ArticlePDF AvailableLiterature Review

Non-stenotic Carotid Plaques in Embolic Stroke of Unknown Source

Frontiers
Frontiers in Neurology
Authors:
  • Wolfson Centre for Prevention of Stroke and Dementia - University of Oxford
  • Central Zone AHS

Abstract and Figures

Embolic stroke of unknown source (ESUS) represents one in five ischemic strokes. Ipsilateral non-stenotic carotid plaques are identified in 40% of all ESUS. In this narrative review, we summarize the evidence supporting the potential causal relationship between ESUS and non-stenotic carotid plaques; discuss the remaining challenges in establishing the causal link between non-stenotic plaques and ESUS and describe biomarkers of potential interest for future research. In support of the causal relationship between ESUS and non-stenotic carotid plaques, studies have shown that plaques with high-risk features are five times more prevalent in the ipsilateral vs. the contralateral carotid and there is a lower incidence of atrial fibrillation during follow-up in patients with ipsilateral non-stenotic carotid plaques. However, non-stenotic carotid plaques with or without high-risk features often coexist with other potential etiologies of stroke, notably atrial fibrillation (8.5%), intracranial atherosclerosis (8.4%), patent foramen ovale (5–9%), and atrial cardiopathy (2.4%). Such puzzling clinical associations make it challenging to confirm the causal link between non-stenotic plaques and ESUS. There are several ongoing studies exploring whether select protein and RNA biomarkers of plaque progression or vulnerability could facilitate the reclassification of some ESUS as large vessel strokes or help to optimize secondary prevention strategies.
This content is subject to copyright.
MINI REVIEW
published: 22 September 2021
doi: 10.3389/fneur.2021.719329
Frontiers in Neurology | www.frontiersin.org 1September 2021 | Volume 12 | Article 719329
Edited by:
Christopher Bladin,
Monash University, Australia
Reviewed by:
Klearchos Psychogios,
Metropolitan Hospital, Greece
Patricia Martínez Sánchez,
Torrecárdenas University
Hospital, Spain
Jae Won Song,
University of Pennsylvania,
United States
*Correspondence:
Joseph Kamtchum-Tatuene
kamtchum@ualberta.ca
Specialty section:
This article was submitted to
Stroke,
a section of the journal
Frontiers in Neurology
Received: 02 June 2021
Accepted: 30 August 2021
Published: 22 September 2021
Citation:
Kamtchum-Tatuene J, Nomani AZ,
Falcione S, Munsterman D, Sykes G,
Joy T, Spronk E, Vargas MI and
Jickling GC (2021) Non-stenotic
Carotid Plaques in Embolic Stroke of
Unknown Source.
Front. Neurol. 12:719329.
doi: 10.3389/fneur.2021.719329
Non-stenotic Carotid Plaques in
Embolic Stroke of Unknown Source
Joseph Kamtchum-Tatuene 1
*, Ali Z. Nomani 2, Sarina Falcione 2, Danielle Munsterman 2,
Gina Sykes 2, Twinkle Joy 2, Elena Spronk 2, Maria Isabel Vargas 3and Glen C. Jickling 2
1Faculty of Medicine and Dentistry, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada,
2Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada, 3Division of
Neuroradiology, Department of Radiology and Medical Imaging, Geneva University Hospital, Geneva, Switzerland
Embolic stroke of unknown source (ESUS) represents one in five ischemic strokes.
Ipsilateral non-stenotic carotid plaques are identified in 40% of all ESUS. In this narrative
review, we summarize the evidence supporting the potential causal relationship between
ESUS and non-stenotic carotid plaques; discuss the remaining challenges in establishing
the causal link between non-stenotic plaques and ESUS and describe biomarkers of
potential interest for future research. In support of the causal relationship between
ESUS and non-stenotic carotid plaques, studies have shown that plaques with high-risk
features are five times more prevalent in the ipsilateral vs. the contralateral carotid and
there is a lower incidence of atrial fibrillation during follow-up in patients with ipsilateral
non-stenotic carotid plaques. However, non-stenotic carotid plaques with or without
high-risk features often coexist with other potential etiologies of stroke, notably atrial
fibrillation (8.5%), intracranial atherosclerosis (8.4%), patent foramen ovale (5–9%), and
atrial cardiopathy (2.4%). Such puzzling clinical associations make it challenging to
confirm the causal link between non-stenotic plaques and ESUS. There are several
ongoing studies exploring whether select protein and RNA biomarkers of plaque
progression or vulnerability could facilitate the reclassification of some ESUS as large
vessel strokes or help to optimize secondary prevention strategies.
Keywords: stroke, carotid stenosis, carotid plaque, biomarkers, atherosclerosis
INTRODUCTION
Ischemic stroke is considered cryptogenic when no definite cause is identified during the baseline
etiological workup (1). According to the Cryptogenic Stroke/Embolic Stroke of Undetermined
Source International Working Group, the baseline etiological workup should include brain
imaging with computed tomography (CT) or magnetic resonance imaging (MRI), assessment
of the heart rhythm with 12-lead ECG and continuous cardiac monitoring for at least 24 h
with automated rhythm detection, transthoracic cardiac ultrasound, and imaging of cervical and
intracranial vessels supplying the infarcted brain region (using CT, MRI, conventional angiography,
or ultrasonography) (2).
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
Cryptogenic strokes represent 30% of all ischemic strokes.
They could be further classified into three subgroups: stroke with
no cause despite complete baseline workup, stroke with multiple
possible underlying causes, and stroke with incomplete baseline
workup (3). In the subgroup of cryptogenic strokes with complete
workup, embolic stroke of unknown source (ESUS) is a clinical
construct referring to non-lacunar ischemic strokes (size >1.5 cm
on CT or >2.0 cm on diffusion MRI) of presumable embolic
origin (superficial/cortical brain lesion) despite the absence of
any obvious sources of cardiac or arterial embolism (e.g., atrial
fibrillation, carotid, or intracranial stenosis >50%) (Figure 1)
(2). ESUS represent 17% of all ischemic strokes with a recurrent
stroke rate of 4.5% per year despite antithrombotic therapy (46).
The definition of ESUS was based on the assumptions that
cryptogenic strokes may be related to covert atrial fibrillation
and that a relationship between non-stenotic atherosclerotic
plaques (causing <50% stenosis) and stroke was unlikely.
However, there is now evidence to suggest that ESUS represents
a heterogeneous group including patients with various other
potential causes of stroke besides atrial fibrillation (79). Such
causes include atrial cardiopathy (10), patent foramen ovale
(PFO) (11), cancer (12), and non-stenotic plaques affecting
the aortic arch or carotid, vertebral, or intracranial arteries
(7,13,14). Atrial cardiopathy is a concept referring to
a dysfunction of the left atrium that is thought to favor
and precede the onset of atrial fibrillation and its eventual
detection by electrocardiographic devices. The diagnosis is
based on the identification of imaging markers (e.g., left atrial
enlargement, spontaneous echocontrast in the left atrium or
the left atrial appendage, atrial fibrosis with delayed gadolinium
enhancement on MRI), electrocardiographic markers (e.g.,
paroxysmal supraventricular tachycardia, increased P-wave
terminal force in V1, interatrial block, prolonged PR), and
blood biomarkers (e.g., N-terminal pro-brain natriuretic peptide,
highly sensitive cardiac troponin T) (10).
Non-stenotic carotid plaques are found in 40% of patients
with ESUS and 10–15% of patients with ESUS have mild stenosis
(20–49%) (2,1517). Here we review the evidence supporting
the relationship between non-stenotic carotid plaques with high-
risk features and stroke in patients with ESUS. We present the
remaining challenges in the process of formally establishing the
causal link between non-stenotic plaques and ESUS, notably
those related to the identification of blood biomarkers of
vulnerable plaque. Finally, we discuss the management of non-
stenotic carotid plaques in patients with ESUS and highlight areas
for future research.
NON-STENOTIC CAROTID PLAQUES AS A
POTENTIAL CAUSE OF ESUS
The relationship between non-stenotic carotid plaques and ESUS
is supported by a set of three clinical observations.
First, in patients with ESUS, carotid plaques are more
prevalent on the side of the stroke than on the contralateral
side. In a cross-sectional study of 85 patients with ESUS,
non-stenotic carotid plaques thicker than 3 mm were
present in 35% of ipsilateral carotid arteries vs. 15% of
the contralateral carotid arteries (18). A similar finding
was observed in a review of 138 ESUS cases from the
prospective multicenter INTERRSeCT study (The Predicting
Early Recanalization and Reperfusion With IV Alteplase
and Other Treatments Using Serial CT Angiography). The
investigators found a non-stenotic carotid plaque ipsilateral to
the stroke in 29.2% of patients and contralateral to the stroke
in 18.7% (17).
Second, in patients with ESUS, there is a lower incidence
of atrial fibrillation detected during follow-up in patients with
ipsilateral non-stenotic carotid plaques than in those without,
thus suggesting that non-stenotic carotid plaques may be
related to the stroke. In 777 participants of the New Approach
Rivaroxaban Inhibition of Factor Xa in a Global Trial vs.
ASA to Prevent Embolism in Embolic Stroke of Undetermined
Source (NAVIGATE-ESUS) trial who were followed up for a
median of 2 years, the incidence of atrial fibrillation was 2.9
per 100 person-years in patients with ipsilateral non-stenotic
carotid plaques vs. 5.0 per 100 person-years in those without
(overall rate: 8.5 vs. 19.0%; adjusted hazard ratio: 0.57, 95%
CI 0.37–0.84) (15).
Third, plaques with high-risk features are more prevalent
on the side of the stroke in patients with ESUS. In a
meta-analysis of 8 studies enrolling 323 patients with ESUS,
plaques with high-risk features were present in 32.5% of
the ipsilateral carotid arteries vs. 4.6% of the contralateral
carotid arteries. More specifically, the odds of finding a
non-stenotic carotid plaque with a ruptured fibrous cap in
the ipsilateral vs. the contralateral carotid artery was 17.5,
reinforcing the idea that non-stenotic carotid plaques should
not be considered as benign coincidental findings in patients
with ESUS (13).
High-risk plaques have features on brain or vascular imaging
that are associated with a higher risk of stroke in patients with
either symptomatic or asymptomatic carotid atherosclerosis,
independent of the grade of stenosis (1924). The most
common high-risk plaque features are echolucency, impaired
cerebrovascular reserve, intraplaque hemorrhage (Figure 1),
silent brain infarcts, lipid-rich necrotic core, large juxtaluminal
black hypoechoic area, large plaque volume, plaque thickness,
microembolic signals, mural thrombus, neovascularization,
plaque irregularity, plaque inflammation or hypermetabolism,
thin or ruptured fibrous cap, and ulceration (19,21,25
31). The American Heart Association combines some of these
features to derive a classification of atherosclerotic plaques into
6 types reflecting increasing instability and risk of cardiovascular
events (Table 1) (3237). On average, high-risk plaque features
are three times more prevalent in patients with symptomatic
vs. asymptomatic carotid stenosis (OR =3.4, 95% CI: 2.5–
4.6) (19). They are detected using various vascular imaging
modalities (Table 2). To date, there are no data on the risk
of recurrent stroke associated with each of the high-risk
features in patients with ESUS. Analysis of secondary outcome
data from the Carotid Plaque Imaging in Acute Stroke study
(CAPIAS; NCT01284933) might help to address this knowledge
gap (35,39).
Frontiers in Neurology | www.frontiersin.org 2September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
FIGURE 1 | Brain and plaque imaging findings in a 64-year-old man with ESUS. (A) Axial angio-CT scan slice showing a hypodense non-stenotic carotid plaque in the
right internal carotid artery (white arrow). (B–E) Axial diffusion-weighted imaging slices (with corresponding ADC maps) showing multiple embolic strokes in the right
pre-and post-central area. (F,G) Coronal and axial T1-weighted black blood sequence showing hyperintensity of the non-stenotic plaque in the right internal carotid
artery (white arrow), thus confirming the presence of intraplaque hemorrhage.
CHALLENGES OF ESTABLISHING CAUSAL
LINK WITH STROKE
Puzzling Clinical Associations
Although studies of high-risk features have provided evidence of
an association between non-stenotic carotid plaques and brain
infarction in patients with ESUS, establishing causality remains
challenging in most cases. The dilemma rests on four clinical
observations. First, high-risk features are often found in plaques
in the absence of related clinical symptoms (19,40). In a meta-
analysis of eight studies enrolling 323 patients with ESUS, a non-
stenotic carotid plaque with high-risk features was identified in
the contralateral carotid artery in 4.6% of cases (95% CI: 0.1–
13.1) (13). Likewise, in a meta-analysis of 64 studies enrolling
20,571 patients with asymptomatic carotid stenosis of various
grades, 26.5% of patients were found to have at least one high-
risk plaque feature (95% CI: 22.9–30.3). The highest prevalence
was observed for neovascularization (43.4%, 95% CI: 31.4–55.8)
and the lowest for mural thrombus (7.3%, 95% CI: 2.5–19.4).
On average, intraplaque hemorrhage was found in 1 out of 5
patients (19). Second, high-risk plaque features are not specific
for symptomatic carotid plaques. In a meta-analysis of data from
20 prospective studies enrolling 1,652 patients with symptomatic
carotid stenosis, high-risk plaque features were identified in <1
in 2 patients (43.3%, 95% CI: 33.6–53.2) (19). Third, in patients
with stroke, there is an association between the presence of high-
risk plaque features and atrial fibrillation. In a study of 68 patients
with embolic stroke, including 45 ESUS, the presence of high-
risk plaque features on carotid ultrasound (ulceration, thickness
3 mm, and echolucency) was independently associated with
detection of atrial fibrillation on admission or during follow-
up (OR =4.5, 95% CI: 1.0–19.6) (41). Fourth, in some patients
with ESUS diagnosed using the current clinical definition, non-
stenotic carotid plaques often coexist with other potential causes
of stroke, including atrial fibrillation (8.5%) (15), intracranial
atherosclerosis (8.4%) (42), PFO (5–9%) (43,44), and atrial
cardiopathy (2.4%) (45).
Lack of Reliable Biomarkers
The identification of an ipsilateral non-stenotic carotid plaque
with or without high-risk features is not sufficient to reclassify
ESUS as stroke due to large vessel disease. Further research is,
Frontiers in Neurology | www.frontiersin.org 3September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
TABLE 1 | American Heart Association comprehensive morphological classification scheme for atherosclerotic lesions (3234).
Plaque type Description
Lipid rich
necrotic core
Fibrous cap Calcification Erosion/rupture Intraplaque
hemorrhage
Thrombus Regression to
normal
Type I (Initial lesion) Initial lesion, accumulation of
smooth muscle cells and isolated
foam cells, absence of a necrotic
core.
