ArticlePDF Available

Multisociety Consensus Quality Improvement Revised Consensus Statement for Endovascular Therapy of Acute Ischemic Stroke

Authors:

Figures

Content may be subject to copyright.
Standards of Practice
Multisociety Consensus Quality
Improvement Revised Consensus
Statement for Endovascular Therapy
of Acute Ischemic Stroke
From the American Association of Neurological Surgeons
(AANS), American Society of Neuroradiology (ASNR),
Cardiovascular and Interventional Radiology Society of Europe
(CIRSE), Canadian Interventional Radiology Association (CIRA),
Congress of Neurological Surgeons (CNS), European Society of
Minimally Invasive Neurological Therapy (ESMINT), European
Society of Neuroradiology (ESNR), European Stroke Organization
(ESO), Society for Cardiovascular Angiography and Interventions
(SCAI), Society of Interventional Radiology (SIR), Society of
NeuroInterventional Surgery (SNIS), and World Stroke
Organization (WSO)
David Sacks MD, Blaise Baxter MD, Bruce C.V. Campbell MBBS, PhD,
Jeffrey S. Carpenter MD, Christophe Cognard MD PhD,
Diederik Dippel MD PhD, Muneer Eesa MD, Urs Fischer MD,
Klaus Hausegger MD, Joshua A. Hirsch MD,
Muhammad Shazam Hussain MD, Olav Jansen MD,
Mahesh V. Jayaraman MD, Alexander A. Khalessi MD MS,
Bryan W. Kluck DO, Sean Lavine MD, Philip M. Meyers MD,
Stephen Ramee MD, Daniel A. Ru
¨fenacht MD,
Clemens M. Schirmer MD PhD and Dierk Vorwerk MD,
From the Department of Interventional Radiology (D.S.), The Reading
Hospital and Medical Center, 6th and Spruce Sts., West Reading, PA
19612; Department of Radiology (B.B.), Erlanger Medical Center,
Chattanooga, Tennessee; Departments of Medicine and Neurology
(B.C.V.C.), Melbourne Brain Centre at the Royal Melbourne Hospital,
University of Melbourne, Parkville, Victoria, Australia; Department of
Radiology (J.S.C.), West Virginia University, Morgantown, West Virginia;
Department of Diagnostic and Therapeutic Neuroradiology (C.C.),
Centre Hospitalier Universitaire de Toulouse, Ho
ˆpital Purpan,
Toulouse, France; Department of Neurology (D.D.), Erasmus University
Medical Center, Rotterdam, The Netherlands; Department of Radiology
(M.E.), University of Calgary, Calgary, Alberta, Canada; Department of
Neurology (U.F.), Inselspital–Universita
¨tsspital Bern, Bern, Switzerland;
Department of Radiology (K.H.), Klagenfurt State Hospital, Klagenfurt
am Wo
¨rthersee, Austria; Neuroendovascular Program, Department of
Radiology (J.A.H.), Massachusetts General Hospital, Boston,
Massachusetts; Cerebrovascular Center, Neurological Institute (M.S.H.),
Cleveland Clinic, Cleveland, Ohio; Department of Radiology and
Neuroradiology (O.J.), Klinik fu¨r Radiologie und Neuroradiologie, Kiel,
Germany; Departments of Diagnostic Imaging, Neurology, and
Neurosurgery (M.V.J.), Warren Alpert School of Medicine at Brown
University, Rhode Island Hospital, Providence, Rhode Island;
Department of Surgery (A.A.K.)
