tissue factor pathway inhibitor (TFPA) and thrombomodulin and formation of clots; activation of complement that promotes coagula-
(TM)) (path e). Finally, NETs can externalize and bind TF, promoting activa- tion and recruits and activates platelets, monocytes and neutrophils,
tion of the extrinsic pathway of coagulation. Platelets support the immuno- thus triggering TF expression; and an abnormal increase in the levels of
thrombotic process by activating the contact-dependent pathway of pro-inflammatory cytokines causing direct cell damage.

and diffuse thrombosis of peripheral small (ADAMTS13), a metalloproteinase that in patients admitted to the ICU than in
vessels have emerged as key features of regulates the size of VWF multimers63. patients not in the ICU, whereas increased
COVID-19 contributing to respiratory Endothelial cell perturbation along with levels of thrombomodulin (a specific
failure6,8,46 (Fig. 2). the release of large VWF multimers and marker of endothelial activation generally
Endothelial cells of lung blood vessels relative insufficient VWF cleavage owing to released during endothelial cell injury)
can be activated by the high levels of ADAMTS13 consumption may account for were associated with increased mortality
pro-inflammatory cytokines (IL-1, IL-6 increased platelet–vessel wall interactions risk35. Higher numbers of circulating
and TNF) and ferritin in severe COVID-19 causing thrombotic microangiopathy. Indeed, endothelial cells were described in patients
(ref.60). Moreover, increased levels of the a relative deficiency in ADAMTS13 activity with COVID-19, especially among those
endothelial adhesion protein VWF were was previously described in inflammatory admitted to the ICU, and the presence
reported both in patients admitted to the conditions with high levels of IL-6, as found of circulating endothelial cells positively
ICU and in non-critically ill patients34,35,61,62, in patients with SARS-CoV-2 infection64. correlated with platelet and lymphocyte
and this increase seems to be associated Other markers of endothelial cell injury have counts and with the classical endothelial
with a mild reduction in the activity of a been recently investigated. Higher levels of marker soluble vascular cell adhesion
disintegrin and metalloproteinase with a soluble P-selectin (a marker of endothelial molecule 1 (sVCAM1)65. This was recently
thrombospondin type 1 motif, member 13 and platelet activation) were observed confirmed by another report from Italy,

NATuRE REVIEWS | Immunology

0123456789();:
Perspectives

showing that patients with COVID-19 had of endothelial dysfunction in COVID-19 endothelial dysfunction in pulmonary
increased circulating endothelial cells and (ref.66). microvessels acts as a trigger for
higher plasma levels of soluble intercellular Considering the important role of the immunothrombosis, resulting in the
adhesion molecule 1 (sICAM1) and endothelium in regulating haemostasis, coagulopathy observed in patients with
sVCAM1, further supporting the concept fibrinolysis and vessel wall permeability, COVID-19 (refs27,47,67,68). In addition,
endothelial dysfunction is recognized
A as a risk factor of microvascular
a b dysfunction through a shift towards
vasoconstriction, by promoting ischaemia,
inflammation and a procoagulant
state69. In addition, the inflammatory
environment triggers the expression of
activated tissue factor on endothelial cells,
macrophages and neutrophils, which
amplifies activation of the coagulation
cascade within the lungs60. These events
200 μm 100 μm fuel a positive immunothrombotic
feedback loop in which hyperinflammation
c d triggers the hypercoagulant state, with
thrombosis occurring within pulmonary
microvessels. Lastly, platelet activation
within the microvasculature is increased
in patients with severe COVID-19 and is
associated with poor outcomes70,71. Platelet–
vessel wall interactions, mediated by surface
receptors (integrins and selectins) and
adhesive proteins (VWF and fibrinogen),
80 μm impair vessel wall integrity or increase
200 μm
endothelial injury, ultimately leading to
B microvascular obstruction72. Moreover,
a b platelets were found to be hyperactivated
in patients with COVID-19 and to be
a source of procoagulant extracellular
vesicles73 and polyphosphates, which can
exacerbate the thromboinflammatory
cascade via neutrophils74. Of importance,
activated platelets are critically involved
in neutrophil extracellular trap (NET)
900 μm 600 μm formation71, which is an essential
element of immunothrombosis. All of
c d e these elements support the hypothesis
that endothelial dysfunction, or
endotheliopathy35,75, and platelet activation
are key features of COVID-19-associated
coagulopathy and may mediate the damage
leading to severe disease.
60 μm
COVID-19-associated ARDS. COVID-
Fig. 2 | Seminal pathology features of patients with CoVID-19. A | Hyaline membrane formation 19-related ARDS is considered atypical,
is a typical finding in patients with alveolar damage irrespective of being ventilated or not (blue arrows with its main characteristic being a
in panels a and b). Organizing fibrosis (blue arrowhead) and fibrin thrombi in small blood vessels (black dissociation between hypoxaemia
arrows) with oedema, along with extensive haemorrhage (haematoxylin and eosin staining) and hya- severity and relatively good respiratory
line membranes (blue arrows) (panel c). The bronchial respiratory mucosa is almost entirely intact and mechanics76,77. From a clinical standpoint,
no squamous metaplasia is evident (panel d), different to that observed in patients with severe acute two phenotypes are described67,78. Patients
respiratory syndrome coronavirus 2 (SARS-CoV-2). B | Two thrombi, one in a small pulmonary artery with ‘type L’ have high lung compliance,
(blue arrow) and one in a pulmonary venule (blue arrowhead), in the haematoxylin and eosin staining lower lung weight as estimated by computed
(panel a) and in the CD61 immunostaining for platelets within thrombi (panel b). Megakaryocytes
tomography scan and poor response to
within small vessels and alveolar capillaries (panel c, blue arrow). CD61 immunostaining of a fibrin- and
positive end-expiratory pressure. Patients
platelet-rich thrombus in a small vessel (panel d), with a megakaryocyte stained below (blue arrow).
Small, perivascular aggregates of lymphocytes (panel e). COVID-19, coronavirus disease 2019. Images with ‘type H’ are those with a deteriorating
reprinted from The Lancet, 8, Fox, S. E. et al., Pulmonary and cardiac pathology in African American disease or suboptimally treated disease
patients with COVID-19: an autopsy series from New Orleans, 681–686, Copyright (2020), with that present with areas of accumulation
permission from Elsevier (ref.6). of inflammatory exudate in the alveoli as