Absent Absent Absent Absent Absent Absent Possible
Type II (Intimal
xanthoma)
Multiple layers of foam cells,
previously referred to as “fatty
streak”
Absent Absent Absent Absent Absent Absent Possible
Type III
(pre-atheroma)
Smooth muscle cells in a
proteoglycan-rich extracellular
matrix, multiple layers of foam
cells, non-confluent extracellular
lipid pools
Absent Present (ill-defined) Absent Absent Absent Absent Possible
Type IV (atheroma) Confluent extracellular lipids Present
(well-formed)
Present
(well-defined)
Absent Absent Absent Absent Not possible
Type Va
(Fibroatheroma)
Confluent extracellular lipids with
prominent proliferative
fibromuscular layer
Present
(well-formed)
Present (thick) PossibleaAbsent Absent Absent Not possible
Type VI
(Complicated
atheroma)b
Inflammatory lesion with at least
one high-risk feature
Present (large) Present (thin or
eroded)
Possible (partial
calcification)
Possible (VIa if
present alone)
Possible (VIb if
present alone)
Possible (VIc if
present alone)
Not possible
aThe plaque is assigned category Vb if predominantly calcified (fibro-calcific) or category Vc if predominantly fibrous (collagen-rich atheroma with smaller lipid core).
bThe plaque is assigned category VIabc if erosion/ulceration, intraplaque hemorrhage and luminal thrombus are present concurrently.
Frontiers in Neurology | www.frontiersin.org 4September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
TABLE 2 | High-risk plaque features commonly used in clinical practice (13,21,2531).
High-risk plaque
featuresa
Imaging
modality of
choice
DescriptionbAlternative imaging
modalities
Prevalence (%)in patients with ESUS
AHA type IV, V, VI
(3537)
MRI Plaque with large lipid-rich necrotic core
(>40% of the vessel circumference), ruptured
fibrous cap, mural thrombus, or intraplaque
hemorrhage (see below).
CT, US In three studies including 82 patients with
ESUS, an AHA plaque type IV-VI was
found in the ipsilateral carotid in 38% of
cases on average (3537).
Echolucency US Hypoechoic area within the plaque on B-mode
(reference =sternocleidomastoid muscle)
Not applicable In a study of 44 patients with ESUS, an
ipsilateral echolucent non-stenotic carotid
plaque was found in 50.0% (38)
Impaired
cerebrovascular
reserve
TCD <10% increase of blood flow in the ipsilateral
MCA while breathing 5% CO2for 2 min.
BOLD-MRI Not applicable for non-stenotic plaques
Intraplaque
hemorrhage
MRI Intraplaque hyperintensity on T1W FAT SAT
(black blood) and 3D-TOF
MRI In five studies including 162 patients,
intraplaque hemorrhage was found in the
ipsilateral carotid in 24.4% of cases (13).
Ipsilateral silent brain
infarcts
MRI Non-lacunar hyperintensity of the brain
parenchyma, in the territory of the internal
carotid artery, visible on T2W and FLAIR, or
DWI (if acute)
CT (would appear as a
hypodensity)
No data available for patients with ESUS
Lipid-rich necrotic core MRI Collection of foam cells, cholesterol crystals
and apoptotic cells that appears
iso/hyper-intense on T1W and iso/hypo-intense
on T2W.
CT, US (although it is
difficult to make the
difference with
intraplaque
hemorrhage on these
modalities)
No data available for patients with ESUS
Microembolic signals TCD Random audible transient increase (variable
threshold) of the Doppler signal within the
monitored arterial blood flow, generating a
high-intensity signal on the doppler imaging
(PWV and M-Mode), visible and moving in the
direction of the flow. Duration of recording
1 h.c
Not applicable No data available for patients with ESUS
Mural thrombus MRI Filling defect on contrast MRI, hyperintense
signal adjacent to the lumen on T1W
CT, US In three studies enrolling 94 patients with
ESUS, plaque thrombus was identified in
the ipsilateral carotid in 6.9% of cases (13).
Neovascularization CEUS Enhancement of the plaque on pulse inversion
harmonic imaging (microbubbles carried into
the plaque by the blood entering the
neovessels)
DCE-MRI No data available for patients with ESUS
Plaque irregularity MRI 0.3–0.9 mm fluctuations of the surface of the
plaque
CT, CEUS No data available for patients with ESUS
Thin/ruptured fibrous
cap
MRI Disrupted or invisible dark band adjacent to the
lumen on 3D-TOF
CEUS In two studies enrolling 50 patients with
ESUS, a thin or ruptured fibrous cap was
found in the ipsilateral carotid in 23.6% of
cases (13).
Ulceration MRI Depression >1 mm on the surface of the
plaque
CTA, CEUS (the
threshold is 2 mm in
ultrasound studies)
No data available for patients with ESUS
aThe following high-risk features are used less often: juxta-luminal black hypoechoic area and plaque volume assessed by ultrasound, plaque inflammation measured by standardized
(18) F-FDG uptake on positron emission tomography-computed tomography, carotid temperature assessed by microwave radiometry.
bFor simplicity, the description of each high-risk feature is based on its appearance on the imaging modality of choice.
cThe sound threshold and the number of MES for a positive examination is variable across studies.
AHA, American Heart Association; BOLD, blood oxygen level-dependent; CEUS, contrast-enhanced ultrasound; CI, confidence interval; CT, computed tomography; DCE, dynamic
contrast-enhanced; ESUS, embolic stroke of undetermined source; FLAIR, fluid-attenuated inversion recovery; MCA, middle cerebral artery; MRI, Magnetic Resonance Imaging; T1W,
T1-weighted imaging; T2W, T2-weighted imaging; TCD, transcranial Doppler ultrasound; and 3D-TOF, 3-dimensional time of flight.
therefore, needed to determine whether combination of vascular
imaging findings, clinical data, and candidate biomarkers of
plaque progression/instability or atheroembolism (4682) into
multiparameter scores could improve the ability to (1) establish a
causal link between ESUS and a non-stenotic carotid plaque, (2)
predict plaque progression or stroke recurrence, and (3) select
Frontiers in Neurology | www.frontiersin.org 5September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
patients who might benefit from adjuvant anti-inflammatory
and lipid-lowering therapies as briefly discussed in the next
section. Some biomarkers of plaque progression and instability
that warrant further investigation specifically in patients with
ESUS are presented in Table 3. There are several ongoing projects
exploring biomarkers in patients with ESUS or cryptogenic
stroke, notably the Searching for Explanations for Cryptogenic
Stroke in the Young: Revealing the Etiology, Triggers, and
Outcome study (SECRETO, NCT01934725) (95), the Carotid
Plaque Imaging in Acute Stroke study (CAPIAS, NCT01284933)
(35), and the Biomarkers of Acute Stroke Etiology study
(BASE, NCT02014896) (96). Efforts to establish a causal
relationship between non-stenotic carotid stenosis and ESUS
using biomarkers and multimodal vascular imaging in well-
phenotyped prospective cohorts will also benefit from research
aiming to identify alternative causes of stroke in patients with
ESUS (14,68,97104).
CHALLENGES OF SECONDARY STROKE
PREVENTION
As a result of the challenges to determine the root cause of an
ESUS, the optimal treatment strategy for patients with ESUS
remains unclear, and a tailored approach would likely be the
most appropriate (9). In this section, we briefly describe the
strategies that have been explored so far and discuss possible
future directions.
Dual Antiplatelet Therapy and Antiplatelet
Switch
Following the results of the Platelet-Oriented Inhibition in
New TIA and Minor Ischemic Stroke (POINT) (105) and the
Clopidogrel in High-Risk Patients with Acute Non-disabling
Cerebrovascular Events (CHANCE) (106) trials, patients with
ESUS are treated with Aspirin-based dual antiplatelet therapy
for 21 days provided that their baseline NIHSS is low. After 3
weeks, patients ideally return to single antiplatelet therapy and
switching from Aspirin to Clopidogrel is considered in patients
who had an ESUS while on Aspirin (107). A meta-analysis
of data from CHANCE and POINT showed that extending
the treatment beyond 3 weeks might increase the bleeding
risk without additional benefit for secondary stroke prevention
(108). Whether the presence of ipsilateral non-stenotic carotid
plaque with or without high-risk features would modify the
magnitude (absolute risk reduction) and duration (beyond 21
days) of the benefits derived from dual antiplatelet therapy
in patients with ESUS remains unknown. In patients allergic
to Clopidogrel and in carriers of a CYP2C19 loss of function
allele, Ticagrelor might be an alternative according to findings
of the Acute Stroke or Transient Ischemic Attack Treated with
Ticagrelor and ASA [acetylsalicylic acid] for Prevention of Stroke
and Death (THALES) trial (109112). The ongoing Clopidogrel
with Aspirin in High-risk patients with Acute Non-disabling
Cerebrovascular Events II (CHANCE-2, NCT04078737) trial is
evaluating the superiority of the Ticagrelor-Aspirin combination
over Clopidogrel-Aspirin therapy in CYP2C19 loss of function
carriers with minor stroke or transient ischemic attack (TIA)
(113). There is currently no evidence supporting the use of
dual antiplatelet therapies not containing Aspirin or triple
antiplatelet therapies (with or without Aspirin) for secondary
stroke prevention in patients with acute stroke or TIA (114).
Anticoagulation
The New Approach Rivaroxaban Inhibition of Factor Xa in a
Global Trial vs. ASA [Acetylsalicylic Acid] to Prevent Embolism
in Embolic Stroke of Undetermined Source (NAVIGATE-ESUS)
and the Randomized Double-Blind Evaluation in Secondary
Stroke Prevention Comparing The Efficacy Of Oral Thrombin
Inhibitor Dabigatran Etexilate for Secondary Stroke Prevention
in Patients With Embolic Stroke of Undetermined Source (RE-
SPECT-ESUS) trials have shown that universal full-dose oral
anticoagulation is not an effective strategy to reduce the risk of
stroke recurrence in patients with ESUS (5,6). These results are
likely explained by the heterogeneity of stroke mechanisms in
patients with ESUS as discussed earlier, with atrial fibrillation
being diagnosed in only 24.8% of cases at 24 months using
insertable cardiac monitors (115). Moreover, there is no evidence
that patients with ESUS and ipsilateral non-stenotic carotid
plaques should be treated differently than those without plaques.
In a subgroup analysis of data from 2,905 patients with non-
stenotic carotid plaques enrolled in the NAVIGATE-ESUS trial,
there was no difference between Rivaroxaban and Aspirin
with respect to the prevention of ipsilateral ischemic stroke
[Hazard ratio [HR] =0.6, 95% CI: 0.2–1.9]. Major bleeding
complications were significantly more frequent in patients taking
anticoagulation (HR =3.7, 95% CI: 1.6–8.7) (16).
In the Cardiovascular Outcomes for People Using
Anticoagulation Strategies (COMPASS) trial, the combination
Rivaroxaban-Aspirin (2.5 mg twice daily plus Aspirin 100 mg
once per day) was superior to Aspirin alone (100 mg once daily)
for the prevention of cardioembolic strokes (HR =0.4, 95% CI:
0.2–0.8) and ESUS (HR =0.3, 95% CI: 0.1–0.7) but there was
no effect on the incidence of stroke due to moderate-to-severe
carotid stenosis (HR =0.9, 95% CI: 0.5–1.6) (116). Although
these results suggest that the combination of Aspirin and
low-dose Rivaroxaban could be an effective secondary stroke
prevention strategy, they are not directly applicable to patients
with ESUS since all patients with acute stroke (<1 month)
were excluded from the trial due to the perceived higher risk
of major intracranial bleeding (117). Furthermore, the baseline
proportion of patients with non-stenotic carotid plaque, with or
without high-risk features, was not reported. The prevalence of
ipsilateral non-stenotic carotid plaque in participants diagnosed
with ESUS during follow-up was also not reported.
According to currently available data, patients with
ESUS and features of atrial cardiopathy, notably atrial
enlargement, constitute the only subgroup that may benefit
from anticoagulation (118). However, since these results are
derived from a post-hoc analysis of the NAVIGATE-ESUS trial,
they might not be used to justify universal prescription of
anticoagulation until confirmation is obtained in dedicated
trials. The ongoing Atrial Cardiopathy and Antithrombotic
Drugs in Prevention After Cryptogenic Stroke (ARCADIA,
Frontiers in Neurology | www.frontiersin.org 6September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
TABLE 3 | Biomarkers of potential interest for the study of non-stenotic carotid plaques in ESUS.
Biomarker Type Main source Key evidence Specific target
of a drug
previously tested
in human trials
References
Lectin-like
oxidized LDL
receptor 1 (LOX-1)
Protein Endothelial cells,
smooth muscle
cells, fibroblasts
In 4,703 participants from the Malmo Diet and Cancer Cohort, higher plasma levels of soluble LOX-1
were associated with higher risk of stroke during a mean follow-up of 16.5 years (HR =1.5, 95% CI:
1.3–2.4).
In 202 patients undergoing carotid endarterectomy, plasma levels of soluble LOX-1 were correlated with
the plaque content of oxidized LDL, proinflammatory cytokines, and matrix metalloproteinases.
No (4649,59,75)
Omentin-1 Protein Visceral adipose
tissue, stromal
vascular cells,
lung, heart,
placenta, ovaries
In 173 patients with acute ischemic stroke, serum levels of omentin-1 were lower in subjects with
unstable plaque (n=38, echolucent, thin fibrous cap, ulcerated) than in those with stable plaques
(median of 53 vs. 62 ng/mL).
No (69)
Lipoprotein-
associated
phospholipase A2
(Lp-PLA2)
Protein Monocytes,
macrophages, T
lymphocytes, and
mast cells
In 1,946 participants of the Northern Manhattan study, there was a dose-response relationship between
Lp-PLA2 mass and the risk of first-ever stroke due to large vessel atherosclerosis (HR =1.4, 4.5, and
5.1 for quartiles 2, 3, and 4 compared with quartile 1 in multivariable survival analysis).
Yes (Darapladib) (52,53,83)
Chitinase-3-like-1
(YKL-40)
Protein Inflammatory cells In 1,132 patients with carotid atherosclerotic plaques of various grades, higher levels of YKL-40 were
associated with plaque instability (n=855, echolucency) after adjusting for various demographic and
cardiovascular risk factors (OR =2.1 and 1.7 for quartiles 3 and 4, respectively).
No (56,59)
Granzyme B Protein T lymphocytes In 67 patients with severe carotid stenosis undergoing revascularization, higher plasma levels of
granzyme B were found in patients with unstable plaques (n=16, echolucent) than in those with stable
plaques (median of 492.0 vs. 143.8 pg/mL)
No (57)
Vimentin Protein Endothelial cells,
macrophages, and
astrocytes
In 4,514 patients with carotid plaques in the Malmo Diet and Cancer Cohort, higher plasma levels of
vimentin at baseline were associated with the incidence of ischemic stroke after a mean follow-up of 22
years (HR =1.66, 95% CI: 1.23–2.25).
Yes (Withaferin-A) (65,84)
Macrophage
chemoattractant
protein
(MCP-1/CCL2)
Protein Monocytes In the Athero-EXPRESS biobank, higher plaque levels of MCP-1 levels were found in symptomatic (vs.
asymptomatic) plaques and in vulnerable (vs. stable) plaques.
No (61)
Matrix
metalloproteinase
9 (MMP9)
Protein Macrophages,
foam cells
Serum levels of MMP9 were higher in large artery atherosclerosis strokes (n=26, 1,137 ng/mL) vs.
cardioembolic strokes (n=86, 517 ng/mL). MMP9 >1,110 ng/mL had 85% sensitivity and 52%
specificity for differentiating large vessel from cardioembolic strokes.
No (59,66)
Complement 5b-9 Protein Liver In 70 patients with acute ischemic stroke, serum C5b-9 levels were higher in patients with unstable
plaques (n=37) than in those with stable plaques (median of 875 vs. 786 ng/mL). There was also a
positive correlation with plaque burden and grade of stenosis.