University of California San Diego Health, San Diego, California;
Interventional Cardiology (B.W.K.), Heart Care Group, Allentown,
Pennsylvania; Departments of Neurological Surgery and Radiology
(S.L.), Columbia University Medical Center/New York–Presbyterian
Hospital, New York, New York; Departments of Radiology and
Neurological Surgery (P.M.M.), Columbia University College of
Physicians & Surgeons, New York, New York; Interventional
Cardiology, Heart & Vascular Institute (S.R.), Ochsner Medical Center,
New Orleans, Louisiana; Neuroradiology Division (D.A.R.), Swiss Neuro
Institute–Clinic Hirslanden, Zu¨ rich, Switzerland; Department of
Neurosurgery and Neuroscience Center (C.M.S.), Geisinger Health
System, Wilkes-Barre, Pennsylvania; and Diagnostic and Interventional
Radiology Institutes (D.V.), Klinikum Ingolstadt, Ingolstadt, Germany
This article was previously published in the Journal of Vascular and
Interventional Radiology. The articles are identical except for minor stylistic
and spelling differences in keeping with each journal’s style. The JVIR article
should be usedwhen citing this article ina journal. ßSociety of Interventional
Radiology, 2018 J Vasc Interv Radiol 2018; 29:441–453. https://doi.org/10.
1016/j.jvir.2017.11.026. Permissions: www.elsevier.com/permissions
Corresponding author:
David Sacks, Department of Interventional Radiology, The Reading
Hospital and Medical Center, 6th and Spruce Sts., West Reading, PA
19612.
Email: david.sacks@towerhealth.org
International Journal of Stroke, 0(0)
International Journal of Stroke
0(0) 1–21
DOI: 10.1177/1747493018778713
journals.sagepub.com/home/wso
Abbreviations
ASPECTS: Alberta Stroke Program Early Computed Tomography Score; EVT: endovascular therapy; mRS: modified
Rankin scale; mTICI: modified thrombolysis in cerebral infarction; NIHSS: National Institutes of Health Stroke Scale;
QI: quality improvement; SAH: subarachnoid hemorrhage; SICH: symptomatic intracranial hemorrhage; SITS-MOST: Safe
Implementation of Thrombolysis in Stroke Monitoring Study; TICI: thrombolysis in cerebral infarction: TIMI: thromb-
olysis in myocardial infarction; TPA: tissue plasminogen activator.
Received: 17 November 2017; accepted: 23 November 2017
Introduction
Endovascular therapy (EVT) for acute ischemic stroke
in selected patients has recently been proven effective in
several clinical trials, and the widespread adoption of
thrombectomy into routine clinical practice has begun.
However, these acute stroke services are resource-inten-
sive, including advanced cerebral imaging and highly
trained multidisciplinary hospital teams rapidly
responding to emergency activation. Despite the previ-
ous acceptance of intravenous fibrinolysis for acute
ischemic stroke and the development of designated
stroke centers (1), ischemic stroke remains a leading
cause of adult death and disability (2). Many patients
are not candidates for fibrinolysis, and intravenous
therapy is relatively ineffective for severe strokes as a
result of large cerebral artery occlusions. Moreover, it is
uncertain if the benefits of endovascular stroke treat-
ment in the trial setting can be generalized to clinical
care provided by hospitals and teams of varying train-
ing, experience, and case volume. In other medical dis-
ciplines, rapid technologic advancement required
guidelines to utilize these tools effectively and respon-
sibly (3). Quality-improvement (QI) metrics for the out-
comes of endovascular ischemic stroke treatment were
published by a multisociety, multispecialty, inter-
national consensus group in 2013 (4). These QI metrics
have been accepted at a national level in Great Britain
and Ireland (5) but have yet to be included into stroke
center accreditation requirements in the United States.
Subsequent to the publication of the prior QI guide-
lines, 8 randomized trials and several meta-analyses of
EVT have been published (6–20). These randomized
trials have established EVT as standard of care when
available (5,21–23), and provide additional data on
which to update the metrics and benchmarks of the
previous paper (4). Therefore, it is now appropriate to
revise the prior QI document based on new evidence.
Revision of this QI consensus statement remains
focused on processes of care and patient outcomes.