www.nature.com/nri

0123456789();:
Perspectives

Box 2 | The role of complement in sepsis strongly stimulate the production of NETs
from healthy neutrophils71,80,85. In addition,
The complement system is an important component of the innate immune system and activates neutrophils isolated from patients with
following infections. After complement activation (through the classic, alternative or lectin COVID-19 showed elevated NET release
pathways), the common pathway is responsible for the production of C3a and C5a, then stimulating
at baseline similar to phorbol myristate
formation of the C5b–9 membrane attack complex, which causes the lysis of pathogen cells176.
acetate (PMA)-stimulated neutrophils
In a mouse model of intranasal infection with severe acute respiratory syndrome coronavirus
(SARS-CoV), early activation of the complement system, especially complement component C3, from healthy donors, indicating that the
caused acute respiratory distress syndrome (ARDS)177. However, C3-deficient mice experienced COVID-19 plasma environment promotes
reduced respiratory dysfunction, limited neutrophil and monocyte lung infiltration, and showed NET formation80. The virus itself is also
lower levels of cytokines both in the lungs and the blood177. Excessive activation of complement in reported to directly activate neutrophils
the advanced phases of sepsis may therefore cause tissue injury contributing to organ failure. towards NETosis, promoting epithelial cell
The complement and coagulation pathways are closely linked178,179. In particular, complement end death89. NETs may provide a scaffold for the
products can trigger the release of procoagulant products and reduce the production of natural deposition of C3, properdin and factor B,
anticoagulants178. Additionally, the binding of C1q to its receptor on platelets triggers the expression thus directly participating in complement
of integrins and P-selectin180, whereas C3 stimulates platelet activation and aggregation. The
activation90. This mechanism may fuel
activation of C5 during the inflammatory response is responsible for the expression of tissue factor
an amplifying feedback loop promoting
on leukocytes and endothelial cells181,182. Complement may indirectly induce procoagulant changes;
for example, activated C3 and C5 contribute to cytokine release soon after the innate immune further NET formation and platelet and
response starts, particularly increasing the concentrations of tumour necrosis factor (TNF) and endothelial cell activation91. The observation
interleukin-6 (IL-6). of antiphospholipid antibodies, which can
directly stimulate NETosis92, in patients with
COVID-19, as well as in other infections93,
observed through computed tomography SARS-CoV-2 infection80,85. Compared suggests that SARS-CoV-2 infection might
scan, low lung compliance, greater lung with controls, patients with COVID-19 synergize with antiphospholipid antibodies
weight and good response to positive have higher levels of serum85 or plasma80 to promote the immunothrombotic process.
end-expiratory pressure. This clinical picture myeloperoxidase (MPO)–DNA complexes, Indeed, higher titres of antiphospholipid
couples with endothelial damage, breaking which are biomarkers of circulating antibodies were associated with increased
down pulmonary vasoregulation, stimulating NET fragments, and further increases activity of neutrophils and platelets and more
ventilation–perfusion mismatch — which is were found in those patients requiring severe respiratory disease94.
mainly responsible for hypoxaemia — and invasive mechanical ventilation. Whether NETs are direct contributors
supporting immunothrombosis. In this MPO–DNA complexes and thrombin– to the dysregulated cytokine response is
respect, the events leading to atypical antithrombin complexes were positively still unclear. Indeed, NETs participate in
ARDS can be summarized by the term correlated in patients with COVID-19, and acute lung injury by inducing macrophage
‘microvascular COVID-19 lung vessels the anticoagulant dabigatran or complement release of IL-1β, which in turn can
obstructive thromboinflammatory targeting inhibited NETosis86, further increase NET formation95,96. In addition,
syndrome’ (MicroCLOTS)79. According to suggesting a central role for NETs in the most inflammatory mediators that are
this proposed definition, ARDS is the result immunothrombotic milieu. MPO–DNA increased in patients with COVID-19
of a hyperimmune reaction of the host levels return to normal in convalescent are known to regulate neutrophil activity
to SARS-CoV-2 that is both directly plasma, indicating that this is a transient through the expression of chemotactic
mediated by the virus through cellular increase80. High-dimensional flow products16,97. These data suggest that
injury and release of pro-inflammatory cytometric analysis of circulating neutrophils dysregulated cytokine release might be
signals and indirectly mediated by revealed a highly activated phenotype in sustained by crosstalk between neutrophils
complement activation (Box 2), leading patients with most severe COVID-19 (ref.71). and macrophages mediated by NETs,
to leukocyte recruitment and increased Neutrophils can be found embedded in fibrin resulting in a deranged or exaggerated
release of pro-inflammatory cytokines. clots and in close association with platelets immunothrombotic status.
This results in a severe tissue injury with within microthrombi in the lungs80, heart
vascular endothelial dysfunction and and kidneys of patients with COVID-19 COVID-19: a syndrome of deregulated
alveolar cell damage, which may explain (ref.71). In severe disease, neutrophils adopted immunothrombosis? On the basis of the
the progressive worsening of ventilation– the so-called low-density phenotype that aforementioned evidence, we propose that
perfusion mismatch and pulmonary is more prone to spontaneously form exaggerated immunothrombosis, occurring
microthrombosis79. NETs; this greater NETosis was linked for the most part within lung microvessels,
to increased intravascular aggregation, drives the clinical manifestations of
NETs in COVID-19-associated leading to microthrombosis and COVID-19 (ref.3), although systemic
immunothrombosis. NETs are likely organ damage87. NETs have also been viraemia early in the disease course may
to be important players in deregulated identified in the lung parenchyma and also be important98 (Fig. 1). The occurrence
immunothrombosis in COVID-19 (ref.80). alveolar space in autopsy case reports9,88. of severe events, such as acute respiratory
Indeed, ARDS can be considered as a Interestingly, neutrophils from healthy failure and ARDS, may be explained by a
NETopathy81; higher levels of NETs were donors showed increased adhesion to deregulated process of immunothrombosis
described in the plasma and bronchoalveolar activated platelets in plasma from patients involving the pulmonary microcirculation.
fluid of patients with transfusion-associated with COVID-19 compared with that from In support of this, there is increasing
and pneumonia-related ARDS compared control patients, causing enhanced NET evidence that COVID-19 is a disease of
with those without ARDS82–84. NETs are generation71. Indeed, both serum and the endothelium35 that results in elevated
markers of disease severity in patients with plasma from patients with COVID-19 levels of PAI1 and VWF, increased platelet

NATuRE REVIEWS | Immunology

0123456789();:
Perspectives

activation and a state of hypercoagulability, antithrombin-dependent factor Xa Multi-factorial Adaptive Platform Trial
with consequent venous, arterial and inhibitor) in patients with COVID-19 was for Community-Acquired Pneumonia
microvascular thrombosis. The exact factors reported in two papers, with contrasting (REMAP-CAP) (NCT02735707); the
triggering this endotheliopathy have not results about its safety109,110. The same applies Accelerating COVID-19 Therapeutic
yet been recognized, but potentially include to direct oral anticoagulants, the use of Interventions and Vaccines-4 (ACTIV-4)
the virus itself (causing endothelial cell which in patients with COVID-19 has been trial (NCT04505774); and the
injury that triggers the coagulation cascade little studied to date111,112. Several trials of Antithrombotic Therapy to Ameliorate
involving fibrin and platelets47,99), immune heparin and other anticoagulant agents are Complications of COVID-19 (ATTACC)
cell (neutrophil and/or macrophage and in progress: the Randomized, Embedded, trial (NCT04372589). These trials are
platelet73,100,101) infiltration, hypoxaemia
(that can induce hypoxia-inducible
glossary
transcription factors that upregulate
tissue factor expression102 and lead to Antithrombin Microthrombosis
fibrin-based clot formation, thus supporting A glycoprotein produced by the liver that inactivates The formation of thrombi (clots) within small vessels,
different enzymes of the coagulation cascade. which is typical of the immunothrombotic process.
a thromboinflammatory feedback loop), Antithrombin activity is strongly increased by the Microthrombosis is often encountered in patients with
complement-mediated damage or a surge in anticoagulant medication heparin, which enhances coronavirus disease 2019 (COVID-19).
pro-inflammatory cytokine (such as IL-1β the binding of antithrombin to factor IIa (thrombin)
and IL-6) release with direct cell damage. and factor Xa. NLRP3 inflammasome
Macromolecular structure that is responsible for sensing
Cytokine storm danger and triggering a local or systemic inflammatory
Thrombosis management in COVID-19 A sudden surge in circulating levels of pro-inflammatory response. Following NLRP3 inflammasome activation,
If we consider COVID-19 as a vascular cytokines, such as interleukin-1 (IL-1), IL-6, tumour large quantities of pro-inflammatory cytokines (in
disease primarily involving the endothelium, necrosis factor (TNF) and interferon-γ (IFNγ). particular IL‑1β) are released for extracellular secretion.
an ideal therapeutic approach would be both Clinically, this can result in hypotension, acute These cytokines are important in the inflammatory
renal failure, impaired pulmonary function and response and responsible for symptoms such as fever.
antithrombotic and anti-inflammatory. Here, even death.
we describe some of the main approaches Neutrophil extracellular trap
that are being explored to address these D-dimer (NET). Fibrillary structure consisting of DNA complexed with
features of COVID-19 (Table 1). A small protein derived from fibrin degradation histones (H3 and H4) and proteins from neutrophil granules,
that is found in the blood once a blood clot is especially neutrophil elastase (NE), myeloperoxidase
degraded. The name comes from the presence (MPO) and cathepsin G. NETs act as a mesh that traps
Antithrombotic therapies. As the of two D fragments of the fibrin protein united by microorganisms and exposes them to neutrophil-derived
coagulation cascade is dysregulated in a cross-link. effector molecules. NETs also provide a scaffold for
COVID-19, anticoagulation approaches thrombus formation and activation of coagulation.
have been explored for the treatment, Disseminated intravascular coagulation
(DIC). An acquired syndrome characterized by the Positive end-expiratory pressure
particularly of patients with incident intravascular activation of coagulation without a Pressure within the lung alveoli above atmospheric
venous thromboembolism (VTE) or precise localization in response to microvascular injury, pressure that is recorded at the end of expiration. Besides
acute coronary syndrome due to plaque leading to organ dysfunction when sufficiently intrinsic positive end-expiratory pressure (caused by an
rupture103. Indeed, a study reported that 40% severe. DIC involves large production of thrombin incomplete exhalation), extrinsic positive end-expiratory
accompanied by platelet activation and consumption, pressure is provided by a ventilator during mechanical
of patients hospitalized with COVID-19
altered fibrinolysis and microvascular thrombosis. ventilation and is meant to reduce end-expiratory alveolar
are at high risk of VTE104 and data from a It can occur in critically ill patients with overwhelming collapse, thus improving hypoxaemia or reducing
large retrospective cohort of patients in the infection, fulminant sepsis, extensive tissue damage or ventilator-associated lung injury in patients with acute
United States suggested that anticoagulation malignancy. respiratory distress syndrome (ARDS).
may reduce mortality, especially among
Haemostasis Thrombosis
those with severe disease requiring invasive The combination of events that result in cessation of The formation of a thrombus (clot) inside blood vessels,
mechanical ventilation105. Pharmacological bleeding. Haemostatic processes include constriction resulting in partial or complete vessel occlusion.
VTE prophylaxis, including daily low of blood vessels, platelet aggregation and coagulation
molecular weight heparin or twice-daily (clotting) of the blood. Thrombotic microangiopathy
Pathology characterized by formation of thrombi in
subcutaneous unfractionated heparin, may
Hyaline thrombi capillaries owing to endothelial damage. In particular,
also be of benefit to patients hospitalized Clots occurring in small vessels due to the formation decreased levels of the protease a disintegrin and
with COVID-19 who are bedridden or need of hyaline membranes and surfactant dysfunction metalloproteinase with a thrombospondin type 1 motif,
intensive care, or patients discharged from after an inflammatory injury. These formations are member 13 (ADAMTS13) lead to an accumulation of
hospital but with relevant risk of VTE — the pathological equivalent of acute respiratory ultra-large von Willebrand factor (VWF) multimers, which
distress syndrome (ARDS) occurring in patients promote platelet–vessel wall interactions, causing thrombus
that is, individuals with reduced mobility, with severe-to-critical coronavirus disease 2019 formation. The clinical presentation comprises multiple
pro-thrombotic comorbidities (active (COVID-19). organ dysfunctions, especially in the brain and kidneys.
cancer or obesity), older age, a history
of VTE and persistently elevated levels of Lung compliance Ventilation–perfusion mismatch
A measure of the ability of the lungs to stretch and Impairment of the ventilation to perfusion ratio, meaning
D-dimer103,106,107.
expand. A low compliance refers to that of a stiff lung, that a part of the lung is adequately oxygenated but not
Studies of heparin treatment of as occurs in fibrosis. A high compliance means a flexible perfused, or vice versa, caused by diseases such as
COVID-19 have shown positive results103,106 lung, as seen in emphysema. pneumonia and pulmonary embolism.
(Supplementary Table 1), and this is now one
of a growing number of agents available to Macrophage activation syndrome Von Willebrand factor
A syndrome in which a dysregulated activation (VWF). The main adhesive protein mediating interactions
manage COVID-19 either as a prophylactic and proliferation of macrophages and T cells between platelets and the vessel wall, leading to platelet
or therapeutic regimen108 (Table 1). The leads to increased release of pro-inflammatory aggregation. Preformed multimers are released from
use of fondaparinux (a selective indirect cytokines. Weibel–Palade bodies of activated endothelial cells.