Yes (Eculizumab) (76,85)
Interleukin 1β
(IL-1β)
Protein Monocytes,
macrophages
A higher expression of IL-1βand other components of the NLRP3 inflammasome was observed in 30
plaques when compared with 10 healthy mesenteric arteries, both at the protein and the mRNA level.
Yes (Anakinra,
Rilonacept,
Canakinumab)
(77,8688)
Interleukin 6 (IL-6) Protein Monocytes,
macrophages
In a sub-analysis of data from 703 participants of the population-based Tromsø study, higher plasma
levels of IL-6 were independently associated with plaque progression after a 6-year follow-up (OR 1.4,
95% CI 1.1–1.8 per 1 SD increase in IL-6 level).
Yes (Ziltivekimab,
Tocilizumab)
(7174)
(Continued)
Frontiers in Neurology | www.frontiersin.org 7September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
TABLE 3 | Continued
Biomarker Type Main source Key evidence Specific target
of a drug
previously tested
in human trials
References
C-Reactive Protein
(CRP)
Protein Hepatocytes,
white blood cells,
adipocytes,
smooth muscle
cells
In a prospective observational study enrolling 271 participants, higher levels of CRP (quartile 4 vs. 1)
were associated with plaque progression after a follow-up of 37 months (OR =1.8, 95% CI: 1.03–2.99).
No (78,89)
CD36 Protein Various cells
including
monocytes,
endothelial cells,
adipocytes,
platelets.
In 62 patients with severe carotid stenosis undergoing revascularization, plasma levels of soluble CD36
were higher in those with symptomatic (n=31) and unstable (echolucent, n=20) plaques.
No (60)
Lipoprotein (a) Lipoprotein Food/Liver In 876 consecutive patients with carotid atherosclerosis (2.5% occlusions), plasma lipoprotein (a) was an
independent predictor of carotid occlusion (OR=1.7, 95% CI: 1.2–2.3 per 1 SD increase), suggesting
that it plays a role in plaque destabilization/rupture, thrombosis, and impaired fibrinolysis.
In 225 patients with coronary artery disease who underwent intra-coronary optical coherence
tomography imaging of culprit plaque, the prevalence of thin fibrous cap atheroma was significantly
higher in the group with higher serum lipoprotein (a) levels (>25 mg/dL, n=87): 23 vs. 11%.
Yes (AKCEA-
Apo(a)-LRx)
(7981,90,91)
Non-HDL
cholesterol
Lipoproteins Food/Liver In 2,888 patients with carotid plaque, including 1,505 with vulnerable plaques (echolucent, irregular, or
ulcerated), higher serum levels of non-HDL cholesterol were independently associated with plaque
vulnerability (OR =1.5 for tertile 3 vs. 1, 95% CI: 1.2–1.8).
Yes (various class
of lipid lowering
drugs)
(51,92,93)
Uric acid Xanthine (purine
derivatives)
Various cells In a study including 88 patients with carotid plaques (44 symptomatic), serum uric acid levels were
significantly higher in patients with symptomatic plaques (7.4 vs. 5.4 mg/dL) who also had higher plaque
expression of xanthine oxidase as assessed by immunohistochemistry.
Yes (allopurinol) (82)
Neutrophil count Cells NA In 60 patients with recently symptomatic carotid artery disease, higher neutrophil count (>5,900/µL) was
associated with detection of microembolic signals on transcranial Doppler monitoring.
No (58)
miR-199b-3p,
miR-27b-3p,
miR-130a-3p,
miR- 221-3p, and
miR-24-3p
RNA Various cells In 60 patients with moderate or severe asymptomatic carotid stenosis, higher plasma levels of the
micro-RNAs were associated with plaque progression (n=19) after 2 years of follow-up.
No (62)
miR-200c RNA Various cells In 22 patients undergoing carotid endarterectomy, higher levels of miR-200c were found in patients with
unstable plaques (echolucent symptomatic) and were positively correlated with biomarkers of plaque
instability (matrix metalloproteinase—MMP1, MMP9; interleukin 6, macrophage chemoattractant protein
1—MCP-1)
No (59,94)
Resistin and
chimerin mRNA
RNA Various cells In an analysis of 165 carotid plaque (67% unstable based on histological criteria), Resistin and chemerin
mRNA expression was 80 and 32% lower, respectively, in unstable vs. stable plaques.
No (70)
Frontiers in Neurology | www.frontiersin.org 8September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
NCT03192215) (101), Apixaban for Treatment of Embolic
Stroke of Undetermined Source (ATTICUS, NCT02427126),
and A Study on BMS-986177 (oral factor XIa inhibitor) for
the Prevention of a Stroke in Patients Receiving Aspirin and
Clopidogrel (AXIOMATIC-SSP, NCT03766581) trials will,
hopefully, provide conclusive results to guide patient care.
Likewise, in the Oxford Vascular Study, a large patent foramen
ovale is present in 36% of patients with a cryptogenic stroke
aged >60 years (119) and associated with a 2.5 times higher risk
of recurrent ischemic stroke (120), thus suggesting it might be
worth trialing PFO closure or anticoagulation in elderly patients
with a large PFO. However, the causal relationship between
the PFO and the recurrent stroke was not formally established
and the prevalence of ipsilateral non-stenotic carotid plaque
not reported. Because PFO closure or anticoagulation are not
expected to prevent strokes due to large vessel atherosclerosis,
trials of PFO closure or anticoagulation in elderly patients with
a large PFO should carefully plan subgroup analyses according
to the presence of alternative candidate causes of the recurrent
stroke, notably an atrial cardiopathy or an ipsilateral non-stenotic
carotid plaque that may coexist with PFO (43,44,121).
Other Therapies and Interventions
Currently, patients with ESUS receive intensive lipid-lowering
therapy (e.g., statins, ezetimibe) to achieve a level of LDL
cholesterol <70 mg/dL (1.8 mmol/L) as early as possible after
stroke (122124). The treatment is maintained long-term if
well-tolerated, even in older adults (125128). Specific targets
of LDL cholesterol have not been assessed in patients with
ESUS and it is unknown if the presence of an ipsilateral non-
stenotic carotid plaque would modify the effect of lipid-lowering
drugs as suggested by findings of the Stroke Prevention by
Aggressive Reduction in Cholesterol Levels (SPARCL) (129).
Furthermore, the potential role of newer classes of lipid-lowering
drugs for plaque stabilization and secondary stroke prevention
is yet to be defined. Such drugs include proprotein convertase
subtilisin/kexin type 9 (PCSK9) inhibitors (small interfering
RNA—inclisiran or monoclonal antibodies—evolocumab or
alirocumab) and Apo(a) antisense oligonucleotides that reduce
plasma levels of both LDL cholesterol and lipoprotein(a) [Lp(a)];
as well as anti- angiopoietin-like 3 monoclonal antibodies that
do not affect Lp(a) levels and bempedoic acid (92,130135).
Like ezetimibe (93,136), the new lipid-lowering drugs may be
useful as add-on or statin-sparing agents in cases of allergy or
intolerance to statins, familial hypercholesterolemia, refractory
hypercholesterolemia, or in patients with high Lp(a) levels at
the time of stroke since statins increase plasma levels of Lp(a)
(90,137). There are reports of an association between high
Lp(a) levels and cryptogenic stroke (138,139) suggesting that
Lp(a) could represent a biomarker to guide optimization of lipid-
lowering therapy in patients with ESUS as is the case in other
cardiovascular diseases.
Systemic inflammation, a hallmark of atherosclerosis,
modulates the risk of stroke and the effect of lipid-lowering
agents (140142). This explains the benefit of various anti-
inflammatory drugs (e.g., canakinumab, colchicine) for
the prevention of atherosclerotic cardiovascular diseases
(86,87,143). In patients with ESUS and ipsilateral non-stenotic
carotid plaque, the effect of anti-inflammatory agents is worth
exploring, especially in those with high-risk plaque features
since they would not be offered revascularization procedures as
first-line treatment according to current guidelines (144146).
Data from the ongoing Colchicine for Prevention of Vascular
Inflammation in Non-Cardioembolic Stroke (CONVINCE,
NCT02898610) might answer the question of whether patients
with ESUS with or without ipsilateral non-stenotic carotid
plaques would benefit from the addition of low-dose colchicine
to best medical therapy for secondary stroke prevention (147).
The relevance of serial vascular imaging to monitor carotid
plaque progression and stability is another aspect of the
management that remains unexplored.
Besides pharmacological treatments, there is a variety of
lifestyle interventions that are beneficial for cardiovascular
risk reduction and are recommended by the American Heart
Association for secondary stroke prevention no matter the
suspected underlying etiology. Such interventions include
smoking cessation, regular physical activity, weight loss,
improved sleep hygiene, avoidance of noise and air pollution,
reduction of salt and sugar intake, higher consumption of fish,
fruits, and vegetables (148155).
CONCLUSION
ESUS is a common subtype of stroke that is frequently associated
with an ipsilateral non-stenotic carotid plaque. Evidence suggests
that advanced multimodal vascular imaging and biomarkers
might help reclassify some ESUS as large vessel strokes. However,
the precise algorithm for this reclassification remains to be
designed. Despite significant research efforts since the term
ESUS was coined in 2014, the optimal management strategy
for patients with ESUS remains unclear. There are several
ongoing trials investigating various interventions. While waiting
for more evidence to support the design of tailored therapeutic
guidelines for the various well-phenotyped subgroups of patients
with ESUS, clinicians should continue to fully implement
all previously validated stroke prevention strategies, whether
an ipsilateral non-stenotic carotid plaque is present or not.
Such strategies include short-term dual antiplatelet therapy if
appropriate, long-term intensive lipid lowering therapy, control
of modifiable cardiovascular risk factors (e.g., hypertension,
diabetes, smoking, obesity), and lifestyle changes.
AUTHOR CONTRIBUTIONS
JK-T did the literature search and wrote the manuscript. MV
and JK-T prepared the figure. AN, SF, DM, GS, TJ, ES, MV, and
GJ critically revised the manuscript. All authors approved the
final version.
FUNDING
GJ received research grant support from Canadian Institutes
of Health Research (CIHR), Heart and Stroke Foundation,
Frontiers in Neurology | www.frontiersin.org 9September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
University Hospital Foundation, Canada Foundation for
Innovation (CFI), and National Institutes of Health (NIH).
JK-T was supported by the Faculty of Medicine and Dentistry
Motyl Graduate Studentship in Cardiac Sciences, an Alberta
Innovates Graduate Student Scholarship, the Ballermann
Translational Research Fellowship, the Izaak Walton Killam
Memorial Scholarship, and the Andrew Stewart Memorial
Graduate Prize.
REFERENCES
1. Adams HP Jr, Bendixen BH, Kappelle LJ, Biller J, Love BB, Gordon
DL, et al. Classification of subtype of acute ischemic stroke. Definitions
for use in a multicenter clinical trial. TOAST. Trial of Org 10172 in
Acute Stroke Treatment. Stroke. (1993) 24:35–41. doi: 10.1161/01.STR.
24.1.35
2. Hart RG, Diener HC, Coutts SB, Easton JD, Granger CB,
O’Donnell MJ, et al. Embolic strokes of undetermined source:
the case for a new clinical construct. Lancet Neurol. (2014)
13:429–38. doi: 10.1016/S1474-4422(13)70310-7
3. Tsivgoulis G, Katsanos AH, Kohrmann M, Caso V, Lemmens R,
Tsioufis K, et al. Embolic strokes of undetermined source: theoretical
construct or useful clinical tool? Ther Adv Neurol Disord. (2019)
12:1756286419851381. doi: 10.1177/1756286419851381
4. Hart RG, Catanese L, Perera KS, Ntaios G, Connolly SJ. Embolic stroke of
undetermined source: a systematic review and clinical update. Stroke. (2017)
48:867–72. doi: 10.1161/STROKEAHA.116.016414
5. Hart RG, Sharma M, Mundl H, Kasner SE, Bangdiwala SI, Berkowitz SD,et al.
Rivaroxaban for stroke prevention after embolic stroke of undetermined
source. N Engl J Med. (2018) 378:2191–201. doi: 10.1056/NEJMoa1802686
6. Diener HC, Sacco RL, Easton JD, Granger CB, Bernstein RA,
Uchiyama S, et al. Dabigatran for prevention of stroke after
embolic stroke of undetermined source. N Engl J Med. (2019)
380:1906–17. doi: 10.1056/NEJMoa1813959
7. Ntaios G. Embolic stroke of undetermined source: JACC review topic of the
week. J Am Coll Cardiol. (2020) 75:333–40. doi: 10.1016/j.jacc.2019.11.024
8. Li L, Yiin GS, Geraghty OC, Schulz UG, Kuker W, Mehta Z, et al. Incidence,
outcome, risk factors, and long-term prognosis of cryptogenic transient
ischaemic attack and ischaemic stroke: a population-based study. Lancet
Neurol. (2015) 14:903–13. doi: 10.1016/S1474-4422(15)00132-5
9. Kamel H, Merkler AE, Iadecola C, Gupta A, Navi BB. Tailoring the approach
to embolic stroke of undetermined source: a review. JAMA Neurol. (2019)
76:855–61. doi: 10.1001/jamaneurol.2019.0591
10. Yaghi S, Kamel H, Elkind MSV. Atrial cardiopathy: a mechanism
of cryptogenic stroke. Expert Rev Cardiovasc Ther. (2017) 15:591–
9. doi: 10.1080/14779072.2017.1355238
11. Kasner SE, Swaminathan B, Lavados P, Sharma M, Muir K, Veltkamp
R, et al. Rivaroxaban or aspirin for patent foramen ovale and
embolic stroke of undetermined source: a prespecified subgroup
analysis from the NAVIGATE ESUS trial. Lancet Neurol. (2018)
17:1053–60. doi: 10.1016/S1474-4422(18)30319-3
12. Navi BB, Kasner SE, Elkind MSV, Cushman M, Bang OY, DeAngelis LM.
Cancer and embolic stroke of undetermined source. Stroke. (2021) 52:1121–
30. doi: 10.1161/STROKEAHA.120.032002
13. Kamtchum-Tatuene J, Wilman A, Saqqur M, Shuaib A, Jickling GC.
carotid plaque with high-risk features in embolic stroke of undetermined
source: systematic review and meta-analysis. Stroke. (2020) 51:311–
4. doi: 10.1161/STROKEAHA.119.027272
14. Tao L, Li XQ, Hou XW, Yang BQ, Xia C, Ntaios G, et al. Intracranial
atherosclerotic plaque as a potential cause of embolic stroke of undetermined
source. J Am Coll Cardiol. (2021) 77:680–91. doi: 10.1016/j.jacc.2020.12.015
15. Ntaios G, Perlepe K, Sirimarco G, Strambo D, Eskandari A,
Karagkiozi E, et al. Carotid plaques and detection of atrial fibrillation
in embolic stroke of undetermined source. Neurology. (2019)
92:e2644–52. doi: 10.1212/WNL.0000000000007611
16. Ntaios G, Swaminathan B, Berkowitz SD, Gagliardi RJ, Lang W, Siegler JE,
et al. Efficacy and safety of rivaroxaban versus aspirin in embolic stroke of
undetermined source and carotid atherosclerosis. Stroke. (2019) 50:2477–
85. doi: 10.1161/STROKEAHA.119.025168
17. Ospel JM, Singh N, Marko M, Almekhlafi M, Dowlatshahi D, Puig J,
et al. Prevalence of ipsilateral nonstenotic carotid plaques on computed
tomography angiography in embolic stroke of undetermined source. Stroke.