Other documents address standards for physician train-
ing (24,25) and recommendations for patient selection
and treatment methods (5,23). As in the previous guide-
lines, it is intended that these benchmarks be used in a
quality-improvement program to assess and improve
processes and outcomes in acute stroke revasculariza-
tion. The benchmarks provide the consensus process
and outcome consensus measures called for by the
Stroke Treatment Academic Industry Roundtable
(STAIR) IX academic industry roundtable for the
next generation of endovascular trials (26). The bench-
marks may also be suitable for accreditation of stroke
intervention programs. Most of the metrics apply to the
role of the interventional physician, regardless of spe-
cialty or particular board certification, but comprehen-
sive stroke care requires a broad multidisciplinary
process involving care that ranges from emergency dis-
patch of paramedics through acute hospital care and
posttreatment subacute rehabilitation. Therefore,
although it is not the intention of this document to
assess in detail the quality of facilities, some of the met-
rics also apply to institutional policies and procedures
for stroke care.
Materials and methods
A literature search was conducted using Ovid and
EMBASE from 2012 (from the last date of the litera-
ture search for the first publication of these metrics) (4)
to October 2015 using article titles that included the
following: (acute ischemic stroke OR cerebrovascular
accident OR stroke) AND (intra-arterial OR intraarter-
ial OR endovascular OR angioplasty OR stent OR
stent retriever OR mechanical thrombectomy OR
thrombolysis OR tissue plasminogen activator [TPA]
OR TPA OR urokinase OR streptokinase OR alteplase
OR tenecteplase). Additional articles were then soli-
cited from writing group members. An evidence table
(Table E1, available online at www.jvir.org) was con-
structed by using articles that were randomized con-
trolled trials, registries, or case series of at least 100
patients, and some case series of less than 100 patients
were included if the series provided uniquely useful
data. From the evidence table, metrics were chosen
that were believed to be important markers of quality
of care. Thresholds for metrics were then chosen by
consensus of the writing group based on review of the
evidence table. Consensus was defined as 80% of the
writing group. If consensus was not achieved during
International Journal of Stroke, 0(0)
2International Journal of Stroke 0(0)
discussion, a modified Delphi process was used to
obtain consensus (27). If consensus was not achieved
after the modified Delphi process, a threshold was not
chosen. The evidence table was then updated by using
the same search terms in February 2017 at the time of
completion of the draft of the document to allow
updating of the metrics if appropriate.
Standards for developing clinical practice guidelines
were reviewed (28). It was determined that the majority
of these standards were not applicable for this docu-
ment that updates quality benchmarks for processes
and outcomes of care rather than creating recommen-
dations for types of patient care. For this reason, this
revision has been changed to a consensus statement
rather than a guideline.
Definitions
Measures and metrics will depend on the definition of a
good outcome or a complication and the time at which
patients are assessed for these outcomes, as many
patients show gradual improvement following an ische-
mic stroke. Numerous trials have used varying defin-
itions for similar concepts. The definitions used in this
document were derived from review of these trials and
then consensus of the writing group.
Ischemic central nervous system infarction.—A
uniformly accepted simple definition of central nervous
system infarction remains elusive. A successful multi-
disciplinary attempt arrived at a definition as
follows (29):
Central nervous system infarction is defined as brain,
spinal cord, or retinal cell death due to ischemia,
based on:
1. Pathological, imaging, or other objective evidence of
cerebral, spinal cord, or retinal focal ischemic injury
in a defined vascular distribution; or
2. Clinical evidence of cerebral, spinal cord, or retinal
focal ischemic injury based on symptoms persisting
at least 24 hours or until death, and other etiologies
excluded.
Door-to-event time.—The term ‘‘door’’ is used to
determine the time of onset of medical care, as in
‘‘door to time of computed tomography (CT) ima-
ging.’’ It is defined as the time of arrival in the emer-
gency department for an outpatient or the time first
discovered to have a stroke for an inpatient. When
patients are transferred, ‘‘door’’ refers to the arrival
(ie, registration) time at the receiving facility.
Time to thrombus.—Time to thrombus is considered
to represent the start of endovascular lytic infusion or
first placement of a mechanical device in the target
vessel.
Successful revascularization.—Successful revascular-
ization is considered to represent modified thromboly-
sis in cerebral infarction (mTICI) (30,31) grade 2b or 3
flow through the previously occluded vessel segment
(Table 1).