www.nature.com/nri

0123456789();:
Perspectives

Table 1 | Putative therapeutic approaches for CoVID-19


Therapeutic agent Target outcome Clinical trial Refs
Antithrombotic drugs
Heparin Inactivation of thrombin and Controversial evidence on mortality – 105,152–154

activated factor X through an and need for mechanical ventilation


antithrombin-dependent mechanism from observational studies
Garadacimab Factor II blockade One study is ongoing NCT04409509 –
Nafamostat Serine protease inhibitor of thrombin, Ongoing trials based on in vitro NCT04352400, NCT04418128 –
mesylate plasmin and trypsin blockade of MERS-CoV infection
Tissue-type Conversion of plasminogen into Benefit in a small study of patients NCT04357730 119

plasminogen plasmin, leading to breakdown of with severe COVID-19


activator cross-linked fibrin A phase IIa trial in patients with
COVID-19-related ARDS is ongoing
Dipyridamole Phosphodiesterase inhibitor In vitro suppression of SARS-CoV-2 NCT04391179 121,124,125

reversibly inhibiting platelet replication and improvement of


aggregation; in addition, it potentiates lung pathology in a model of viral
vascular-protective effects of pneumonia
endothelium-derived nitric oxide
In 14 patients, reported improvement
Possible activity against in clinical status compared with
positive-stranded RNA viruses and controls
prevention of NET formation
A trial is ongoing
Ticagrelor P2Y12-receptor antagonist and Ongoing trials NCT02735707 , NCT04518735 –
attenuation of NET formation
Anti-inflammatory drugs
NE inhibitors NE inhibition blocks NET formation Sivelestat has been approved to treat – 131–133

and reduces SARS-CoV-2 spike protein ARDS in Japan and South Korea, but
proteolytic activation findings are inconsistent
No current indication is available for
SARS-CoV-2-related ARDS
DNase I Dissolution of NETs Dornase alpha has been tested in NCT04402944, NCT04355364, 138,139

two case studies in patients with NCT04432987 , NCT04359654,


COVID-19-associated ARDS NCT04445285, NCT04402970
Several trials are ongoing
Ruxolitinib JAK1/2 inhibitor In a small pilot study, clinical NCT04338958 142,155,156

improvement was observed after


1 week of treatment
In a small RCT, ruxolitinib plus SOC
did not significantly accelerate
clinical improvement in patients with
severe COVID-19
A trial is ongoing in Germany
Eculizumab and C5 inhibitors blocking MAC formation Eculizumab showed good efficacy in NCT04288713, NCT04369469, 147,148

ravulizumab reducing inflammatory biomarkers NCT04390464


In ICU patients, eculizumab improved
15-day survival and hypoxia
compared with SOC
Trials with eculizumab and
ravulizumab are ongoing
AMY-101 Inhibition of C3 cleavage by direct Limited experience is now available, EudraCT number 2020-001550- 149

binding with encouraging results 22, EudraCT number 2020-


004408-32, NCT04395456
Three phase II clinical trials in patients
with COVID-19-associated ARDS are
ongoing
ARDS, acute respiratory distress syndrome; COVID-19, coronavirus disease 2019; ICU, intensive care unit, JAK, Janus kinase; MAC, membrane attack complex;
MERS-CoV, Middle East respiratory syndrome coronavirus; NE, neutrophil elastase; NET, neutrophil extracellular trap; RCT, randomized controlled trial; SARS-CoV-2,
severe acute respiratory syndrome coronavirus 2; SOC, standard of care.

testing whether the use of full-dose versus However, enrolment of critically ill patients therapy may depend on the time of
low-dose unfractionated heparin or low has been temporarily stopped owing to administration; if given too late, thrombi in
molecular weight heparin benefits patients futility and safety concerns for this subgroup the microcirculation and large vessels may
with COVID-19 requiring ICU admission. of patients. Response to this anticoagulant already be too extensive.

NATuRE REVIEWS | Immunology

0123456789();:
Perspectives

An alternative approach to Anti-inflammatory therapies. released NETs include JAK1/2 inhibition