(2020) 51:1743–9. doi: 10.1161/STROKEAHA.120.029404
18. Coutinho JM, Derkatch S, Potvin AR, Tomlinson G, Kiehl TR, Silver FL, et al.
Nonstenotic carotid plaque on CT angiography in patients with cryptogenic
stroke. Neurology. (2016) 87:665–72. doi: 10.1212/WNL.0000000000002978
19. Kamtchum-Tatuene J, Noubiap JJ, Wilman AH, Saqqur M, Shuaib A,
Jickling GC. Prevalence of high-risk plaques and risk of stroke in patients
with asymptomatic carotid stenosis: a meta-analysis. JAMA Neurol. (2020)
77:1018–27. doi: 10.1001/jamaneurol.2020.2658
20. Schindler A, Schinner R, Altaf N, Hosseini AA, Simpson RJ, Esposito-Bauer
L, et al. Prediction of stroke risk by detection of hemorrhage in carotid
plaques: meta-analysis of individual patient data. JACC Cardiovasc Imaging.
(2019) 13(2 Pt 1):395–406. doi: 10.1016/j.jcmg.2019.03.028
21. Saba L, Saam T, Jager HR, Yuan C, Hatsukami TS, Saloner D, et al.
Imaging biomarkers of vulnerable carotid plaques for stroke risk prediction
and their potential clinical implications. Lancet Neurol. (2019) 18:559–
72. doi: 10.1016/S1474-4422(19)30035-3
22. Bos D, Arshi B, van den Bouwhuijsen QJA, Ikram MK, Selwaness M,
Vernooij MW, et al. Atherosclerotic carotid plaque composition and
incident stroke and coronary events. J Am Coll Cardiol. (2021) 77:1426–
35. doi: 10.1016/j.jacc.2021.01.038
23. Kelly PJ, Camps-Renom P, Giannotti N, Marti-Fabregas J, McNulty JP,
Baron JC, et al. A risk score including carotid plaque inflammation and
stenosis severity improves identification of recurrent stroke. Stroke. (2020)
51:838–45. doi: 10.1161/STROKEAHA.119.027268
24. Baradaran H, Gupta A. Extracranial vascular disease: carotid
stenosis and plaque imaging. Neuroimaging Clin N Am. (2021)
31:157–66. doi: 10.1016/j.nic.2021.02.002
25. Bayer-Karpinska A, Schindler A, Saam T. Detection of vulnerable plaque in
patients with cryptogenic stroke. Neuroimaging Clin N Am. (2016) 26:97–
110. doi: 10.1016/j.nic.2015.09.008
26. Fabiani I, Palombo C, Caramella D, Nilsson J, De Caterina R. Imaging of the
vulnerable carotid plaque: role of imaging techniques and a research agenda.
Neurology. (2020) 94:922–32. doi: 10.1212/WNL.0000000000009480
27. Paraskevas KI, Veith FJ, Spence JD. How to identify which patients
with asymptomatic carotid stenosis could benefit from endarterectomy or
stenting. Stroke Vasc Neurol. (2018) 3:92–100. doi: 10.1136/svn-2017-000129
28. Ringelstein EB, Droste DW, Babikian VL, Evans DH, Grosset DG, Kaps
M, et al. Consensus on microembolus detection by TCD. International
Consensus Group on Microembolus Detection. Stroke. (1998) 29:725–
9. doi: 10.1161/01.STR.29.3.725
29. Saam T, Ferguson MS, Yarnykh VL, Takaya N, Xu D, Polissar
NL, et al. Quantitative evaluation of carotid plaque composition
by in vivo MRI. Arterioscler Thromb Vasc Biol. (2005) 25:234–
9. doi: 10.1161/01.ATV.0000149867.61851.31
30. Markus HS, Harrison MJ. Estimation of cerebrovascular reactivity using
transcranial Doppler, including the use of breath-holding as the vasodilatory
stimulus. Stroke. (1992) 23:668–73. doi: 10.1161/01.STR.23.5.668
31. Rafailidis V, Li X, Sidhu PS, Partovi S, Staub D. Contrast imaging ultrasound
for the detection and characterization of carotid vulnerable plaque.
Cardiovasc Diagn Ther. (2020) 10:965–81. doi: 10.21037/cdt.2020.01.08
32. Stary HC. Natural history and histological classification of atherosclerotic
lesions: an update. Arterioscler Thromb Vasc Biol. (2000) 20:1177–
8. doi: 10.1161/01.ATV.20.5.1177
33. Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons
from sudden coronary death: a comprehensive morphological classification
scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol. (2000)
20:1262–75. doi: 10.1161/01.ATV.20.5.1262
Frontiers in Neurology | www.frontiersin.org 10 September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
34. Saba L, Brinjikji W, Spence JD, Wintermark M, Castillo M, Borst GJD, et al.
Roadmap consensus on carotid artery plaque imaging and impact on therapy
strategies and guidelines: an international, multispecialty, expert review and
position statement. AJNR Am J Neuroradiol. (2021). doi: 10.3174/ajnr.A7223.
[Epub ahead of print].
35. Bayer-Karpinska A, Schwarz F, Wollenweber FA, Poppert H, Boeckh-
Behrens T, Becker A, et al. The carotid plaque imaging in acute stroke
(CAPIAS) study: protocol and initial baseline data. BMC Neurol. (2013)
13:201. doi: 10.1186/1471-2377-13-201
36. Freilinger TM, Schindler A, Schmidt C, Grimm J, Cyran C, Schwarz
F, et al. Prevalence of nonstenosing, complicated atherosclerotic
plaques in cryptogenic stroke. JACC Cardiovasc Imaging. (2012)
5:397–405. doi: 10.1016/j.jcmg.2012.01.012
37. Hyafil F, Schindler A, Sepp D, Obenhuber T, Bayer-Karpinska A, Boeckh-
Behrens T, et al. High-risk plaque features can be detected in non-stenotic
carotid plaques of patients with ischaemic stroke classified as cryptogenic
using combined (18)F-FDG PET/MR imaging. Eur J Nucl Med Mol Imaging.
(2016) 43:270–9. doi: 10.1007/s00259-015-3201-8
38. Buon R, Guidolin B, Jaffre A, Lafuma M, Barbieux M, Nasr N, et al.
Carotid ultrasound for assessment of nonobstructive carotid atherosclerosis
in young adults with cryptogenic stroke. J Stroke Cerebrovasc Dis. (2018)
27:1212–6. doi: 10.1016/j.jstrokecerebrovasdis.2017.11.043
39. Kopczak A, Schindler A, Bayer-Karpinska A, Koch ML, Sepp D, Zeller J, et al.
Complicated carotid artery plaques as a cause of cryptogenic stroke. J Am
Coll Cardiol. (2020) 76:2212–22. doi: 10.1016/j.jacc.2020.09.532
40. Sun J, Underhill HR, Hippe DS, Xue Y, Yuan C, Hatsukami TS. Sustained
acceleration in carotid atherosclerotic plaque progression with intraplaque
hemorrhage: a long-term time course study. JACC Cardiovasc Imaging.
(2012) 5:798–804. doi: 10.1016/j.jcmg.2012.03.014
41. Grosse GM, Sieweke JT, Biber S, Ziegler NL, Gabriel MM, Schuppner
R, et al. Nonstenotic carotid plaque in embolic stroke of undetermined
source: interplay of arterial and atrial disease. Stroke. (2020) 51:3737–
41. doi: 10.1161/STROKEAHA.120.030537
42. Ameriso SF, Amarenco P, Pearce LA, Perera KS, Ntaios G,
Lang W, et al. Intracranial and systemic atherosclerosis in
the NAVIGATE ESUS trial: recurrent stroke risk and response
to antithrombotic therapy. J Stroke Cerebrovasc Dis. (2020)
29:104936. doi: 10.1016/j.jstrokecerebrovasdis.2020.104936
43. Ntaios G, Sagris D, Strambo D, Perlepe K, Sirimarco G, Georgiopoulos
G, et al. Carotid atherosclerosis and patent foramen ovale in embolic
stroke of undetermined source. J Stroke Cerebrovasc Dis. (2021)
30:105409. doi: 10.1016/j.jstrokecerebrovasdis.2020.105409
44. Jaffre A, Guidolin B, RuidavetsJB, Nasr N, Larrue V. Non-obstructive carotid
atherosclerosis and patent foramen ovale in young adults with cryptogenic
stroke. Eur J Neurol. (2017) 24:663–6. doi: 10.1111/ene.13275
45. Kamel H, Pearce LA, Ntaios G, Gladstone DJ, Perera K, Roine RO,
et al. Atrial cardiopathy and nonstenosing large artery plaque in patients
with embolic stroke of undetermined source. Stroke. (2020) 51:938–
43. doi: 10.1161/STROKEAHA.119.028154
46. Barreto J, Karathanasis SK, Remaley A, Sposito AC. Role of LOX-1 (Lectin-
like oxidized low-density lipoprotein receptor 1) as a cardiovascular risk
predictor: mechanistic insight and potential clinical use. Arterioscler Thromb
Vasc Biol. (2021) 41:153–66. doi: 10.1161/ATVBAHA.120.315421
47. Hofmann A, Brunssen C, Wolk S, Reeps C, Morawietz H. Soluble
LOX-1: a novel biomarker in patients with coronary artery disease,
stroke, and acute aortic dissection? J Am Heart Assoc. (2020)
9:e013803. doi: 10.1161/JAHA.119.013803
48. Markstad H, Edsfeldt A, Yao Mattison I, Bengtsson E, Singh P,
Cavalera M, et al. High levels of soluble lectinlike oxidized low-density
lipoprotein receptor-1 are associated with carotid plaque inflammation
and increased risk of ischemic stroke. J Am Heart Assoc. (2019)
8:e009874. doi: 10.1161/JAHA.118.009874
49. Yokota C, Sawamura T, Watanabe M, Kokubo Y, Fujita Y, Kakino A,
et al. High levels of soluble lectin-like oxidized low-density lipoprotein
receptor-1 in acute stroke: an age- and sex-matched cross-sectional
study. J Atheroscler Thromb. (2016) 23:1222–6. doi: 10.5551/jat.
32466
50. Li XM, Jin PP, Xue J, Chen J, Chen QF, Luan XQ, et al. Role of sLOX-1 in
intracranial artery stenosis and in predicting long-term prognosis of acute
ischemic stroke. Brain Behav. (2018) 8:e00879. doi: 10.1002/brb3.879
51. Wu J, Zhang J, Wang A, Chen S, Wu S, Zhao X. Association
between non-high-density lipoprotein cholesterol levels and asymptomatic
vulnerable carotid atherosclerotic plaques. Eur J Neurol. (2019) 26:1433–
8. doi: 10.1111/ene.13973
52. Katan M, Moon YP, Paik MC, Wolfert RL, Sacco RL, Elkind MS.
Lipoprotein-associated phospholipase A2 is associated with atherosclerotic
stroke risk: the Northern Manhattan Study. PLoS ONE. (2014)
9:e83393. doi: 10.1371/journal.pone.0083393
53. Yang M, Wang A, Li J, Zhao X, Liu L, Meng X, et al. Lp-PLA2 and
dual antiplatelet agents in intracranial arterial stenosis. Neurology. (2019)
94:e181–9. doi: 10.1212/WNL.0000000000008733
54. Kamtchum-Tatuene J, Jickling GC. Blood biomarkers for
stroke diagnosis and management. Neuromolecular Med. (2019)
21:344–68. doi: 10.1007/s12017-019-08530-0
55. Koenig W, Khuseyinova N. Biomarkers of atherosclerotic plaque
instability and rupture. Arterioscler Thromb Vasc Biol. (2007)
27:15–26. doi: 10.1161/01.ATV.0000251503.35795.4f
56. Wang Y, Li B, Jiang Y, Zhang R, Meng X, Zhao X, et al. YKL-40 is associated
with ultrasound-determined carotid atherosclerotic plaque instability. Front
Neurol. (2021) 12:622869. doi: 10.3389/fneur.2021.622869
57. Skjelland M, Michelsen AE, Krohg-Sorensen K, Tennoe B, Dahl A, Bakke
S, et al. Plasma levels of granzyme B are increased in patients with lipid-
rich carotid plaques as determined by echogenicity. Atherosclerosis. (2007)
195:e142–6. doi: 10.1016/j.atherosclerosis.2007.05.001
58. Nasr N, Ruidavets JB, Arnal JF, Sie P, Larrue V. Association
of neutrophil count with microembolization in patients with
symptomatic carotid artery stenosis. Atherosclerosis. (2009)
207:519–23. doi: 10.1016/j.atherosclerosis.2009.05.003
59. Jiao Y, Qin Y, Zhang Z, Zhang H, Liu H, Li C. Early identification of carotid
vulnerable plaque in asymptomatic patients. BMC Cardiovasc Disord. (2020)
20:429. doi: 10.1186/s12872-020-01709-5
60. Handberg A, Skjelland M, Michelsen AE, Sagen EL, Krohg-Sorensen
K, Russell D, et al. Soluble CD36 in plasma is increased in patients
with symptomatic atherosclerotic carotid plaques and is related to
plaque instability. Stroke. (2008) 39:3092–5. doi: 10.1161/STROKEAHA.108.
517128
61. Georgakis MK, van der Laan SW, Asare Y, Mekke JM, Haitjema S,
Schoneveld AH, et al. Monocyte-chemoattractant protein-1 levels in human
atherosclerotic lesions associate with plaque vulnerability. Arterioscler
Thromb Vasc Biol. (2021) 1:2038–48. doi: 10.1161/ATVBAHA.121.316091
62. Dolz S, Gorriz D, Tembl JI, Sanchez D, Fortea G, Parkhutik V,
et al. Circulating microRNAs as novel biomarkers of stenosis
progression in asymptomatic carotid stenosis. Stroke. (2017)
48:10–6. doi: 10.1161/STROKEAHA.116.013650
63. Basic J, Stojkovic S, Assadian A, Rauscher S, Duschek N, Kaun C, et al. The
relevance of vascular endothelial growth factor, hypoxia inducible factor-1
alpha, and clusterin in carotid plaque instability. J Stroke Cerebrovasc Dis.