Symptomatic intracranial hemorrhage.—
Symptomatic intracranial hemorrhage (SICH) is a par-
enchymal hematoma type II (per the Safe
Implementation of Thrombolysis in Stroke
Monitoring Study [SITS-MOST] definition) (32) or
subarachnoid hemorrhage (SAH) with neurologic
deterioration leading to an increase in National
Institutes of Health Stroke Scale (NIHSS) score >4
or leading to death within 36 hours of treatment.
Because of the risk of vessel perforation during endo-
vascular procedures, SAH has been added as a cause of
intracranial hemorrhage to the SITS-MOST SICH def-
inition (33).
This definition is similar to that used in the recent
randomized trials of EVT (7,11,15). Several of the
authors of those trials have joined others in proposing
Table 1. mTICI Revascularization Scale Scores (30,31,113)
Score Description
0 No perfusion, complete obstruction; no flow past occlusion of ‘‘major’’ vessel
1 Perfusion past initial obstruction but limited distal branch filling with little/slow distal perfusion
2a Partial perfusion: <50% of ‘‘major’’ vascular territory perfused (eg, filling and complete perfusion through one
M2 division)
2b Partial perfusion: 50% of major vascular territory is filled, but there is not complete and normal perfusion of
entire territory
3 Complete or full perfusion with filling of all distal branches
mTICI: modified thrombolysis in cerebral infarction.
International Journal of Stroke, 0(0)
Sacks et al. 3
a new definition of SICH (34). These new definitions
have not yet been validated on a larger scale, adopted in
stroke trials, or applied to the outcomes of the recent
randomized trials. Therefore, the original definition of
SICH is maintained in the present revision of the con-
sensus statement and modified to include any intracra-
nial hemorrhage associated with a decrease in NIHSS
score >4 or death within 24 hours of the end of the
revascularization procedure (20).
Good clinical outcome.—A good clinical outcome is a
measure of neurologic functional with a score of 0–2 on
the modified Rankin scale (mRS; Table 2) (35) assessed
90 days after treatment. This does not exclude clinically
significant benefit in patients in whom an mRS score of
2 is not achieved.
Indications and Contraindications
EVT for acute ischemic stroke with large vessel occlu-
sion is established in guidelines as the standard of care
(22,36). If the patient is also eligible for intravenous
TPA, this drug should be administered as a ‘‘bridging’’
strategy in parallel without delaying thrombectomy.
Waiting to assess ‘‘response’’ to TPA is strongly dis-
couraged (22), as clinical improvement may not indi-
cate recanalization. The rate of TPA-induced
recanalization before thrombectomy (performed with-
out delay) was <10% in recent randomized trials
(11,13,20). Proceeding directly to thrombectomy (ie,
direct thrombectomy) should be performed in appro-
priate candidates with a contraindication to TPA,
including risk of hemorrhage or when >4.5 hours
have elapsed since stroke onset.
Indications and contraindications for EVT are based
on subgroup analyses of randomized trials and case
series. Clinical trials tend to have more restrictive cri-
teria, whereas case series represent more of a ‘‘real-
world’’ experience. Potential selection criteria are
based on stroke severity, time (ie, duration of symp-
toms), imaging, clot location, age, and comorbidities.
Stroke severity.—Clinical trials have set variable
NIHSS score limits for eligibility, often requiring 6,
8, or 10 points. The Multicenter Randomized Clinical
Trial of Endovascular Treatment for Acute Ischemic
Stroke in the Netherlands (MR CLEAN) trial had a
minimum NIHSS score of 2 and Extending the Time
for Thrombolysis in Emergency Neurological Deficits-
Intra-arterial (EXTEND-IA) had no NIHSS score
limits, but, given the requirement for large vessel occlu-
sion, few patients with NIHSS scores <6 were enrolled
(37). Individual patient data meta-analysis of five posi-
tive randomized trials (14) demonstrated highly consist-
ent treatment effects across the NIHSS score spectrum,
at least for NIHSS scores 6. Data from observational
studies have demonstrated an important incidence of
large vessel occlusion in patients with clinically mild
stroke and a propensity for these patients to later
experience neurologic deterioration (38). The risk/bene-
fit in patients with low NIHSS scores therefore needs to
be carefully considered, and future studies have to
address whether endovascular procedures are beneficial
in patients with mild symptoms and proximal vessel
occlusion. There are no data supporting an upper
limit on stroke severity.