pharmacologically disrupt the coagulation Immunomodulatory therapies, such as with ruxolitinib142, dipyridamole124,
cascade involves blockade of factor XII inhibitors of IL-6, IL-1, GM-CSF and Janus ticagrelor126 and neonatal NET-inhibitory
(FXII), which is responsible for activation kinases (JAKs), aim to blunt the cytokine factor, an endogenous peptide produced
of the contact pathway (Table 1). FXII dysregulation associated with severe in newborns that restricts the ability of
blockade has been shown to protect from COVID-19 (refs15,127,128) (Table 1), but little is neutrophils to produce NETs143. Neonatal
occlusive thrombosis in animal models known about their impact on the thrombotic NET-inhibitory factor was shown to prevent
without impairing haemostasis113,114. Thus, complications of the disease. COVID-19 plasma-induced NET formation
a FXII-blocking antibody (garadacimab) is Given that NETs have a direct role by healthy donor neutrophils in vitro, but
currently being explored in a multicentre, in the immunothrombotic process in the mechanism of action remains to be
double-blind, placebo-controlled phase II COVID-19, blocking NETs may help elucidated80.
trial in patients with respiratory distress to improve prognosis (Table 1). Some Finally, evidence is accumulating on
and COVID-19 (NCT04409509). Similarly, drugs targeting NETs already exist or are the role of complement activation in the
a serine protease inhibitor of thrombin, under development, such as inhibitors pathogenesis and severity of SARS-CoV-2
plasmin and trypsin known as nafamostat of neutrophil elastase (NE), PAD4 (ref.144) as well as on complement-mediated
mesylate (used in Japan for the treatment of and gasdermin D129. For example, NE thrombotic microangiopathy in patients
pancreatitis and DIC and the prevention inhibitors can block NET formation, limit with COVID-19 (ref.145). Thus, therapies
of clots during dialysis115) is being the toxic activities of NE itself on lung for thrombotic microangiopathy —
investigated in clinical trials for COVID-19 connective tissue and reduce spike protein complement inhibitors — may be used in
(NCT04352400, NCT04418128). This drug proteolytic activation that is responsible this setting. C3 blockade can inhibit the
showed potential to block Middle East for SARS-CoV-2 binding to host cells130. production of both C3a and C5a, but also of
respiratory syndrome coronavirus infection Sivelestat, an NE inhibitor, has been intrapulmonary C3a and the release of IL-6
in vitro116 (Table 1). approved to treat ARDS in Japan and South from alveolar macrophages146. Currently,
The hypofibrinolytic state observed Korea, but findings are inconsistent as the two FDA-approved complement inhibitors
in ARDS has been targeted by the use severity of lung injury is likely to represent are available, eculizumab and ravulizumab,
of tissue-type plasminogen activator, the most important predictor for patient both of which bind to C5 and sterically
which is responsible for the conversion outcome131–133. However, no evidence is block the cleavage of C5 to C5a, and thus
of plasminogen into plasmin, leading to currently available in favour of using NE membrane attack complex formation.
the breakdown of cross-linked fibrin117,118. inhibitors for COVID-19-induced ARDS. Four patients with COVID-19 receiving
Indeed, tissue-type plasminogen activator NETs can be degraded by recombinant eculizumab intravenously showed a prompt
infusion showed benefit in three patients with DNase I (dornase alpha), which is reduction in C-reactive protein levels and
severe COVID-19 (ref.119), and a phase IIa currently given by inhalation to patients an improvement of the lesions observed
trial in patients with COVID-19-related with cystic fibrosis to target NETs in the through chest computed tomography scan147.
ARDS is currently ongoing (NCT04357730). airways134. Dornase alpha may help to Among 35 patients admitted to the ICU with
The antiplatelet agent dipyridamole may dissolve respiratory secretions in patients COVID-19, eculizumab treatment improved
have therapeutic potential120 (Table 1). This with COVID-19 (ref.135), reducing the risk 15-day survival and hypoxia compared with
agent, apart from its antiplatelet function, for secondary infections and improving patients treated with standard care alone148.
was shown to provide broad-spectrum ventilation, and may also reduce the risk Trials with eculizumab (NCT04288713)
antiviral activity (especially against of progression towards ARDS, as shown in and ravulizumab (NCT04369469,
positive-stranded RNA viruses), suppress animal models136,137. Two case studies have NCT04390464) are currently ongoing.
inflammation and favour mucosal healing, shown that administration of dornase alpha In a report from Italy, a patient with
and prevent acute injury and fibrosis in the is well tolerated in patients with COVID-19- severe COVID-19-related ARDS was
lungs, heart and kidney121–123. Dipyridamole associated ARDS138,139, and several trials are safely and successfully treated with the
treatment in patients with COVID-19 may underway to determine the efficacy of this compstatin-based complement C3 inhibitor
also have the added benefit of preventing approach (NCT04402944, NCT04355364, AMY-101 (ref.149). A recent study comparing
NETosis by promoting 3′,5′-cyclic adenosine NCT04432987, NCT04359654, two different strategies targeting C3 and
monophosphate (cAMP) generation in NCT04445285, NCT04402970). Nebulizer C5 has shown a general beneficial impact
neutrophils, as was shown in the context administration of DNase provides direct in patients with COVID-19, although C3
of antiphospholipid syndrome124. Liu et al. access to NETs within the alveolar space, but inhibition led to a broader therapeutic
demonstrated that dipyridamole can there may be a barrier to reaching systemic effect blocking cytokine dysregulation and
suppress SARS-CoV-2 replication in vitro efficacy owing to the high reported levels of NET generation, thereby having a positive
and improve lung pathology in an animal platelet factor 4 (PF4) in severe COVID-19 impact on immunothrombosis-driven
model of viral pneumonia through a type I (ref.80). PF4 binding to NETs leads to their disease pathways150. The C3 inhibitor
interferon response125. This drug was tested compaction and decreased susceptibility AMY-101 is currently being evaluated in
in 14 patients and led to an improvement in to DNase degradation140. Therefore, DNase three phase II clinical trials in patients with
clinical status compared with controls125. administration is unlikely to ameliorate COVID-19-associated ARDS (EudraCT
A trial of dipyridamole is currently ongoing NET-mediated thrombosis, although the number 2020-001550-22, EudraCT number
(NCT04391179). Another antiplatelet agent, combination with heparin (which also digests 2020-004408-32, NCT04395456). Another
ticagrelor, showed the ability to attenuate NETs141) may be able to overcome this DNase C3 inhibitor, the pegylated compound
NET formation126. Accordingly, two trials of resistance. APL-9, is being tested in a phase I/II
ticagrelor are ongoing or recently completed Other promising approaches to prevent randomized controlled trial for severe
(NCT02735707, NCT04518735). NET formation rather than target already COVID-19 (NCT04402060). The C1