(2019) 28:1540–5. doi: 10.1016/j.jstrokecerebrovasdis.2019.03.009
64. Ammirati E, Moroni F, Norata GD, Magnoni M, Camici PG. Markers
of inflammation associated with plaque progression and instability
in patients with carotid atherosclerosis. Mediators Inflamm. (2015)
2015:718329. doi: 10.1155/2015/718329
65. Xiao J, Chen L, Melander O, Orho-Melander M, Nilsson J, Borne
Y, et al. Circulating vimentin is associated with future incidence of
stroke in a population-based cohort study. Stroke. (2021) 52:937–
44. doi: 10.1161/STROKEAHA.120.032111
66. Alhazmi H, Bani-Sadr A, Bochaton T, Paccalet A, Da Silva CC, Buisson
M, et al. Large vessel cardioembolic stroke and embolic stroke of
undetermined source share a common profile of matrix metalloproteinase-9
level and susceptibility vessel sign length. Eur J Neurol. (2021) 28:1977–
83. doi: 10.1111/ene.14806
67. Jickling GC, Xu H, Stamova B, Ander BP, Zhan X, Tian Y, et al. Signatures of
cardioembolic and large-vessel ischemic stroke. Ann Neurol. (2010) 68:681–
92. doi: 10.1002/ana.22187
Frontiers in Neurology | www.frontiersin.org 11 September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
68. Choi KH, Kim JH, Kim JM, Kang KW, Lee C, Kim JT, et al.
d-dimer level as a predictor of recurrent stroke in patients
with embolic stroke of undetermined source. Stroke. (2021)
52:2292–301. doi: 10.1161/STROKEAHA.120.033217
69. Xu T, Zuo P, C ao L, Gao Z, Ke K. Omentin-1 is associated with carotid plaque
instability among ischemic stroke patients. J Atheroscler Thromb. (2018)
25:505–11. doi: 10.5551/jat.42135
70. Yanofsky R, Sancho C, Gasbarrino K, Zheng H, Doonan RJ, Jaunet
F, ET AL. Expression of Resistin, Chemerin, and Chemerin’s receptor
in the unstable carotid atherosclerotic plaque. Stroke. (2021) 52:2537–
46. doi: 10.1161/STROKEAHA.120.030228
71. Eltoft A, Arntzen KA, Wilsgaard T, Mathiesen EB, Johnsen SH.
Interleukin-6 is an independent predictor of progressive atherosclerosis
in the carotid artery: the Tromso Study. Atherosclerosis. (2018) 271:1–
8. doi: 10.1016/j.atherosclerosis.2018.02.005
72. Ridker PM. From RESCUE to ZEUS: will interleukin-6 inhibition with
ziltivekimab prove effective for cardiovascular event reduction? Cardiovasc
Res. (2021). doi: 10.1093/cvr/cvab231. [Epub ahead of print].
73. Ridker PM, Devalaraja M, Baeres FMM, Engelmann MDM,
Hovingh GK, Ivkovic M, et al. IL-6 inhibition with ziltivekimab
in patients at high atherosclerotic risk (RESCUE): a double-
blind, randomised, placebo-controlled, phase 2 trial. Lancet. (2021)
397:2060–9. doi: 10.1016/S0140-6736(21)00520-1
74. Ridker PM, Rane M. Interleukin-6 signaling and anti-interleukin-
6 therapeutics in cardiovascular disease. Circ Res. (2021)
128:1728–46. doi: 10.1161/CIRCRESAHA.121.319077
75. Pothineni NVK, Karathanasis SK, Ding Z, Arulandu A, Varughese
KI, Mehta JL. LOX-1 in atherosclerosis and myocardial ischemia:
biology, genetics, and modulation. J Am Coll Cardiol. (2017) 69:2759–
68. doi: 10.1016/j.jacc.2017.04.010
76. Si W, He P, Wang Y, Fu Y, Li X, Lin X, et al. Complement complex
C5b-9 levels are associated with the clinical outcomes of acute ischemic
stroke and carotid plaque stability. Transl Stroke Res. (2018) 10:279–
86. doi: 10.1007/s12975-018-0658-3
77. Shi X, Xie WL, Kong WW, Chen D, Qu P. Expression of the NLRP3
inflammasome in carotid atherosclerosis. J Stroke Cerebrovasc Dis. (2015)
24:2455–66. doi: 10.1016/j.jstrokecerebrovasdis.2015.03.024
78. Arthurs ZM, Andersen C, Starnes BW, Sohn VY, Mullenix PS, Perry
J. A prospective evaluation of C-reactive protein in the progression
of carotid artery stenosis. J Vasc Surg. (2008) 47:744–50; discussion
751. doi: 10.1016/j.jvs.2007.11.066
79. Klein JH, Hegele RA, Hackam DG, Koschinsky ML, Huff MW, Spence JD.
Lipoprotein(a) is associated differentially with carotid stenosis, occlusion,
and total plaque area. Arterioscler Thromb Vasc Biol. (2008) 28:1851–
6. doi: 10.1161/ATVBAHA.108.169292
80. Muramatsu Y, Minami Y, Kato A, Katsura A, Sato T, Kakizaki R, et al.
Lipoprotein (a) level is associated with plaque vulnerability in patients with
coronary artery disease: an optical coherence tomography study. Int J Cardiol
Heart Vasc. (2019) 24:100382. doi: 10.1016/j.ijcha.2019.100382
81. Rehberger Likozar A, Zavrtanik M, Sebestjen M. Lipoprotein(a) in
atherosclerosis: from pathophysiology to clinical relevance and treatment
options. Ann Med. (2020) 52:162–77. doi: 10.1080/07853890.2020.1775287
82. Ganji M, Nardi V, Prasad M, Jordan KL, Bois MC, Franchi F,
et al. carotid plaques from symptomatic patients are characterized by
local increase in xanthine oxidase expression. Stroke. (2021) 52:1636–
42. doi: 10.1161/STROKEAHA.120.032964
83. Stability Investigators, White HD, Held C, Stewart R, Tarka E, Brown R, et al.
Darapladib for preventing ischemic events in stable coronary heart disease.
N Engl J Med. (2014) 370:1702–11. doi: 10.1056/NEJMoa1315878
84. Pires N, Gota V, Gulia A, Hingorani L, Agarwal M, Puri A. Safety
and pharmacokinetics of Withaferin-A in advanced stage high grade
osteosarcoma: a phase I trial. J Ayurveda Integr Med. (2020) 11:68–
72. doi: 10.1016/j.jaim.2018.12.008
85. Thurman JM. New anti-complement drugs: not so far away. Blood. (2014)
123:1975–6. doi: 10.1182/blood-2014-02-555805
86. Ridker PM. Anticytokine agents: targeting interleukin signaling pathways
for the treatment of atherothrombosis. Circ Res. (2019) 124:437–
50. doi: 10.1161/CIRCRESAHA.118.313129
87. Ridker PM. From CANTOS to CIRT to COLCOT to clinic: will
all atherosclerosis patients soon be treated with combination lipid-
lowering and inflammation-inhibiting agents? Circulation. (2020) 141:787–
9. doi: 10.1161/CIRCULATIONAHA.119.045256
88. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne
C, et al. Antiinflammatory therapy with canakinumab for atherosclerotic
disease. N Engl J Med. (2017) 377:1119–31. doi: 10.1056/NEJMoa1707914
89. Ridker PM. From C-reactive protein to interleukin-6 to interleukin-1:
moving upstream to identify novel targets for atheroprotection. Circ Res.
(2016) 118:145–56. doi: 10.1161/CIRCRESAHA.115.306656
90. Kamtchum-Tatuene J, Jickling GC. Letter by Kamtchum-Tatuene
and Jickling Regarding Article, “Elevated Lp(a) (Lipoprotein[a])
Levels Increase Risk of 30-Day Major Adverse Cardiovascular
Events in Patients Following Carotid Endarterectomy”. Stroke. (2021)
52:e64–5. doi: 10.1161/STROKEAHA.120.032698
91. Tsimikas S, Karwatowska-Prokopczuk E, Gouni-Berthold I, Tardif JC,
Baum SJ, Steinhagen-Thiessen E, et al. Lipoprotein(a) reduction in
persons with cardiovascular disease. N Engl J Med. (2020) 382:244–
55. doi: 10.1056/NEJMoa1905239
92. Hegele RA, Tsimikas S. Lipid-lowering agents. Circ Res. (2019) 124:386–
404. doi: 10.1161/CIRCRESAHA.118.313171
93. Michos ED, McEvoy JW, Blumenthal RS. Lipid management for the
prevention of atherosclerotic cardiovascular disease. N Engl J Med. (2019)
381:1557–67. doi: 10.1056/NEJMra1806939
94. Magenta A, Sileno S, D’Agostino M, Persiani F, Beji S, Paolini
A, et al. Atherosclerotic plaque instability in carotid arteries:
miR-200c as a promising biomarker. Clin Sci. (2018) 132:2423–
36. doi: 10.1042/CS20180684
95. Putaala J, Martinez-Majander N, Saeed S, Yesilot N, Jakala P, Nerg O, et al.
Searching for explanations for cryptogenic stroke in the young: revealing the
triggers, causes, and outcome (SECRETO): rationale and design. Eur Stroke
J. (2017) 2:116–25. doi: 10.1177/2396987317703210
96. Jauch EC, Barreto AD, Broderick JP, Char DM, Cucchiara BL, Devlin TG,
et al. Biomarkers of Acute Stroke Etiology (BASE) study methodology. Transl
Stroke Res. (2017) 8:424–8. doi: 10.1007/s12975-017-0537-3
97. Chang AD, Ignacio GC, Akiki R, Grory BM, Cutting SS, Burton
T, et al. increased left atrial appendage density on computerized
tomography is associated with cardioembolic stroke. J Stroke Cerebrovasc
Dis. (2020) 29:104604. doi: 10.1016/j.jstrokecerebrovasdis.2019.
104604
98. Ricci B, Chang AD, Hemendinger M, Dakay K, Cutting S, Burton T, et al.
A simple score that predicts paroxysmal atrial fibrillation on outpatient
cardiac monitoring after embolic stroke of unknown source. J Stroke
Cerebrovasc Dis. (2018) 27:1692–6. doi: 10.1016/j.jstrokecerebrovasdis.2018.
01.028
99. Ntaios G, Perlepe K, Lambrou D, Sirimarco G, Strambo D,
Eskandari A, et al. External performance of the HAVOC score
for the prediction of new incident atrial fibrillation. Stroke. (2020)
51:457–61. doi: 10.1161/STROKEAHA.119.027990
100. Ntaios G, Perlepe K, Lambrou D, Sirimarco G, Strambo D, Eskandari A, et al.
Identification of patients with embolic stroke of undetermined source and
low risk of new incident atrial fibrillation: the AF-ESUS score. Int J Stroke.
(2021) 16:29–38. doi: 10.1177/1747493020925281
101. Kamel H, Longstreth WT Jr, Tirschwell DL, Kronmal RA, Broderick
JP, Palesch YY, et al. The AtRial cardiopathy and antithrombotic
drugs in prevention after cryptogenic stroke randomized trial: rationale
and methods. Int J Stroke. (2018) 14:207–14. doi: 10.1177/17474930187
99981
102. Zhang K, Kamtchum-Tatuene J, Li M, Jickling GC. Cardiac natriuretic
peptides for diagnosis of covert atrial fibrillation after acute ischaemic stroke:
a meta-analysis of diagnostic accuracy studies. Stroke Vasc Neurol. (2020)
6:128–32. doi: 10.1136/svn-2020-000440
103. Goyal M, Singh N, Marko M, Hill MD, Menon BK,
Demchuk A, et al. Embolic stroke of undetermined source
and symptomatic nonstenotic carotid disease. Stroke. (2020)
51:1321–5. doi: 10.1161/STROKEAHA.119.028853
104. Strambo D, Sirimarco G, Nannoni S, Perlepe K, Ntaios G, Vemmos K,
et al. Embolic stroke of undetermined source and patent foramen ovale: risk
Frontiers in Neurology | www.frontiersin.org 12 September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
of paradoxical embolism score validation and atrial fibrillation prediction.
Stroke. (2021) 52:1643–52. doi: 10.1161/STROKEAHA.120.032453
105. Johnston SC, Easton JD, Farrant M, Barsan W, Conwit RA, Elm JJ, et al.
Clopidogrel and aspirin in acute ischemic stroke and high-risk TIA. N Engl J
Med. (2018) 379:215–25. doi: 10.1056/NEJMoa1800410
106. Wang Y, Wang Y, Zhao X, Liu L, Wang D, Wang C, et al. Clopidogrel with
aspirin in acute minor stroke or transient ischemic attack. N Engl J Med.
(2013) 369:11–9. doi: 10.1056/NEJMoa1215340
107. Lee M, Saver JL, Hong KS, Rao NM, Wu YL, Ovbiagele B.
antiplatelet regimen for patients with breakthrough strokes while
on aspirin: a systematic review and meta-analysis. Stroke. (2017)
48:2610–3. doi: 10.1161/STROKEAHA.117.017895
108. Pan Y, Elm JJ, Li H, Easton JD, Wang Y, Farrant M, et al. Outcomes
associated with clopidogrel-aspirin use in minor stroke or transient ischemic
attack: a pooled analysis of clopidogrel in high-risk patients with acute non-
disabling cerebrovascular events (CHANCE) and platelet-oriented inhibition
in new TIA and minor ischemic stroke (POINT) trials. JAMA Neurol. (2019)
76:1466–73. doi: 10.1001/jamaneurol.2019.2531
109. Damman P, Woudstra P, Kuijt WJ, de Winter RJ, James SK. P2Y12 platelet
inhibition in clinical practice. J Thromb Thrombolysis. (2012) 33:143–
53. doi: 10.1007/s11239-011-0667-5
110. Pan Y, Chen W, Xu Y, Yi X, Han Y, Yang Q, et al. Genetic polymorphisms
and clopidogrel efficacy for acute ischemic stroke or transient ischemic
attack: a systematic review and meta-analysis. Circulation. (2017) 135:21–
33. doi: 10.1161/CIRCULATIONAHA.116.024913
111. Johnston SC, Amarenco P, Denison H, Evans SR, Himmelmann A, James S,
et al. Ticagrelor and aspirin or aspirin alone in acute ischemic stroke or TIA.
N Engl J Med. (2020) 383:207–17. doi: 10.1056/NEJMoa1916870
112. Li ZX, Xiong Y, Gu HQ, Fisher M, Xian Y, Johnston SC, Wang YJ.
P2Y12 inhibitors plus aspirin versus aspirin alone in patients with minor
stroke or high-risk transient ischemic attack. Stroke. (2021) 52:2250–
7. doi: 10.1161/STROKEAHA.120.033040
113. Wang Y, Johnston C, Bath PM, Meng X, Jing J, Xie X, et al. Clopidogrel
with aspirin in high-risk patients with acute non-disabling cerebrovascular
events II (CHANCE-2): rationale and design of a multicentre randomised
trial. Stroke Vasc Neurol. (2021) 6:280–5. doi: 10.1136/svn-2020-000791
114. Xiong Y, Bath PM. Antiplatelet therapy for transient
ischemic attack and minor stroke. Stroke. (2020) 51:3472–
4. doi: 10.1161/STROKEAHA.120.031763
115. Noubiap JJ, Agbaedeng TA, Kamtchum-Tatuene J, Fitzgerald JL,
Middeldorp ME, Kleinig T, et al. Rhythm monitoring strategies for
atrial fibrillation detection in patients with cryptogenic stroke: a
systematic review and meta-analysis. Int J Cardiol Heart Vasc. (2021)
34:100780. doi: 10.1016/j.ijcha.2021.100780
116. Perera KS, Ng KKH, Nayar S, Catanese L, Dyal L, Sharma M, et al.
Association between low-dose rivaroxaban with or without aspirin
and ischemic stroke subtypes: a secondary analysis of the COMPASS
trial. JAMA Neurol. (2020) 77:43–8. doi: 10.1001/jamaneurol.201
9.2984
117. Sharma M, Hart RG, Connolly SJ, Bosch J, Shestakovska O, Ng KKH,
et al. Stroke outcomes in the COMPASS trial. Circulation. (2019) 139:1134–
45. doi: 10.1161/CIRCULATIONAHA.118.035864
118. Healey JS, Gladstone DJ, Swaminathan B, Eckstein J, Mundl H, Epstein
AE, et al. Recurrent stroke with rivaroxaban compared with aspirin
according to predictors of atrial fibrillation: secondary analysis of the
NAVIGATE esus randomized clinical trial. JAMA Neurol. (2019) 76:764–
73. doi: 10.1001/jamaneurol.2019.0617
119. Mazzucco S, Li L, Binney L, Rothwell PM, Oxford vascular study
phenotyped C. prevalence of patent foramen ovale in cryptogenic
transient ischaemic attack and non-disabling stroke at older ages:
a population-based study, systematic review, and meta-analysis.
Lancet Neurol. (2018) 17:609–17. doi: 10.1016/S1474-4422(18)
30167-4
120. Mazzucco S, Li L, Rothwell PM. Prognosis of cryptogenic stroke with
patent foramen ovale at older ages and implications for trials: a population-
based study and systematic review. JAMA Neurol. (2020) 77:1279–
87. doi: 10.1001/jamaneurol.2020.1948
121. Yaghi S, Boehme AK, Hazan R, Hod EA, Canaan A, Andrews
HF, et al. Atrial cardiopathy and cryptogenic stroke: a cross-
sectional pilot study. J Stroke Cerebrovasc Dis. (2016) 25:110–
4. doi: 10.1016/j.jstrokecerebrovasdis.2015.09.001
122. Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC,
Becker K, et al. 2018 Guidelines for the early management of patients with
acute ischemic stroke: a guideline for healthcare professionals from the
American Heart Association/American Stroke Association. Stroke. (2018)
49:e46–110. doi: 10.1161/STR.0000000000000158
123. Amarenco P, Kim JS, Labreuche J, Charles H, Abtan J, Bejot Y, et al. A
comparison of two LDL cholesterol targets after ischemic stroke. N Engl J
Med. (2020) 382:9. doi: 10.1056/NEJMoa1910355
124. Turan TN, Voeks JH, Chimowitz MI, Roldan A, LeMatty
T, Haley W, et al. Rationale, design, and implementation of
intensive risk factor treatment in the CREST2 trial. Stroke. (2020)
51:2960–71. doi: 10.1161/STROKEAHA.120.030730
125. Cannon CP. Don’t stop the statin! Eur Heart J. (2019) 40:3526–
8. doi: 10.1093/eurheartj/ehz629
126. Raal FJ, Mohamed F. Never too old to benefit from lipid-lowering treatment.
Lancet. (2020) 396:1608–9. doi: 10.1016/S0140-6736(20)32333-3
127. Cheung BMY, Lam KSL. Never too old for statin treatment? Lancet. (2019)
393:379–80. doi: 10.1016/S0140-6736(18)32263-3
128. Dearborn-Tomazos JL, Hu X, Bravata DM, Phadke MA, Baye FM, Myers
LJ, et al. Deintensification or no statin treatment is associated with higher
mortality in patients with ischemic stroke or transient ischemic attack.
Stroke. (2021) 52:2521–9. doi: 10.1161/STROKEAHA.120.030089
129. Sillesen H, Amarenco P, Hennerici MG, Callahan A, Goldstein LB, Zivin
J, et al. Atorvastatin reduces the risk of cardiovascular events in patients
with carotid atherosclerosis: a secondary analysis of the Stroke Prevention
by Aggressive Reduction in Cholesterol Levels (SPARCL) trial. Stroke. (2008)
39:3297–302. doi: 10.1161/STROKEAHA.108.516450
130. Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson J, et al.
Efficacy and safety of evolocumab in reducing lipids and cardiovascular
events. N Engl J Med. (2015) 372:1500–9. doi: 10.1056/NEJMoa1500858
131. Schwartz GG, Steg PG, Szarek M, Bhatt DL, Bittner VA, Diaz R, et al.
Alirocumab and cardiovascular outcomes after acute coronary syndrome. N
Engl J Med. (2018) 379:2097–107. doi: 10.1056/NEJMoa1801174
132. Julius U, Tselmin S, Schatz U, Fischer S, Bornstein SR. Lipoprotein(a) and
proprotein convertase subtilisin/kexin type 9 inhibitors. Clin Res Cardiol
Suppl. (2019) 14:45–50. doi: 10.1007/s11789-019-00099-z
133. Rosenson RS, Burgess LJ, Ebenbichler CF, Baum SJ, Stroes ESG, Ali S, et al.
Evinacumab in patients with refractory hypercholesterolemia. N Engl J Med.
(2020) 383:2307–19. doi: 10.1056/NEJMoa2031049
134. Ruscica M, Zimetti F, Adorni MP, Sirtori CR, Lupo MG, Ferri N.
Pharmacological aspects of ANGPTL3 and ANGPTL4 inhibitors: new
therapeutic approaches for the treatment of atherogenic dyslipidemia.
Pharmacol Res. (2020) 153:104653. doi: 10.1016/j.phrs.2020.104653
135. Di Minno A, Lupoli R, Calcaterra I, Poggio P, Forte F, Spadarella G, et al.
Efficacy and safety of Bempedoic acid in patients with hypercholesterolemia:
systematic review and meta-analysis of randomized controlled trials. J Am
Heart Assoc. (2020) 9:e016262. doi: 10.1161/JAHA.119.016262
136. Awad K, Mikhailidis DP, Katsiki N, Muntner P, Banach M, Lipid,
Blood Pressure Meta-Analysis Collaboration Group. Effect of
ezetimibe monotherapy on plasma Lipoprotein(a) concentrations
in patients with primary hypercholesterolemia: a systematic review
and meta-analysis of randomized controlled trials. Drugs. (2018)
78:453–62. doi: 10.1007/s40265-018-0870-1
137. Tsimikas S, Gordts P, Nora C, Yeang C, Witztum JL. Statin
therapy increases lipoprotein(a) levels. Eur Heart J. (2020)
41:2275–84. doi: 10.1093/eurheartj/ehz310
138. Beheshtian A, Shitole SG, Segal AZ, Leifer D, Tracy RP, Rader DJ, et al.
Lipoprotein (a) level, apolipoprotein (a) size, and risk of unexplained
ischemic stroke in young and middle-aged adults. Atherosclerosis. (2016)
253:47–53. doi: 10.1016/j.atherosclerosis.2016.08.013
139. Lin WV, Vickers A, Prospero Ponce CM, Lee AG. Elevated lipoprotein(a)
levels as the cause of cryptogenic stroke in a young Ashkenazi Jewish female.
Can J Ophthalmol. (2019) 54:e126–8. doi: 10.1016/j.jcjo.2018.07.011
Frontiers in Neurology | www.frontiersin.org 13 September 2021 | Volume 12 | Article 719329
Kamtchum-Tatuene et al. Non-stenotic Carotid Plaques in ESUS
140. Esenwa CC, Elkind MS. Inflammatory risk factors, biomarkers and
associated therapy in ischaemic stroke. Nat Rev Neurol. (2016) 12:594–
604. doi: 10.1038/nrneurol.2016.125
141. Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J,
Bittencourt MS, et al. Atherosclerosis. Nat Rev Dis Primers. (2019)
5:56. doi: 10.1038/s41572-019-0106-z
142. Puri R, Nissen SE, Arsenault BJ, St John J, Riesmeyer JS, Ruotolo G, et al.
Effect of C-reactive protein on Lipoprotein(a)-associated cardiovascular risk
in optimally treated patients with high-risk vascular disease: a prespecified
secondary analysis of the ACCELERATE trial. JAMA Cardiol. (2020) 5:1136–
43. doi: 10.1001/jamacardio.2020.2413
143. Lawler PR, Bhatt DL, Godoy LC, Luscher TF, Bonow RO, Verma
S, et al. Targeting cardiovascular inflammation: next steps in clinical
translation. Eur Heart J. (2021) 42:113–31. doi: 10.1093/eurheartj/
ehaa099
144. Brott TG, Halperin JL, Abbara S, Bacharach JM, Barr JD, Bush
RL, et al. 2011 ASA/ACCF/AHA/AANN/AANS/ACR/ASNR/CNS/
SAIP/SCAI/SIR/SNIS/SVM/SVS guideline on the management of patients
with extracranial carotid and vertebral artery disease: executive summary:
a report of the American College of Cardiology Foundation/American
Heart Association Task Force on Practice Guidelines, and the American
Stroke Association, American Association of Neuroscience Nurses,
American Association of Neurological Surgeons, American College of
Radiology, American Society of Neuroradiology, Congress of Neurological
Surgeons, Society of Atherosclerosis Imaging and Prevention, Society for
Cardiovascular Angiography and Interventions, Society of Interventional
Radiology, Society of NeuroInterventional Surgery, Society for Vascular
Medicine, and Society for Vascular Surgery. J Am Coll Cardiol. (2011)
57:1002–44. doi: 10.1016/j.jacc.2010.11.005
145. Naylor AR, Ricco JB, de Borst GJ, Debus S, de Haro J, Halliday A, et al.
Editor’s Choice - management of atherosclerotic carotid and vertebral
artery disease: 2017 clinical practice guidelines of the European Society
for Vascular Surgery (ESVS). Eur J Vasc Endovasc Surg. (2018) 55:3–
81. doi: 10.1016/j.ejvs.2017.06.021
146. Bonati LH, Kakkos S, Berkefeld J, de Borst GJ, Bulbulia R,
Halliday A, et al. European Stroke Organisation guideline
on endarterectomy and stenting for carotid artery stenosis.
Eur Stroke J. (2021) 6:I–XLVII. doi: 10.1177/239698732110
26990
147. Katsanos AH, Palaiodimou L, Price C, Giannopoulos S, Lemmens R,
Kosmidou M, et al. Colchicine for stroke prevention in patients with
coronary artery disease: a systematic review and meta-analysis. Eur J Neurol.
(2020) 27:1035–8. doi: 10.1111/ene.14198
148. Kleindorfer DO, Towfighi A, Chaturvedi S, Cockroft KM, Gutierrez J,
Lombardi-Hill D, et al. 2021 Guideline for the prevention of stroke in
patients with stroke and transient ischemic attack: a guideline From the
American Heart Association/American Stroke Association. Stroke. (2021)
52:e364–467. doi: 10.1161/STR.0000000000000375
149. Rippe JM. Lifestyle strategies for risk factor reduction, prevention, and
treatment of cardiovascular disease. Am J Lifestyle Med. (2019) 13:204–
12. doi: 10.1177/1559827618812395
150. Munzel T, Sorensen M, Daiber A. Transportation noise
pollution and cardiovascular disease. Nat Rev Cardiol. (2021)
18:619–36. doi: 10.1038/s41569-021-00532-5
151. Munzel T, Sorensen M, Gori T, Schmidt FP, Rao X, Brook FR, et al.
Environmental stressors and cardio-metabolic disease: part II-mechanistic
insights. Eur Heart J. (2017) 38:557–64. doi: 10.1093/eurheartj/ehw294
152. Munzel T, Sorensen M, Gori T, Schmidt FP, Rao X, Brook J,
et al. Environmental stressors and cardio-metabolic disease: part
I-epidemiologic evidence supporting a role for noise and air
pollution and effects of mitigation strategies. Eur Heart J. (2017)
38:550–6. doi: 10.1093/eurheartj/ehw269
153. McAlpine CS, Kiss MG, Rattik S, He S, Vassalli A, Valet C, et al. Sleep
modulates haematopoiesis and protects against atherosclerosis. Nature.
(2019) 566:383–7. doi: 10.1038/s41586-019-0948-2
154. Leng Y, Cappuccio FP, Wainwright NW, Surtees PG, Luben R,
Brayne C, et al. Sleep duration and risk of fatal and nonfatal
stroke: a prospective study and meta-analysis. Neurology. (2015)
84:1072–9. doi: 10.1212/WNL.0000000000001371
155. Estruch R, Ros E, Salas-Salvado J, Covas MI, Corella D, Aros F, et al.
Primary prevention of cardiovascular disease with a mediterranean diet
supplemented with extra-virgin olive oil or nuts. N Engl J Med. (2018)
378:e34. doi: 10.1056/NEJMoa1800389
Conflict of Interest: The authors declare that the research was conducted in the
absence of any commercial or financial relationships that could be construed as a
potential conflict of interest.
Publisher’s Note: All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their affiliated organizations, or those of
the publisher, the editors and the reviewers. Any product that may be evaluated in
this article, or claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Copyright © 2021 Kamtchum-Tatuene, Nomani, Falcione, Munsterman, Sykes, Joy,
Spronk, Vargas and Jickling. This is an open-access article distributed under the
terms of the Creative Commons Attribution License (CC BY). The use, distribution
or reproduction in other forums is permitted, provided the original author(s) and
the copyright owner(s) are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Frontiers in Neurology | www.frontiersin.org 14 September 2021 | Volume 12 | Article 719329
... 20 Therefore, antiplatelet therapy can of course reduce the risk of subsequent cerebrovascular events in most of these patients, but the efficacy may largely be limited in a certain subgroup of patients with IPH. 21 Recent meta-analysis has analyzed the risk for ipsilateral ischemic stroke and any operative stroke or death in patients with mild carotid stenosis. As the results, the annual risk in medically treated patients was 3.0% in patients with 30-49% stenosis and 1.8% in those with <30% stenosis. ...
Preprint
Background and Purpose – Plaque composition, but not stenosis degree, may play a key role in the development of recurrent ischemic events in patients with symptomatic, mild (<50%) carotid stenosis. This multi-center prospective cohort study was aimed to determine their clinical and radiological features and to evaluate the benefits of carotid endarterectomy (CEA) for them. Methods ? This study included 124 patients with cerebrovascular or retinal ischemic events ipsilateral to mild carotid stenosis. Best medical therapy (BMT) was administered to all participants. CEA or carotid artery stenting (CAS) was implemented at each institution's discretion. Baseline and 6-, 12-, and 24-month follow-up data were collected. Primary endpoint was ipsilateral ischemic stroke. Secondary endpoints included any stroke, ipsilateral TIA, ipsilateral ocular symptoms, any death, and plaque progression requiring CEA/CAS. Multivariate Cox proportional hazard model was used to evaluate the predictors for each endpoint. Results – Of 124 patients, 70 patients (56.5%) had the history of ipsilateral ischemic stroke and 51 (43.5%) had been treated with antiplatelet agents. Mean stenosis degree was 22.4±13.7%. Plaque composition was categorized into fibrous plaque in 22 patients, lipid-rich/necrotic core (LR/NC) in 25, and intraplaque hemorrhage (IPH) in 69. BMT was indicated for 59 patients, while CEA was performed in 63. The incidence of primary endpoint was significantly higher in BMT group than in CEA group (15.1% vs. 1.7%; HR, 0.18; 95%CI, 0.05–0.84, P=0.03). The predictors for ipsilateral ischemic stroke were CEA (HR, 0.18; 95%CI, 0.05–0.84, P=0.03) and IPH (HR, 1.92; 95%CI, 1.26–4.28, P=0.04). The results on secondary endpoints were very similar. Conclusion – IPH may highly predict subsequent cerebrovascular events, whereas CEA may reduce these risk during a 2-year follow-up in patients with symptomatic, mild carotid stenosis. Randomized clinical trials is warranted to validate these results.