www.nature.com/nri

0123456789();:
esterase inhibitor ruconest (NCT04414631, 10
First Clinic of Internal Medicine, Department of 22. Blanco-Melo, D. et al. Imbalanced host response to
Internal Medicine and Centre of Excellence for SARS-CoV-2 drives development of COVID-19. Cell
NCT04530136) and the C5a inhibitor IFX-1 181, 1036–1045.e9 (2020).
Biomedical Research (CEBR), University of Genoa,
(NCT04333420) are being tested as well. Genoa, Italy.
23. Toldo, S. et al. Inflammasome formation in the lungs of
patients with fatal COVID-19. Inflamm. Res. 70, 7–10
11
Deutsches Zentrum für Herz-Kreislauf-Forschung (2021).
Conclusions (DZHK) e.V., Partner Site Munich Heart Alliance,
24. Rodrigues, T. S. et al. Inflammasomes are activated in
response to SARS-CoV-2 infection and are associated
Progressive advancements in our Munich, Germany. with COVID-19 severity in patients. J. Exp. Med. 218,
understanding of the pathophysiology of 12
Medizinische Klinik und Poliklinik I, Klinikum der e20201707 (2021).
25. Quartuccio, L. et al. Profiling COVID-19 pneumonia
COVID-19 have been crucial for better Universität, Ludwig-Maximilians-Universität, Munich,
progressing into the cytokine storm syndrome: results
management of COVID-19. Severe Germany. from a single Italian centre study on tocilizumab
versus standard of care. J. Clin. Virol. 129, 104444
COVID-19 is not limited to the respiratory Department of Medicine and Cardio-metabolic
13
(2020).
Programme — NIHR UCLH/UCL BRC, University College
tract but is a multisystem syndrome in 26. Henry, B. M., de Oliveira, M. H. S., Benoit, S.,
London Hospitals NHS Foundation Trust, London, UK. Plebani, M. & Lippi, G. Hematologic, biochemical and
which the vascular endothelium is the most ✉e-mail: aldo.bonaventura@asst-settelaghi.it immune biomarker abnormalities associated with
damaged organ, characterized by a state of severe illness and mortality in coronavirus disease
https://doi.org/10.1038/s41577-021-00536-9 2019 (COVID-19): a meta-analysis. Clin. Chem. Lab.
immunothrombosis within microvessels Med. 58, 1021–1028 (2020).
(MicroCLOTS) and a predisposition to Published online xx xx xxxx 27. McGonagle, D., O’Donnell, J. S., Sharif, K.,
Emery, P. & Bridgewood, C. Immune mechanisms of
macrothrombosis. McGonagle et al. recently 1. Pascarella, G. et al. COVID-19 diagnosis and pulmonary intravascular coagulopathy in COVID-19
pointed out the potential role of thrombosis management: a comprehensive review. J. Intern. Med. pneumonia. Lancet Rheumatol. 2, e437–e445
288, 192–206 (2020). (2020).
in pulmonary veins distal to the alveolar 2. Bonaventura, A. et al. Targeting GM-CSF in COVID-19 28. Wang, C. et al. Alveolar macrophage dysfunction
capillary bed, which should act as a clot filter, pneumonia: rationale and strategies. Front. Immunol. and cytokine storm in the pathogenesis of two
11, 1625 (2020). severe COVID-19 patients. EBioMedicine 57, 102833
suggesting this could be a SARS-CoV-2- 3. McFadyen, J. D., Stevens, H. & Peter, K. The emerging (2020).
related vasculitis responsible for ischaemic threat of (micro)thrombosis in COVID-19 and its 29. Adachi, T. et al. Clinicopathologic and
therapeutic implications. Circ. Res. 127, 571–587 immunohistochemical findings from autopsy of
manifestations in different organs151. Based (2020). patient with COVID-19, Japan. Emerg. Infect. Dis. 26,
on all of these features, anticoagulant 4. Ranucci, M. et al. The procoagulant pattern of patients 2157–2161 (2020).
with COVID-19 acute respiratory distress syndrome. 30. Liao, M. et al. Single-cell landscape of bronchoalveolar
therapy and immunomodulatory J. Thromb. Haemost. 18, 1747–1751 (2020). immune cells in patients with COVID-19. Nat. Med.
agents are likely necessary to blunt the 5. Gupta, A. et al. Extrapulmonary manifestations of 26, 842–844 (2020).
COVID-19. Nat. Med. 26, 1017–1032 (2020). 31. Alon, R. et al. Leukocyte trafficking to the lungs
hyperinflammatory and prothrombotic 6. Fox, S. E. et al. Pulmonary and cardiac pathology in and beyond: lessons from influenza for COVID-19.
conditions. Evidence from clinical trials is African American patients with COVID-19: an autopsy Nat. Rev. Immunol. 21, 49–64 (2021).
series from New Orleans. Lancet Respir. Med. 8, 32. Tay, M. Z., Poh, C. M., Renia, L., MacAry, P. A.
still limited and consensus-based guidelines 681–686 (2020). & Ng, L. F. P. The trinity of COVID-19: immunity,
recommend anticoagulation therapy based 7. Wichmann, D. et al. Autopsy findings and venous inflammation and intervention. Nat. Rev. Immunol.
thromboembolism in patients with COVID-19: a 20, 363–374 (2020).
on an individual risk profile. With regard prospective cohort study. Ann. Intern. Med. 173, 33. Tang, N., Li, D., Wang, X. & Sun, Z. Abnormal
to immunomodulatory therapies, evidence 268–277 (2020). coagulation parameters are associated with poor
is accumulating about a beneficial role in 8. Carsana, L. et al. Pulmonary post-mortem findings
in a series of COVID-19 cases from northern Italy:
prognosis in patients with novel coronavirus pneumonia.
J. Thromb. Haemost. 18, 844–847 (2020).
COVID-19. Timing of administration, a two-centre descriptive study. Lancet Infect. Dis. 20, 34. Panigada, M. et al. Hypercoagulability of COVID-19
however, is a challenging point that needs 9.
1135–1140 (2020).
Schurink, B. et al. Viral presence and immunopathology
patients in intensive care unit: a report of
thromboelastography findings and other parameters
to be carefully considered, and data about a in patients with lethal COVID-19: a prospective autopsy of hemostasis. J. Thromb. Haemost. 18, 1738–1742
consistent effect on immunothrombosis are cohort study. Lancet Microbe 1, e290–e299 (2020). (2020).
10. Engelmann, B. & Massberg, S. Thrombosis as an 35. Goshua, G. et al. Endotheliopathy in COVID-19-
still lacking. intravascular effector of innate immunity. Nat. Rev. associated coagulopathy: evidence from a single-centre,
Immunol. 13, 34–45 (2013). cross-sectional study. Lancet Haematol. 7, e575–e582
Aldo Bonaventura   1,2,3 ✉, Alessandra Vecchié1,3, 11. Wiersinga, W. J., Rhodes, A., Cheng, A. C., Peacock, S. J. (2020).
Lorenzo Dagna4,5, Kimberly Martinod   6, & Prescott, H. C. Pathophysiology, transmission, 36. Iba, T., Levy, J. H., Levi, M., Connors, J. M. &
Dave L. Dixon1,7, Benjamin W. Van Tassell1,7, diagnosis, and treatment of coronavirus disease 2019 Thachil, J. Coagulopathy of coronavirus disease 2019.
(COVID-19): a review. JAMA 324, 782–793 (2020). Crit. Care Med. 48, 1358–1364 (2020).
Francesco Dentali8, Fabrizio Montecucco   9,10,
12. Berlin, D. A., Gulick, R. M. & Martinez, F. J. Severe 37. Lodigiani, C. et al. Venous and arterial
Steffen Massberg   11,12, Marcel Levi13 and COVID-19. N. Engl. J. Med. 383, 2451–2460 (2020). thromboembolic complications in COVID-19 patients
Antonio Abbate1 13. Lavezzo, E. et al. Suppression of a SARS-CoV-2 admitted to an academic hospital in Milan, Italy.
outbreak in the Italian municipality of Vo′. Nature Thromb. Res. 191, 9–14 (2020).
1
Pauley Heart Center, Division of Cardiology,
584, 425–429 (2020). 38. Iba, T., Connors, J. M. & Levy, J. H. The coagulopathy,
Department of Internal Medicine, Virginia 14. Siddiqi, H. K. & Mehra, M. R. COVID-19 illness in endotheliopathy, and vasculitis of COVID-19. Inflamm.
Commonwealth University, Richmond, VA, USA. native and immunosuppressed states: a clinical- Res. 69, 1181–1189 (2020).
therapeutic staging proposal. J. Heart Lung Transpl. 39. Gazzaruso, C. et al. Association between antithrombin
2
First Clinic of Internal Medicine, Department of
39, 405–407 (2020). and mortality in patients with COVID-19. A possible
Internal Medicine, University of Genoa, Genoa, Italy. 15. Buckley, L. F. et al. Role for anti-cytokine therapies in link with obesity. Nutr. Metab. Cardiovasc. Dis. 30,
3
Department of Internal Medicine, ASST dei Sette severe coronavirus disease 2019. Crit. Care Explor. 2, 1914–1919 (2020).
e0178 (2020). 40. Mir, N., D'Amico, A., Dasher, J., Tolwani, A. &
Laghi, Varese, Italy. 16. Huang, C. et al. Clinical features of patients infected Valentine, V. Understanding the andromeda
4
Unit of Immunology, Rheumatology, Allergy and Rare with 2019 novel coronavirus in Wuhan, China. Lancet strain — the role of cytokine release, coagulopathy
Diseases, IRCCS San Raffaele Scientific Institute, Milan, 395, 497–506 (2020). and antithrombin III in SARS-CoV2 critical illness.
17. Vecchie, A. et al. IL-18 and infections: is there Blood Rev. 45, 100731 (2021).
Italy. a role for targeted therapies? J. Cell Physiol. 236, 41. Wang, Y. et al. SARS-CoV-2 infection of the liver
Vita-Salute San Raffaele University, Milan, Italy.
5 1638–1657 (2021). directly contributes to hepatic impairment in
18. Fajgenbaum, D. C. & June, C. H. Cytokine storm. patients with COVID-19. J. Hepatol. 73, 807–816
6
Center for Molecular and Vascular Biology, N. Engl. J. Med. 383, 2255–2273 (2020). (2020).
Department of Cardiovascular Sciences, KU Leuven, 19. Mehta, P. et al. COVID-19: consider cytokine storm 42. Portincasa, P., Krawczyk, M., Machill, A., Lammert, F.
Leuven, Belgium. syndromes and immunosuppression. Lancet 395, & Di Ciaula, A. Hepatic consequences of COVID-19
1033–1034 (2020). infection. Lapping or biting? Eur. J. Intern. Med. 77,
7
Department of Pharmacotherapy and Outcome 20. Kox, M., Waalders, N. J. B., Kooistra, E. J., Gerretsen, J. 18–24 (2020).
Science, School of Pharmacy, Virginia Commonwealth & Pickkers, P. Cytokine levels in critically ill patients 43. Whyte, C. S., Morrow, G. B., Mitchell, J. L.,
University, Richmond, VA, USA. with COVID-19 and other conditions. JAMA 324, Chowdary, P. & Mutch, N. J. Fibrinolytic abnormalities
1565–1567 (2020). in acute respiratory distress syndrome (ARDS) and
8
Department of Medicine and Surgery, Insubria 21. Leisman, D. E. et al. Cytokine elevation in severe and versatility of thrombolytic drugs to treat COVID-19.
University, Varese, Italy. critical COVID-19: a rapid systematic review, J. Thromb. Haemost. 18, 1548–1555 (2020).
meta-analysis, and comparison with other 44. Gralinski, L. E. et al. Mechanisms of severe acute
9
IRCCS Ospedale Policlinico San Martino Genova — inflammatory syndromes. Lancet Respir. Med. 8, respiratory syndrome coronavirus-induced acute lung
Italian Cardiovascular Network, Genoa, Italy. 1233–1244 (2020). injury. mBio 4, e00271-13 (2013).

NATuRE REVIEWS | Immunology

0123456789();:
Perspectives

45. Iba, T., Warkentin, T. E., Thachil, J., Levi, M. & Levy, J. H. 72. Levi, M. Platelets in critical illness. Semin. Thromb. study with clinical correlation. Virchows Arch. 477,
Proposal of the definition for COVID-19-associated Hemost. 42, 252–257 (2016). 349–357 (2020).
coagulopathy. J. Clin. Med. 10, 191 (2021). 73. Zaid, Y. et al. Platelets can associate with SARS-Cov-2 98. Wolfel, R. et al. Virological assessment of hospitalized
46. Tian, S. et al. Pulmonary pathology of early-phase RNA and are hyperactivated in COVID-19. Circ. Res. patients with COVID-2019. Nature 581, 465–469
2019 novel coronavirus (COVID-19) pneumonia in 127, 1404–1418 (2020). (2020).
two patients with lung cancer. J. Thorac. Oncol. 15, 74. Chrysanthopoulou, A. et al. Interferon λ1/IL-29 and 99. Bradley, B. T. et al. Histopathology and ultrastructural
700–704 (2020). inorganic polyphosphate are novel regulators of findings of fatal COVID-19 infections in Washington
47. Varga, Z. et al. Endothelial cell infection and neutrophil-driven thromboinflammation. J. Pathol. State: a case series. Lancet 396, 320–332 (2020).
endotheliitis in COVID-19. Lancet 395, 1417–1418 243, 111–122 (2017). 100. Manne, B. K. et al. Platelet gene expression and
(2020). 75. Gu, S. X. et al. Thrombocytopathy and function in patients with COVID-19. Blood 136,
48. Schaller, T. et al. Postmortem examination of patients endotheliopathy: crucial contributors to COVID-19 1317–1329 (2020).
with COVID-19. JAMA 323, 2518–2520 (2020). thromboinflammation. Nat. Rev. Cardiol. 18, 101. Zhang, S. et al. SARS-CoV-2 binds platelet ACE2 to
49. Nicholls, J. M. et al. Lung pathology of fatal severe 194–209 (2021). enhance thrombosis in COVID-19. J. Hematol. Oncol.
acute respiratory syndrome. Lancet 361, 1773–1778 76. Gattinoni, L., Chiumello, D. & Rossi, S. COVID-19 13, 120 (2020).
(2003). pneumonia: ARDS or not? Crit. Care 24, 154 (2020). 102. Yan, S. F., Mackman, N., Kisiel, W., Stern, D. M. &
50. McGonagle, D., Sharif, K., O’Regan, A. & 77. Gattinoni, L. et al. COVID-19 does not lead to Pinsky, D. J. Hypoxia/hypoxemia-induced activation
Bridgewood, C. The role of cytokines including a ‘typical’ acute respiratory distress syndrome. of the procoagulant pathways and the pathogenesis of
interleukin-6 in COVID-19 induced pneumonia and Am. J. Respir. Crit. Care Med. 201, 1299–1300 ischemia-associated thrombosis. Arterioscler. Thromb.
macrophage activation syndrome-like disease. (2020). Vasc. Biol. 19, 2029–2035 (1999).
Autoimmun. Rev. 19, 102537 (2020). 78. Gattinoni, L. et al. COVID-19 pneumonia: different 103. Bikdeli, B. et al. COVID-19 and thrombotic or
51. Chen, L. & Hao, G. The role of angiotensin-converting respiratory treatments for different phenotypes? thromboembolic disease: implications for prevention,
enzyme 2 in coronaviruses/influenza viruses and Intensive Care Med. 46, 1099–1102 (2020). antithrombotic therapy, and follow-up: JACC
cardiovascular disease. Cardiovasc. Res. 116, 79. Ciceri, F. et al. Microvascular COVID-19 lung vessels State-of-the-Art Review. J. Am. Coll. Cardiol. 75,
1932–1936 (2020). obstructive thromboinflammatory syndrome 2950–2973 (2020).
52. Magro, C. et al. Complement associated microvascular (MicroCLOTS): an atypical acute respiratory distress 104. Wang, T. et al. Attention should be paid to venous
injury and thrombosis in the pathogenesis of severe syndrome working hypothesis. Crit. Care Resusc. 22, thromboembolism prophylaxis in the management
COVID-19 infection: a report of five cases. Transl Res. 95–97 (2020). of COVID-19. Lancet Haematol. 7, e362–e363
220, 1–13 (2020). 80. Middleton, E. A. et al. Neutrophil extracellular traps (2020).
53. Zhang, H. et al. Histopathologic changes and contribute to immunothrombosis in COVID-19 105. Paranjpe, I. et al. Association of treatment dose
SARS-CoV-2 immunostaining in the lung of a patient acute respiratory distress syndrome. Blood 136, anticoagulation with in-hospital survival among
with COVID-19. Ann. Intern. Med. 172, 629–632 1169–1179 (2020). hospitalized patients with COVID-19. J. Am. Coll.
(2020). 81. Mitsios, A., Arampatzioglou, A., Arelaki, S., Cardiol. 76, 122–124 (2020).
54. Xu, Z. et al. Pathological findings of COVID-19 Mitroulis, I. & Ritis, K. NETopathies? Unraveling 106. Barnes, G. D. et al. Thromboembolism and
associated with acute respiratory distress syndrome. the dark side of old diseases through neutrophils. anticoagulant therapy during the COVID-19 pandemic:
Lancet Respir. Med. 8, 420–422 (2020). Front. Immunol. 7, 678 (2016). interim clinical guidance from the anticoagulation
55. Teuwen, L. A., Geldhof, V., Pasut, A. & Carmeliet, P. 82. Rebetz, J., Semple, J. W. & Kapur, R. The pathogenic forum. J. Thromb. Thrombolysis 50, 72–81 (2020).
COVID-19: the vasculature unleashed. Nat. Rev. involvement of neutrophils in acute respiratory 107. Levi, M. & Hunt, B. J. Thrombosis and coagulopathy in
Immunol. 20, 389–391 (2020). distress syndrome and transfusion-related acute COVID-19: an illustrated review. Res. Pract. Thromb.
56. Goeijenbier, M. et al. Review: Viral infections and lung injury. Transfus. Med. Hemother 45, 290–298 Haemost. 4, 744–751 (2020).
mechanisms of thrombosis and bleeding. J. Med. (2018). 108. Paar, V. et al. Anti-coagulation for COVID-19 treatment:
Virol. 84, 1680–1696 (2012). 83. Bendib, I. et al. Neutrophil extracellular traps are both anti-thrombotic and anti-inflammatory? J. Thromb.
57. Guo, Y., Korteweg, C., McNutt, M. A. & Gu, J. elevated in patients with pneumonia-related acute Thrombolysis 51, 226–231 (2021).
Pathogenetic mechanisms of severe acute respiratory respiratory distress syndrome. Anesthesiology 130, 109. Prandoni, P. et al. The hazard of fondaparinux in
syndrome. Virus Res. 133, 4–12 (2008). 581–591 (2019). non-critically ill patients with COVID-19: retrospective
58. Ding, Y. et al. The clinical pathology of severe acute 84. Lv, X. et al. Extracellular histones are clinically controlled study versus enoxaparin. Thromb. Res.
respiratory syndrome (SARS): a report from China. relevant mediators in the pathogenesis of acute 196, 395–397 (2020).
J. Pathol. 200, 282–289 (2003). respiratory distress syndrome. Respir. Res. 18, 165 110. Russo, V. et al. Fondaparinux use in patients with