... Currently, the traditional clinical diagnosis technologies for AS, such as angiography, optical coherence tomography (OCT) and intravascular ultrasound (IUVS) (Liang et al., 2018;Chow et al., 2021), can only provide the prediction of cardiovascular and cerebrovascular events by quantifying the percentage of lumen occlusion. Plaque only with severe stenosis are considered as dangerous plaque, which greatly underestimates the high-risk of mild-stenosis plaque while a large number of acute vascular events occured in mild-stenosis (stenosis<50%) (Kamtchum-Tatuene et al., 2021). The risk of vulnerable carotid plaque should be attributed not only to the degree of stenosis, but also to the plaque composition. ...
Article
Full-text available
Atherosclerotic plaque remains the primary cause of morbidity and mortality worldwide. Accurate assessment of the degree of atherosclerotic plaque is critical for predicting the risk of atherosclerotic plaque and monitoring the results after intervention. Compared with traditional technology, the imaging technologies of nanoparticles have distinct advantages and great development prospects in the identification and characterization of vulnerable atherosclerotic plaque. Here, we systematically summarize the latest advances of targeted nanoparticle approaches in the diagnosis of atherosclerotic plaque, including multimodal imaging, fluorescence imaging, photoacoustic imaging, exosome diagnosis, and highlighted the theranostic progress as a new therapeutic strategy. Finally, we discuss the major challenges that need to be addressed for future development and clinical transformation.
... As stenosis was assessed non-invasively in our study, the primary analysis was performed assuming a randomly assigned 50% of the cohort had ulcerated plaque, with sensitivity analysis assuming prevalence of plaque ulceration of 25%, based on published prevalence data of plaque ulceration in symptomatic carotid stenosis. 11,12 The SCAIL score was assigned based on degree of carotid stenosis and inflammation measured by the maximum standardised uptake value (SUV max ) on 18 FDG-PET in the symptomatic carotid plaque ( Table 1). ...
Article
Background: The Oxford Carotid Stenosis tool (OCST) and Essen Stroke Risk Score (ESRS) are validated to predict recurrent stroke in patients with and without carotid stenosis. The Symptomatic Carotid Atheroma Inflammation Lumen stenosis (SCAIL) score combines stenosis and plaque inflammation on fluorodeoxyglucose positron-emission tomography (18FDG-PET). We compared SCAIL with OCST and ESRS to predict ipsilateral stroke recurrence in symptomatic carotid stenosis. Patients and methods: We pooled three prospective cohort studies of patients with recent (<30 days) non-severe ischaemic stroke/TIA and internal carotid artery stenosis (>50%). All patients had carotid 18FDG-PET/CT angiography and late follow-up, with censoring at carotid revascularisation. Results: Of 212 included patients, 16 post-PET ipsilateral recurrent strokes occurred in 343 patient-years follow-up (median 42 days (IQR 13-815)).Baseline SCAIL predicted recurrent stroke (unadjusted hazard ratio [HR] 1.96, CI 1.20-3.22, p = 0.007, adjusted HR 2.37, CI 1.31-4.29, p = 0.004). The HR for OCST was 0.996 (CI 0.987-1.006, p = 0.49) and for ESRS was 1.26 (CI 0.87-1.82, p = 0.23) (all per 1-point score increase). C-statistics were: SCAIL 0.66 (CI 0.51-0.80), OCST 0.52 (CI 0.40-0.64), ESRS 0.61 (CI 0.48-0.74). Compared with ESRS, addition of plaque inflammation (SUVmax) to ESRS improved risk prediction when analysed continuously (HR 1.51, CI 1.05-2.16, p = 0.03) and categorically (ptrend = 0.005 for risk increase across groups; HR 3.31, CI 1.42-7.72, p = 0.006; net reclassification improvement 10%). Findings were unchanged by further addition of carotid stenosis. Conclusions: SCAIL predicted recurrent stroke, had discrimination better than chance, and improved the prognostic utility of ESRS, suggesting that measuring plaque inflammation may improve risk stratification in carotid stenosis.
... Of note, the study cohort consisted of patients with low-to moderate-grade carotid stenosis and may not be representative for patients with more significant stenosis. Nevertheless, as patients with ischemic cerebral symptoms and a carotid stenosis below 50% (NASCET) are not recommended for intervention by most guidelines [35] and rather categorized as suffering from 'embolic stroke of unknown source' [36], these patients would likely benefit the most from a predictive biomarker, such as plasma BLVRB, to guide clinical decision making. Lastly, it should be noted that the assessment of changes in plasma BLVRB levels following VEGFR-2 was performed exclusively in a preclinical mouse model. ...
Article
Full-text available
Background: Intraplaque hemorrhage (IPH) is a hallmark of atherosclerotic plaque instability. Biliverdin reductase B (BLVRB) is enriched in plasma and plaques from patients with symptomatic carotid atherosclerosis and functionally associated with IPH. Objective: We explored the biomarker potential of plasma BLVRB through (1) its correlation with IPH in carotid plaques assessed by magnetic resonance imaging (MRI), and with recurrent ischemic stroke, and (2) its use for monitoring pharmacotherapy targeting IPH in a preclinical setting. Methods: Plasma BLVRB levels were measured in patients with symptomatic carotid atherosclerosis from the PARISK study (n = 177, 5 year follow-up) with and without IPH as indicated by MRI. Plasma BLVRB levels were also measured in a mouse vein graft model of IPH at baseline and following antiangiogenic therapy targeting vascular endothelial growth factor receptor 2 (VEGFR-2). Results: Plasma BLVRB levels were significantly higher in patients with IPH (737.32 ± 693.21 vs. 520.94 ± 499.43 mean fluorescent intensity (MFI), p = 0.033), but had no association with baseline clinical and biological parameters. Plasma BLVRB levels were also significantly higher in patients who developed recurrent ischemic stroke (1099.34 ± 928.49 vs. 582.07 ± 545.34 MFI, HR = 1.600, CI [1.092-2.344]; p = 0.016). Plasma BLVRB levels were significantly reduced following prevention of IPH by anti-VEGFR-2 therapy in mouse vein grafts (1189 ± 258.73 vs. 1752 ± 366.84 MFI; p = 0.004). Conclusions: Plasma BLVRB was associated with IPH and increased risk of recurrent ischemic stroke in patients with symptomatic low- to moderate-grade carotid stenosis, indicating the capacity to monitor the efficacy of IPH-preventive pharmacotherapy in an animal model. Together, these results suggest the utility of plasma BLVRB as a biomarker for atherosclerotic plaque instability.
... High-risk ultrasound features reported in the identified studies include degree of stenosis, intraplaque hemorrhage, lipid-rich necrotic core, as well as both echolucency/high echogenicity and low echogenicity, although no quantitative thresholds were provided by the authors 31 (online supplemental table 1). One review discussed the potential advantages of contrast-enhanced ultrasound, 24 which is, however, not routinely used in most centers. ...
Article
Symptomatic non-stenotic carotid plaques (SyNC) are an under-researched and under-recognized source of stroke. Various imaging markers of non-stenotic carotid plaques that are associated with stroke risk have been identified, but these causal relationships need to be confirmed in additional prospective studies. Currently, there exists neither a standardized SyNC definition nor a dedicated set of imaging protocols, although researchers have started to address these shortcomings. Moreover, many neuroradiologists are still unaware of the condition, and hence do not comment on high-risk plaque features other than stenosis in their reports. Regarding SyNC treatment, scant data exist as to whether and to what extent medical, interventional and surgical treatments could influence the course of the disease; the relative lack of data on the ‘natural’ history of untreated SyNC makes treatment comparisons difficult. In our opinion, endovascular SyNC treatment represents the most promising treatment option for SyNC, since it allows for targeted elimination of the embolic source, with few systemic side effects and without the need for general anesthesia. However, currently available carotid devices are designed to treat stenotic lesions, and thus are not optimally designed for SyNC. Developing a device specifically tailored to SyNC could be an important step towards establishing endovascular SyNC treatment in clinical practice. In this review, we provide an overview of the current state of evidence with regard to epidemiological, clinical and imaging features of SyNC, propose a SyNC definition based on imaging and clinical features, and outline a possible pathway towards evidence-based SyNC therapies, with a special focus on endovascular SyNC treatment.
... However, two randomized controlled trials did not show a superiority of oral anticoagulants over antiplatelet therapy in ESUS (4), implying that a proper work-up for identifying the most probable mechanism of stroke etiology is still warranted in patients who suffered a cryptogenic stroke. In addition to atrial cardiopathy, potential embolic sources in ESUS include non-stenotic carotid plaques (5,6), aortic atherosclerosis (7,8), cardiac valvulopathies, coagulopathies or paradoxical embolism via patent foramen ovale (PFO), and others (9). Interestingly, in a sub-analysis of the NAVIGATE-ESUS trial, Ntaios et al. showed that ∼40% of patients with ESUS revealed multiple potential embolic sources (10). ...
Article
Full-text available
Background The identification of the underlying mechanism in ischemic stroke has important implications for secondary prevention. A disintegrin and metalloprotease with a thrombospondin type 1 motif, member 13 (ADAMTS-13) has antithrombotic properties and was repeatedly implicated in the pathophysiology of stroke. In this study, we, therefore, aimed to investigate whether ADAMTS-13 is associated with stroke etiology and the burden of vascular risk factors. Methods We determined ADAMTS-13 activity in two prospectively recruited stroke cohorts in the long-term course after the event. Cohort 1 (n = 88) consisted of patients who suffered a stroke due to embolic stroke of undetermined source (ESUS), cardioembolic stroke due to atrial fibrillation (AF), large-artery atherosclerosis, or small vessel disease. In cohort 2, patients with cryptogenic stroke and patent foramen ovale (PFO) scheduled for PFO closure (n = 38) were enrolled. As measures of vascular risk factor burden, the CHA2DS2VASC score, the Essen Stroke Risk Score (ESRS), and the Risk of Paradoxical Embolism (RoPE) score were calculated, as appropriate. Results ADAMTS-13 activity was lower in patients with AF-related stroke compared to patients with ESUS (p = 0.0227), which was, however, due to confounding by vascular risk factors. ADAMTS-13 activity inversely correlated with the ESRS (r = −0.452, p < 0.001) and CHA2DS2VASC (r = −0.375, p < 0.001) in cohort 1. In accordance with these findings, we found a positive correlation between ADAMTS-13 activity and the RoPE score in cohort 2 (r = 0.413, p = 0.010). Conclusion ADAMTS-13 activity is inversely correlated with the number of vascular risk factors across different stroke etiologies. Further study is warranted to establish ADAMTS-13 as a mediator of cerebrovascular risk.
Article
Background and objectives: Anti-inflammatory therapies reduce major adverse cardiovascular events (MACE) in coronary artery disease but remain unproven after stroke. Establishing the subtype-specific association between inflammatory markers and recurrence risk is essential for optimal selection of patients in randomized trials (RCTs) of anti-inflammatory therapies for secondary stroke prevention. Methods: Using individual participant data (IPD) identified from a systematic review, we analyzed the association between high-sensitivity C-reactive protein, interleukin-6 (IL-6), and vascular recurrence after ischemic stroke or transient ischemic attack. The prespecified coprimary end points were (1) any recurrent MACE (first major coronary event, recurrent stroke, or vascular death) and (2) any recurrent stroke (ischemic, hemorrhagic, or unspecified) after sample measurement. Analyses were performed stratified by stroke mechanism, per quarter and per biomarker unit increase after loge transformation. We then did study-level meta-analysis with comparable published studies not providing IPD. Preferred Reporting Items for Systematic Review and Meta-Analyses IPD guidelines were followed. Results: IPD was obtained from 10 studies (8,420 patients). After adjustment for vascular risk factors and statins/antithrombotic therapy, IL-6 was associated with recurrent MACE in stroke caused by large artery atherosclerosis (LAA) (risk ratio [RR] 2.30, 95% CI 1.21-4.36, p = 0.01), stroke of undetermined cause (UND) (RR 1.78, 1.19-2.66, p = 0.005), and small vessel occlusion (SVO) (RR 1.71, 0.99-2.96, p = 0.053) (quarter 4 [Q4] vs quarter 1 [Q1]). No association was observed for stroke due to cardioembolism or other determined cause. Similar results were seen for recurrent stroke and when analyzed per loge unit increase for MACE (LAA, RR 1.26 [1.06-1.50], p = 0.009; SVO, RR 1.22 [1.01-1.47], p = 0.04; UND, RR 1.18 [1.04-1.34], p = 0.01). High-sensitivity CRP was associated with recurrent MACE in UND stroke only (Q4 vs Q1 RR 1.45 [1.04-2.03], p = 0.03). Findings were consistent on study-level meta-analysis of the IPD results with 2 other comparable studies (20,136 patients). Discussion: Our data provide new evidence for the selection of patients in future RCTs of anti-inflammatory therapy in stroke due to large artery atherosclerosis, small vessel occlusion, and undetermined etiology according to inflammatory marker profile.
Article
Full-text available
Background Disambiguation of embolus pathogenesis in embolic strokes is often a clinical challenge. One common source of embolic stroke is the carotid arteries, with emboli originating due to plaque buildup or perioperatively during revascularization procedures. Although it is commonly thought that thromboemboli from carotid sources travel to cerebral arteries ipsilaterally, there are existing reports of contralateral embolic events that complicate embolus source destination relationship for carotid sources. Here, we hypothesize that emboli from carotid sources can travel to contralateral hemispheres and that embolus interactions with collateral hemodynamics in the circle of Willis influence this process. Methods and Results We use a patient‐specific computational embolus‐hemodynamics interaction model developed in prior works to conduct an in silico experiment spanning 4 patient vascular models, 6 circle of Willis anastomosis variants, and 3 different thromboembolus sizes released from left and right carotid artery sites. This led to a total of 144 different experiments, estimating trajectories and distribution of approximately 1.728 million embolus samples. Across all cases considered, emboli from left and right carotid sources showed nonzero contralateral transport ( P value <−0.05). Contralateral movement revealed a size dependence, with smaller emboli traveling more contralaterally. Detailed analysis of embolus dynamics revealed that collateral flow routes in the circle of Willis played a role in routing emboli, and transhemispheric movement occurred through the anterior and posterior communicating arteries in the circle of Willis. Conclusions We generated quantitative data demonstrating the complex dynamics of finite size thromboembolus particles as they interact with pulsatile arterial hemodynamics and traverse the vascular network of the circle of Willis. This leads to a nonintuitive source‐destination relationship for emboli originating from carotid artery sites, and emboli from carotid sources can potentially travel to cerebral arteries on contralateral hemispheres.