59. Hamming, I. et al. Tissue distribution of ACE2 protein, (2017). COVID-19: a preliminary multicenter real-world
the functional receptor for SARS coronavirus. A first 85. Zuo, Y. et al. Neutrophil extracellular traps in experience. J. Cardiovasc. Pharmacol. 76, 369–371
step in understanding SARS pathogenesis. J. Pathol. COVID-19. JCI Insight 5, e138999 (2020). (2020).
203, 631–637 (2004). 86. Skendros, P. et al. Complement and tissue 111. Rossi, R., Coppi, F., Talarico, M. & Boriani, G. Protective
60. Levi, M. & van der Poll, T. Coagulation and sepsis. factor-enriched neutrophil extracellular traps are role of chronic treatment with direct oral anticoagulants
Thromb. Res. 149, 38–44 (2017). key drivers in COVID-19 immunothrombosis. J. Clin. in elderly patients affected by interstitial pneumonia in
61. Helms, J. et al. High risk of thrombosis in patients Invest. 130, 6151–6157 (2020). COVID-19 era. Eur. J. Intern. Med. 77, 158–160
with severe SARS-CoV-2 infection: a multicenter 87. Leppkes, M. et al. Vascular occlusion by neutrophil (2020).
prospective cohort study. Intensive Care Med. 46, extracellular traps in COVID-19. EBioMedicine 58, 112. Flam, B., Wintzell, V., Ludvigsson, J. F., Martensson, J.
1089–1098 (2020). 102925 (2020). & Pasternak, B. Direct oral anticoagulant use and risk
62. Escher, R., Breakey, N. & Lammle, B. Severe COVID-19 88. Radermecker, C. et al. Neutrophil extracellular traps of severe COVID-19. J. Intern. Med. 289, 411–419
infection associated with endothelial activation. infiltrate the lung airway, interstitial, and vascular (2021).
Thromb. Res. 190, 62 (2020). compartments in severe COVID-19. J. Exp. Med. 217, 113. Larsson, M. et al. A factor XIIa inhibitory antibody
63. Martinelli, N. et al. A relative ADAMTS13 deficiency e20201012 (2020). provides thromboprotection in extracorporeal
supports the presence of a secondary microangiopathy 89. Veras, F. P. et al. SARS-CoV-2-triggered neutrophil circulation without increasing bleeding risk. Sci. Transl
in COVID 19. Thromb. Res. 193, 170–172 (2020). extracellular traps mediate COVID-19 pathology. Med. 6, 222ra217 (2014).
64. Schwameis, M., Schorgenhofer, C., Assinger, A., J. Exp. Med. 217, e20201129 (2020). 114. Jansen, P. M. et al. Inhibition of factor XII in septic
Steiner, M. M. & Jilma, B. VWF excess and 90. O’Flynn, J., Dixon, K. O., Faber Krol, M. C., baboons attenuates the activation of complement
ADAMTS13 deficiency: a unifying pathomechanism Daha, M. R. & van Kooten, C. Myeloperoxidase directs and fibrinolytic systems and reduces the release of
linking inflammation to thrombosis in DIC, malaria, properdin-mediated complement activation. J. Innate interleukin-6 and neutrophil elastase. Blood 87,
and TTP. Thromb. Haemost. 113, 708–718 (2015). Immun. 6, 417–425 (2014). 2337–2344 (1996).
65. Guervilly, C. et al. Circulating endothelial cells as 91. de Bont, C. M., Boelens, W. C. & Pruijn, G. J. M. 115. Asakura, H. & Ogawa, H. Potential of heparin and
a marker of endothelial injury in severe COVID-19. NETosis, complement, and coagulation: a triangular nafamostat combination therapy for COVID-19.
J. Infect. Dis. 222, 1789–1793 (2020). relationship. Cell Mol. Immunol. 16, 19–27 (2019). J. Thromb. Haemost. 18, 1521–1522 (2020).
66. Falcinelli, E. et al. Role of endothelial dysfunction in 92. Yalavarthi, S. et al. Release of neutrophil extracellular 116. Yamamoto, M. et al. Identification of nafamostat as a
the thrombotic complications of COVID-19 patients. traps by neutrophils stimulated with antiphospholipid potent inhibitor of Middle East respiratory syndrome
J. Infect. https://doi.org/10.1016/j.jinf.2020.11.041 antibodies: a newly identified mechanism of coronavirus S protein-mediated membrane fusion using
(2020). thrombosis in the antiphospholipid syndrome. the split-protein-based cell–cell fusion assay. Antimicrob.
67. Marini, J. J. & Gattinoni, L. Management of COVID-19 Arthritis Rheumatol. 67, 2990–3003 (2015). Agents Chemother. 60, 6532–6539 (2016).
respiratory distress. JAMA 323, 2329–2330 (2020). 93. Mendoza-Pinto, C., Garcia-Carrasco, M. & 117. Bonaventura, A., Montecucco, F. & Dallegri, F.
68. Perico, L. et al. Immunity, endothelial injury and Cervera, R. Role of infectious diseases in the Update on the effects of treatment with recombinant
complement-induced coagulopathy in COVID-19. antiphospholipid syndrome (including its catastrophic tissue-type plasminogen activator (rt-PA) in
Nat. Rev. Nephrol. 17, 46–64 (2021). variant). Curr. Rheumatol. Rep. 20, 62 (2018). acute ischemic stroke. Expert. Opin. Biol. Ther. 16,
69. Bonetti, P. O., Lerman, L. O. & Lerman, A. Endothelial 94. Zuo, Y. et al. Prothrombotic autoantibodies in serum 1323–1340 (2016).
dysfunction: a marker of atherosclerotic risk. from patients hospitalized with COVID-19. Sci. Transl 118. Liu, C. et al. Meta-analysis of preclinical studies of
Arterioscler. Thromb. Vasc. Biol. 23, 168–175 Med. 12, eabd3876 (2020). fibrinolytic therapy for acute lung injury. Front.
(2003). 95. Channappanavar, R. & Perlman, S. Pathogenic Immunol. 9, 1898 (2018).
70. Hottz, E. D. et al. Platelet activation and platelet– human coronavirus infections: causes and 119. Wang, J. et al. Tissue plasminogen activator (tPA)
monocyte aggregate formation trigger tissue factor consequences of cytokine storm and immunopathology. treatment for COVID-19 associated acute respiratory
expression in patients with severe COVID-19. Blood Semin. Immunopathol. 39, 529–539 (2017). distress syndrome (ARDS): a case series. J. Thromb.
136, 1330–1341 (2020). 96. Chousterman, B. G., Swirski, F. K. & Weber, G. F. Haemost. 18, 1752–1755 (2020).
71. Nicolai, L. et al. Immunothrombotic dysregulation in Cytokine storm and sepsis disease pathogenesis. 120. Kanthi, Y., Knight, J. S., Zuo, Y. & Pinsky, D. J. New (re)
COVID-19 pneumonia is associated with respiratory Semin. Immunopathol. 39, 517–528 (2017). purpose for an old drug: purinergic modulation may
failure and coagulopathy. Circulation 142, 97. Bosmuller, H. et al. The evolution of pulmonary extinguish the COVID-19 thromboinflammatory
1176–1189 (2020). pathology in fatal COVID-19 disease: an autopsy firestorm. JCI Insight 5, e140971 (2020).