Article
Full-text available
Currently, recombinant tissue plasminogen activator (rtPA) is an effective therapy for ischemic stroke (IS). However, blood–brain barrier (BBB) disruption is a serious side effect of rtPA therapy and may lead to patients’ death. The natural polyphenol apigenin has a good therapeutic effect on IS. Apigenin has potential BBB protection, but the mechanism by which it protects the BBB integrity is not clear. In this study, we used network pharmacology, bioinformatics, molecular docking and molecular dynamics simulation to reveal the mechanisms by which apigenin protects the BBB. Among the 146 targets of apigenin for the treatment of IS, 20 proteins were identified as core targets (e.g., MMP-9, TLR4, STAT3). Apigenin protects BBB integrity by inhibiting the activity of MMPs through anti-inflammation and anti-oxidative stress. These mechanisms included JAK/STAT, the toll-like receptor signaling pathway, and Nitrogen metabolism signaling pathways. The findings of this study contribute to a more comprehensive understanding of the mechanism of apigenin in the treatment of BBB disruption and provide ideas for the development of drugs to treat IS. Graphical abstract
Article
Background: Despite advances in secondary stroke prevention during the last several years, cryptogenic stroke remains associated with a high risk of recurrence. Studies have shown that the recurrence risk is higher in patients with large artery disease in which complex carotid plaques and carotid WEBs are identified. Methods: This is a case series of six patients with cryptogenic recurrent stroke in which conventional imaging and extensive workup did not identify an etiology. Intravascular optic coherence tomography (OCT) was performed using a ballon-guided flow-arrest technique to identify possible covert carotid lesions. Results: We present six cases in which, with the help of OCT, we identified three carotid WEBs with associated thrombosis and two ulcerated carotid artery plaques. Four patients were subsequently treated with endovascular stent placement without complications. OCT permitted the distinction between complicated carotid artery plaque and carotid WEB. Conclusion: Intravascular OCT is a feasible and safe approach to identifying patients with covert carotid wall abnormalities, like carotid WEBs and ulcerated plaques, that are amenable to carotid stenting to reduce recurrent stroke risk.
Article
Full-text available
Current guidelines for primary and secondary prevention of stroke in patients with carotid atherosclerosis are based on the quantification of the degree of stenosis and symptom status. Recent publications have demonstrated that plaque morphology and composition, independent of the degree of stenosis, are important in the risk stratification of carotid atherosclerotic disease. This finding raises the question as to whether current guidelines are adequate or if they should be updated with new evidence, including imaging for plaque phenotyping, risk stratification, and clinical decision-making in addition to the degree of stenosis. To further this discussion, this roadmap consensus article defines the limits of luminal imaging and highlights the current evidence supporting the role of plaque imaging. Furthermore, we identify gaps in current knowledge and suggest steps to generate high-quality evidence, to add relevant information to guidelines currently based on the quantification of stenosis.
Article
Atherosclerotic stenosis of the internal carotid artery is an important cause of stroke. The aim of this guideline is to analyse the evidence pertaining to medical, surgical and endovascular treatment of patients with carotid stenosis. These guidelines were developed based on the ESO standard operating procedure and followed the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) approach. The working group identified relevant questions, performed systematic reviews and meta-analyses of the literature, assessed the quality of the available evidence, and wrote recommendations. Based on moderate quality evidence, we recommend carotid endarterectomy (CEA) in patients with ≥60–99% asymptomatic carotid stenosis considered to be at increased risk of stroke on best medical treatment (BMT) alone. We also recommend CEA for patients with ≥70–99% symptomatic stenosis, and we suggest CEA for patients with 50–69% symptomatic stenosis. Based on high quality evidence, we recommend CEA should be performed early, ideally within two weeks of the last retinal or cerebral ischaemic event in patients with ≥50–99% symptomatic stenosis. Based on low quality evidence, carotid artery stenting (CAS) may be considered in patients < 70 years old with symptomatic ≥50–99% carotid stenosis. Several randomised trials supporting these recommendations were started decades ago, and BMT, CEA and CAS have evolved since. The results of another large trial comparing outcomes after CAS versus CEA in patients with asymptomatic stenosis are anticipated in the near future. Further trials are needed to reassess the benefits of carotid revascularisation in combination with modern BMT in subgroups of patients with carotid stenosis.
Article
Background and Purpose XO (xanthine oxidase) is a key enzyme of uric acid metabolism and is thought to contribute to oxidative pathways that promote atherosclerotic plaque progression, yet its role in plaque destabilization is not well elucidated. We hypothesized that XO is expressed in carotid plaque from symptomatic patients in association with cardiovascular risk factors. Methods Patients were stratified by symptoms, defined as presentation with an ipsilateral cerebral ischemic event. Carotid atherosclerotic plaques were obtained from 44 patients with symptomatic plaque and 44 patients without ischemic cerebral events. Protein expression of XO was evaluated by immunohistochemical staining and the percentage of cells expressing XO and CD68 (macrophage marker) compared between the groups. Biochemical and demographic cardiometabolic risk factors of study participants also were measured. Results Carotid atherosclerotic plaques from symptomatic patients were associated with significantly higher XO expression versus asymptomatic plaque (median [interquartile range]: 1.24 [2.09] versus 0.16 [0.34]; P <0.001) and with significantly higher circulating uric acid levels (mean±SD: 7.36±2.10 versus 5.37±1.79 mg/dL; P <0.001, respectively). In addition, XO expression in atherosclerotic carotid plaque was inversely associated with serum high-density lipoproteins cholesterol levels ( P =0.010, r =−0.30) and directly with circulating uric acid levels ( P <0.001, r =0.45). The average percentage of macrophages that expressed XO was significantly higher in symptomatic versus asymptomatic plaques (median [interquartile range]: 93.37% [25] versus 46.15% [21], respectively; P <0.001). Conclusions XO overexpression in macrophages is associated with increased serum uric acid and low high-density lipoproteins cholesterol levels and may potentially have a mechanistic role in carotid plaque destabilization. The current study supports a potential role for uric acid synthesis pathway as a target for management of carotid atherosclerosis in humans.
Article
Background and purpose We performed a systemic review and meta-analysis to elucidate the effectiveness and safety of dual antiplatelet (DAPT) therapy with P2Y12 inhibitors (clopidogrel/ticagrelor) and aspirin versus aspirin monotherapy in patients with mild ischemic stroke or high-risk transient ischemic attack. Methods Following Preferred Reported Items for Systematic Review and Meta-Analysis standards for meta-analyses, Medline, Embase, Cochrane Central Register of Controlled Trials, and the Cochrane Library were searched for randomized controlled trials that included patients with a diagnosis of an acute mild ischemic stroke or high-risk transient ischemic attack, intervention of DAPT therapy with clopidogrel/ticagrelor and aspirin versus aspirin alone from January 2012 to July 2020. The outcomes included subsequent stroke, all-cause mortality, cardiovascular death, hemorrhage (mild, moderate, or severe), and myocardial infarction. A DerSimonian-Laird random-effects model was used to estimate pooled risk ratio (RR) and corresponding 95% CI in R package meta. We assessed the heterogeneity of data across studies with use of the Cochran Q statistic and I ² test. Results Four eligible trials involving 21 493 participants were included in the meta-analysis. DAPT therapy started within 24 hours of symptom onset reduced the risk of stroke recurrence by 24% (RR, 0.76 [95% CI, 0.68–0.83], I ² =0%) but was not associated with a change in all-cause mortality (RR, 1.30 [95% CI, 0.90–1.89], I ² =0%), cardiovascular death (RR, 1.34 [95% CI, 0.56–3.17], I ² =0%), mild bleeding (RR, 1.25 [95% CI, 0.37–4.29], I ² =94%), or myocardial infarction (RR, 1.45 [95% CI, 0.62–3.39], I ² =0%). However, DAPT was associated with an increased risk of severe or moderate bleeding (RR, 2.17 [95% CI, 1.16–4.08], I ² =41%); further sensitivity tests found that the association was limited to trials with DAPT treatment duration over 21 days (RR, 2.86 [95% CI, 1.75–4.67], I ² =0%) or ticagrelor (RR, 2.17 [95% CI, 1.16–4.08], I ² =37%) but not within 21 days or clopidogrel. Conclusions In patients with noncardioembolic mild stroke or high-risk transient ischemic attack, DAPT with aspirin and clopidogrel/ticagrelor is more effective than aspirin alone for recurrent stroke prevention with a small absolute increase in the risk of severe or moderate bleeding.
Article
Background and purpose: Practice guidelines recommend that most patients receive moderate- or high-potency statins after ischemic stroke or transient ischemic attack (TIA) of atherosclerotic origin. We tested the association of different patterns of potency for prescribed statin therapy-assessed before admission and at hospital discharge for ischemic stroke or TIA-on mortality in a large, nationwide sample of US Veterans. Methods: The study population included patients with an ischemic stroke or TIA occurring during 2011 at any of the 134 Veterans Health Administration facilities. We used electronic outpatient pharmacy files to identify statin dose at hospital admission and within 7 days after hospital discharge. We categorized statin dosing as low, moderate, or high potency; moderate or high potency was considered at goal. We created 6 mutually exclusive groups to reflect patterns of statin potency from hospital admission to discharge: goal to goal, low to goal, goal to low or goal to none (deintensification), none to none, none to low, and low to low. We used logistic regression to compare 30-day and 1-year mortality across statin potency groups. Results: The population included 9380 predominately White (71.1%) men (96.3%) who were hospitalized for stroke or TIA. In this sample, 34.1% of patients (n=3194) were discharged off a statin medication. Deintensification occurred in 14.0% of patients (n=1312) and none to none in 20.5% (n=1924). Deintensification and none to none were associated with a higher odds of mortality as compared with goal to goal (adjusted odds ratio 1-year mortality: deintensification versus goal to goal, 1.26 [95% CI, 1.02-1.57]; none to none versus goal to goal, 1.59 [95% CI, 1.30-1.93]). Adjustments for differences in baseline characteristics using propensity weighted scores demonstrated similar results. Conclusions: Underutilization of statins, including no treatment or underdosing after stroke (deintensification), was observed in approximately one-third of veterans with ischemic stroke or TIA and was associated with higher mortality when compared with patients who were at goal for statin prescription dosing.
Article
Background IL-6 has emerged as a pivotal factor in atherothrombosis. Yet, the safety and efficacy of IL-6 inhibition among individuals at high atherosclerotic risk but without a systemic inflammatory disorder is unknown. We therefore addressed whether ziltivekimab, a fully human monoclonal antibody directed against the IL-6 ligand, safely and effectively reduces biomarkers of inflammation and thrombosis among patients with high cardiovascular risk. We focused on individuals with elevated high-sensitivity CRP and chronic kidney disease, a group with substantial unmet clinical need in whom previous studies in inflammation inhibition have shown efficacy for cardiovascular event reduction. Methods RESCUE is a randomised, double-blind, phase 2 trial done at 40 clinical sites in the USA. Inclusion criteria were age 18 years or older, moderate to severe chronic kidney disease, and high-sensitivity CRP of at least 2 mg/L. Participants were randomly allocated (1:1:1:1) to subcutaneous administration of placebo or ziltivekimab 7·5 mg, 15 mg, or 30 mg every 4 weeks up to 24 weeks. The primary outcome was percentage change from baseline in high-sensitivity CRP after 12 weeks of treatment with ziltivekimab compared with placebo, with additional biomarker and safety data collected over 24 weeks of treatment. Primary analyses were done in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of assigned treatment. The trial is registered with ClinicalTrials.gov, NCT03926117. Findings Between June 17, 2019, and Jan 14, 2020, 264 participants were enrolled into the trial, of whom 66 were randomly assigned to each of the four treatment groups. At 12 weeks after randomisation, median high-sensitivity CRP levels were reduced by 77% for the 7·5 mg group, 88% for the 15 mg group, and 92% for the 30 mg group compared with 4% for the placebo group. As such, the median pairwise differences in percentage change in high-sensitivity CRP between the ziltivekimab and placebo groups, after aligning for strata, were –66·2% for the 7·5 mg group, –77·7% for the 15 mg group, and –87·8% for the 30 mg group (all p<0·0001). Effects were stable over the 24-week treatment period. Dose-dependent reductions were also observed for fibrinogen, serum amyloid A, haptoglobin, secretory phospholipase A2, and lipoprotein(a). Ziltivekimab was well tolerated, did not affect the total cholesterol to HDL cholesterol ratio, and there were no serious injection-site reactions, sustained grade 3 or 4 neutropenia or thrombocytopenia. Interpretation Ziltivekimab markedly reduced biomarkers of inflammation and thrombosis relevant to atherosclerosis. On the basis of these data, a large-scale cardiovascular outcomes trial will investigate the effect of ziltivekimab in patients with chronic kidney disease, increased high-sensitivity CRP, and established cardiovascular disease. Funding Novo Nordisk.
Article
Interleukin-6 (IL-6) is a pivotal cytokine of innate immunity which enacts a broad set of physiologic functions traditionally associated with host defense, immune cell regulation, proliferation, and differentiation. Following recognition of innate immune pathways leading from the NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome to interleukin-1 to IL-6 and on to the hepatically derived clinical biomarker C-reactive protein, an expanding literature has led to understanding of the pro-atherogenic role for IL-6 in cardiovascular disease and thus the potential for interleukin-6 inhibition as a novel method for vascular protection. In this review, we provide an overview of the mechanisms by which IL-6 signaling occurs and how that impacts upon pharmacologic inhibition; describe murine models of IL-6 and atherogenesis; summarize human epidemiologic data outlining the utility of IL-6 as a biomarker of vascular risk; outline genetic data suggesting a causal role for IL-6 in systemic atherothrombosis and aneurysm formation; and then detail the potential role of IL-6 inhibition in stable coronary disease, acute coronary syndromes, heart failure, and the atherothrombotic complications associated with chronic kidney disease and end-stage renal failure. Finally, we review anti-inflammatory and anti-thrombotic findings for ziltivekimab, a novel IL-6 ligand inhibitor being developed specifically for use in atherosclerotic disease and poised to be tested formally in a large scale cardiovascular outcomes trial focused on individuals with chronic kidney disease and elevated levels of C-reactive protein, a population at high residual atherothrombotic risk, high residual inflammatory risk, and considerable unmet clinical need.
Article
Background and Purpose Unstable carotid plaques are a common cause of ischemic strokes. Identifying markers that reflect/contribute to plaque instability has become a prominent focus in cardiovascular research. The adipokines, resistin and chemerin, and ChemR23 (chemerin receptor), may play a role in carotid atherosclerosis, making them potential candidates to assess plaque instability. However, the expression and interrelationship of resistin and chemerin (and ChemR23) protein and mRNA within the carotid atherosclerotic plaque remains elusive. Thus, we investigated herein, the association between plaque mRNA and protein expression of resistin and chemerin (and ChemR23) and carotid plaque instability in humans, and whether sex differences exist in the relationship between these adipokines and plaque instability. Methods Human carotid plaques were processed for immunohistochemical/mRNA analysis of resistin, chemerin, and ChemR23. Plaque instability was assessed by gold-standard histological classifications. A semi-quantitative scoring system was used to determine the intensity of adipokine expression on macrophages/foam cells, as well as the percentage of inflammatory cells stained positive. Plaque adipokine protein expression was also digitally quantified and mRNA expression was assessed by qRT-PCR. Results Resistin and chemerin mRNA expression was 80% and 32% lower, respectively, in unstable versus stable plaques ( P <0.05), while no difference in ChemR23 mRNA expression was observed. In contrast, greater resistin staining intensity and percentage of cells stained positive were detected in unstable versus stable plaques ( P <0.01). Similarly, chemerin and ChemR23 staining intensity and percentage of cells stained were positively associated with plaque instability ( P <0.05). No strong sex-specific relationship was observed between adipokines and plaque instability. Conclusions This study examined the relationship between resistin, chemerin, and ChemR23, and carotid plaque instability, with a specific analysis at the plaque level. We reported a positive association between plaque instability and protein levels of resistin, chemerin, and ChemR23 but a negative association with resistin and chemerin mRNA expression. This suggests these adipokines exert proinflammatory roles in the process of carotid atherosclerosis and may be regulated via a negative feedback regulatory mechanism.