www.nature.com/nri

0123456789();:
Perspectives

121. Fata-Hartley, C. L. & Palmenberg, A. C. Dipyridamole 147. Diurno, F. et al. Eculizumab treatment in patients kinase pathway. J. Immunol. 182, 7997–8004
reversibly inhibits mengovirus RNA replication. with COVID-19: preliminary results from real life ASL (2009).
J. Virol. 79, 11062–11070 (2005). Napoli 2 Nord experience. Eur. Rev. Med. Pharmacol. 171. Bergmann, S. & Hammerschmidt, S. Fibrinolysis
122. Huang, B. et al. Mucosal profiling of pediatric-onset Sci. 24, 4040–4047 (2020). and host response in bacterial infections. Thromb.
colitis and IBD reveals common pathogenics and 148. Annane, D. et al. Eculizumab as an emergency Haemost. 98, 512–520 (2007).
therapeutic pathways. Cell 179, 1160–1176.e24 treatment for adult patients with severe COVID-19 172. Buchanan, J. T. et al. DNase expression allows the
(2019). in the intensive care unit: a proof-of-concept study. pathogen group A Streptococcus to escape killing
123. Insel, P. A. et al. cAMP and Epac in the regulation EClinicalMedicine 28, 100590 (2020). in neutrophil extracellular traps. Curr. Biol. 16,
of tissue fibrosis. Br. J. Pharmacol. 166, 447–456 149. Mastaglio, S. et al. The first case of COVID-19 396–400 (2006).
(2012). treated with the complement C3 inhibitor AMY-101. 173. Beiter, K. et al. An endonuclease allows Streptococcus
124. Ali, R. A. et al. Adenosine receptor agonism protects Clin. Immunol. 215, 108450 (2020). pneumoniae to escape from neutrophil extracellular
against NETosis and thrombosis in antiphospholipid 150. Mastellos, D. C. et al. Complement C3 vs C5 inhibition traps. Curr. Biol. 16, 401–407 (2006).
syndrome. Nat. Commun. 10, 1916 (2019). in severe COVID-19: early clinical findings reveal 174. Cheng, A. G. et al. Contribution of coagulases towards
125. Liu, X. et al. Potential therapeutic effects of differential biological efficacy. Clin. Immunol. 220, Staphylococcus aureus disease and protective
dipyridamole in the severely ill patients with COVID-19. 108598 (2020). immunity. PLoS Pathog. 6, e1001036 (2010).
Acta Pharm. Sin. B 10, 1205–1215 (2020). 151. McGonagle, D., Bridgewood, C., Ramanan, A. V., 175. Guggenberger, C., Wolz, C., Morrissey, J. A. &
126. Mitsios, A. et al. Ticagrelor exerts immune-modulatory Meaney, J. F. M. & Watad, A. COVID-19 vasculitis Heesemann, J. Two distinct coagulase-dependent
effect by attenuating neutrophil extracellular traps. and novel vasculitis mimics. Lancet Rheumatol. 3, barriers protect Staphylococcus aureus from
Int. J. Mol. Sci. 21, 3625 (2020). e224–e233 (2021). neutrophils in a three dimensional in vitro infection
127. Stebbing, J. et al. JAK inhibition reduces SARS-CoV-2 152. Tang, N. et al. Anticoagulant treatment is associated model. PLoS Pathog. 8, e1002434 (2012).
liver infectivity and modulates inflammatory responses with decreased mortality in severe coronavirus disease 176. Gavriilaki, E. & Brodsky, R. A. Complementopathies
to reduce morbidity and mortality. Sci. Adv. 7, 2019 patients with coagulopathy. J. Thromb. Haemost. and precision medicine. J. Clin. Invest. 130,
eabe4724 (2020). 18, 1094–1099 (2020). 2152–2163 (2020).
128. Cavalli, G. et al. Repurposing of biologic and targeted 153. Ayerbe, L., Risco, C. & Ayis, S. The association 177. Gralinski, L. E. et al. Complement activation
synthetic anti-rheumatic drugs in COVID-19 and between treatment with heparin and survival in contributes to severe acute respiratory syndrome
hyper-inflammation: a comprehensive review of patients with COVID-19. J. Thromb. Thrombolysis 50, coronavirus pathogenesis. mBio 9, e01753-18
available and emerging evidence at the peak of the 298–301 (2020). (2018).
pandemic. Front. Pharmacol. 11, 598308 (2020). 154. Nadkarni, G. N. et al. Anticoagulation, bleeding, 178. Markiewski, M. M., Nilsson, B., Ekdahl, K. N.,
129. Barnes, B. J. et al. Targeting potential drivers of mortality, and pathology in hospitalized patients Mollnes, T. E. & Lambris, J. D. Complement and
COVID-19: neutrophil extracellular traps. J. Exp. Med. with COVID-19. J. Am. Coll. Cardiol. 76, 1815–1826 coagulation: strangers or partners in crime? Trends
217, e20200652 (2020). (2020). Immunol. 28, 184–192 (2007).
130. Mohamed, M. M. A., El-Shimy, I. A. & Hadi, M. A. 155. La Rosee, F. et al. The Janus kinase 1/2 inhibitor 179. Ramlall, V. et al. Immune complement and coagulation
Neutrophil elastase inhibitors: a potential prophylactic ruxolitinib in COVID-19 with severe systemic dysfunction in adverse outcomes of SARS-CoV-2
treatment option for SARS-CoV-2-induced respiratory hyperinflammation. Leukemia 34, 1805–1815 infection. Nat. Med. 26, 1609–1615 (2020).
complications? Crit. Care 24, 311 (2020). (2020). 180. Peerschke, E. I., Reid, K. B. & Ghebrehiwet, B.
131. Zeiher, B. G. et al. Neutrophil elastase inhibition 156. Cao, Y. et al. Ruxolitinib in treatment of severe Platelet activation by C1q results in the induction of
in acute lung injury: results of the STRIVE study. coronavirus disease 2019 (COVID-19): a multicenter, αIIb/β3 integrins (GPIIb-IIIa) and the expression
Crit. Care Med. 32, 1695–1702 (2004). single-blind, randomized controlled trial. J. Allergy of P-selectin and procoagulant activity. J. Exp. Med.
132. Aikawa, N. & Kawasaki, Y. Clinical utility of Clin. Immunol. 146, 137–146.e3 (2020). 178, 579–587 (1993).
the neutrophil elastase inhibitor sivelestat for the 157. Hickey, M. J. & Kubes, P. Intravascular immunity: the 181. Ikeda, K. et al. C5a induces tissue factor activity on
treatment of acute respiratory distress syndrome. host–pathogen encounter in blood vessels. Nat. Rev. endothelial cells. Thromb. Haemost. 77, 394–398
Ther. Clin. Risk Manag. 10, 621–629 (2014). Immunol. 9, 364–375 (2009). (1997).
133. Tagami, T. et al. Effect of a selective neutrophil 158. Jackson, S. P., Darbousset, R. & Schoenwaelder, S. M. 182. Ritis, K. et al. A novel C5a receptor-tissue factor
elastase inhibitor on mortality and ventilator-free days Thromboinflammation: challenges of therapeutically cross-talk in neutrophils links innate immunity to
in patients with increased extravascular lung water: targeting coagulation and other host defense coagulation pathways. J. Immunol. 177, 4794–4802
a post hoc analysis of the PiCCO Pulmonary Edema mechanisms. Blood 133, 906–918 (2019). (2006).
Study. J. Intensive Care 2, 67 (2014). 159. Massberg, S. et al. Reciprocal coupling of coagulation
134. Papayannopoulos, V., Staab, D. & Zychlinsky, A. and innate immunity via neutrophil serine proteases. Acknowledgements
Neutrophil elastase enhances sputum solubilization Nat. Med. 16, 887–896 (2010). F.M. is supported by a grant from the Italian Ministry of
in cystic fibrosis patients receiving DNase therapy. 160. von Bruhl, M. L. et al. Monocytes, neutrophils, Health to the Italian Cardiovascular Network (#2754291).
PLoS ONE 6, e28526 (2011). and platelets cooperate to initiate and propagate
135. Zou, Y. et al. Neutrophil extracellular traps promote venous thrombosis in mice in vivo. J. Exp. Med. 209, Author contributions
lipopolysaccharide-induced airway inflammation 819–835 (2012). A.B., A.V., K.M., S.M., M.L. and A.A. contributed to all
and mucus hypersecretion in mice. Oncotarget 9, 161. Kambas, K. et al. Autophagy mediates the delivery aspects of the article. L.D., D.L.D., B.W.V.T., F.D. and F.M.
13276–13286 (2018). of thrombogenic tissue factor to neutrophil contributed to discussion of the content and reviewed and/or
136. Lefrancais, E., Mallavia, B., Zhuo, H., Calfee, C. S. extracellular traps in human sepsis. PLoS ONE 7, edited the manuscript before submission. All authors
& Looney, M. R. Maladaptive role of neutrophil e45427 (2012). approved the final version of the manuscript.
extracellular traps in pathogen-induced lung injury. 162. Bonaventura, A. et al. The pathophysiological
JCI Insight 3, e98178 (2018). role of neutrophil extracellular traps in inflammatory Competing interests
137. Thomas, G. M. et al. Extracellular DNA traps are diseases. Thromb. Haemost. 118, 6–27 (2018). A.B. and A.V. received a travel grant from Kiniksa
associated with the pathogenesis of TRALI in humans 163. Martinod, K. & Wagner, D. D. Thrombosis: tangled up Pharmaceuticals Ltd and honoraria from Effetti s.r.l. (Milan,
and mice. Blood 119, 6335–6343 (2012). in NETs. Blood 123, 2768–2776 (2014). Italy). L.D. has received consultation honoraria from Abbvie,
138. Okur, H. K. et al. Preliminary report of in vitro and 164. Ammollo, C. T., Semeraro, F., Xu, J., Esmon, N. L. & A m g e n , B i o g e n , B r i s to l - M ye rs S q u i b b, C e l l t r i o n ,
in vivo effectiveness of dornase alfa on SARS-CoV-2 Esmon, C. T. Extracellular histones increase plasma GlaxoSmithKline, Novartis, Pfizer, Roche, Sanofi-Genzyme
infection. N. Microbes N. Infect. 37, 100756 (2020). thrombin generation by impairing thrombomodulin- and SOBI. K.M. is an inventor on patent US9642822 and
139. Weber, A. G., Chau, A. S., Egeblad, M., Barnes, B. J. dependent protein C activation. J. Thromb. Haemost. p a t e n t a p p l i c a t i o n s W O 2 01 8 0 2 71 9 5 3 A 1 a n d
& Janowitz, T. Nebulized in-line endotracheal dornase 9, 1795–1803 (2011). US2019167680A1, and is a consultant for PEEL
alfa and albuterol administered to mechanically 165. Nicolai, L., Gaertner, F. & Massberg, S. Therapeutics, Inc. B.W.V.T. has served as a consultant for
ventilated COVID-19 patients: a case series. Platelets in host defense: experimental and Novartis and Serpin Pharma, and has received research sup-
Mol. Med. 26, 91 (2020). clinical insights. Trends Immunol. 40, 922–938 port from Novartis, Kiniksa Pharmaceuticals Ltd, Serpin
140. Gollomp, K. et al. Neutrophil accumulation and NET (2019). Pharma and SOBI. A.A. has served as a consultant for
release contribute to thrombosis in HIT. JCI Insight 3, 166. Ma, A. C. & Kubes, P. Platelets, neutrophils, and Applied Clinical Intel, AstraZeneca, Cromos Pharma, Janssen,
e99445 (2018). neutrophil extracellular traps (NETs) in sepsis. Kiniksa Pharmaceuticals Ltd, Merck, Olatec and Serpin
141. Fuchs, T. A. et al. Extracellular DNA traps J. Thromb. Haemost. 6, 415–420 (2008). Pharma, and has received research support from Janssen,
promote thrombosis. Proc. Natl Acad. Sci. USA 107, 167. Guo, L. & Rondina, M. T. The era of Olatec and Serpin Pharma. All other authors declare no
15880–15885 (2010). thromboinflammation: platelets are dynamic competing interests.
142. Wolach, O. et al. Increased neutrophil extracellular sensors and effector cells during infectious
trap formation promotes thrombosis in diseases. Front. Immunol. 10, 2204 (2019). Peer review information
myeloproliferative neoplasms. Sci. Transl Med. 10, 168. Reinhardt, C. et al. Protein disulfide isomerase acts Nature Reviews Immunology thanks the anonymous reviewers
eaan8292 (2018). as an injury response signal that enhances fibrin for their contribution to the peer review of this work.
143. Yost, C. C. et al. Neonatal NET-inhibitory factor and generation via tissue factor activation. J. Clin. Invest.
related peptides inhibit neutrophil extracellular trap 118, 1110–1122 (2008). Publisher’s note
formation. J. Clin. Invest. 126, 3783–3798 (2016). 169. Semple, J. W., Aslam, R., Kim, M., Speck, E. R. & Springer Nature remains neutral with regard to jurisdictional
144. Java, A. et al. The complement system in COVID-19: Freedman, J. Platelet-bound lipopolysaccharide claims in published maps and institutional affiliations.
friend and foe? JCI Insight 5, e140711 (2020). enhances Fc receptor-mediated phagocytosis of
145. Zhang, Y. et al. Coagulopathy and antiphospholipid IgG-opsonized platelets. Blood 109, 4803–4805 Supplementary information
antibodies in patients with COVID-19. N. Engl. J. Med. (2007). The online version contains supplementary material available
382, e38 (2020). 170. Zhang, G. et al. Lipopolysaccharide stimulates at https://doi.org/10.1038/s41577-021-00536-9.
146. Risitano, A. M. et al. Complement as a target in platelet secretion and potentiates platelet aggregation
COVID-19? Nat. Rev. Immunol. 20, 343–344 (2020). via TLR4/MyD88 and the cGMP-dependent protein © Springer Nature Limited 2021

NATuRE REVIEWS | Immunology

0123456789();:

You might also like