Time.—Most trials of intraarterial lytic agents and
mechanical revascularization devices have historically
required start of treatment within 6 or 8 hours (39–
42) for anterior-circulation strokes. The strongest evi-
dence for EVT is for treatment commenced within 6
hours (14,43). More rapid time to reperfusion has
been linked to improved clinical outcomes and is there-
fore an important consideration in patient selection
(43–45). A few patients in recent trials were treated at
6–8 hours in the Randomized Trial of
Revascularization with Solitaire FR Device versus
Best Medical Therapy in the Treatment of Acute
Table 2. mRS Scores (35)
Score Description
0 No symptoms
1 No significant disability: able to carry out all usual activities despite some symptoms
2 Slight disability: able to look after own affairs without assistance but unable to carry out all previous activities
3 Moderate disability: requires some help but able to walk unassisted
4 Moderately severe disability: unable to attend to own bodily needs without assistance and unable to walk unassisted
5 Severe disability: requires constant nursing care and attention, bedridden, incontinent
6 Dead
mRS: modified Rankin scale.
International Journal of Stroke, 0(0)
4International Journal of Stroke 0(0)
Stroke Due to Anterior Circulation Large Vessel
Occlusion Presenting within Eight Hours of Symptom
Onset (REVASCAT) trial (15) and at 6–12 hours in the
Endovascular Treatment for Small Core and Anterior
Circulation Proximal Occlusion with Emphasis on
Minimizing CT to Recanalization Times (ESCAPE)
trial (13). Individual patient data meta-analysis sug-
gests significant benefit to at least 7 hours, 18 minutes
(46). Observational studies have suggested that patients
presenting at later time points with favorable imaging
findings still benefit from reperfusion (47), and this was
confirmed in the DWI or CTP Assessment with Clinical
Mismatch in the Triage of Wake-Up and Late
Presenting Strokes Undergoing Neurointervention
with Trevo (DAWN) trial (48), which used clinical–
core mismatch criteria to select patients 6–24 hours
after the ‘‘last known well’’ time. In the DAWN trial
(48), independent functional outcome occurred in
48.6% of patients who underwent endovascular treat-
ment versus 13.1% of control patients (P<.0001) with
similar revascularization success as 0–6-hour thrombec-
tomy trials and no variation in treatment effect between
the 6–12-hour and 12–24-hour treatment windows.
Other randomized trials in extended time windows are
ongoing (49,50). Vertebrobasilar occlusions have been
treated at extended times, sometimes more than 24–48
hours after symptom onset (51,52). This is partly
because of the traditional definition of onset as the
last known well time. Patients with basilar artery occlu-
sion may have prodromal mild symptoms in 60% of
cases before the development of severe deficits (53).
The Basilar Artery International Cooperation Study
(BASICS) registry (53) advocated using time of severe
deficit (ie, likely moment of occlusion) and found
that good outcome with reperfusion beyond 9 hours
of that time was extraordinarily rare (53).
Randomized trials in patients with basilar artery occlu-
sion are ongoing (54,55).
Imaging.—Noncontrast CT has been an essential
component of patient selection in randomized trials of
intravenous and endovascular revascularization for
treatment of acute stroke (1,7,11,13,15,20,39–41,56).
Absolute noncontrast CT contraindications to endo-
vascular treatment are similar to those for intravenous
thrombolytic agents and include the presence of acute
intracranial hemorrhage or a significant established
infarct (1).
Infarct size can be approximated on noncontrast CT
by using the Alberta Stroke Program Early CT Score
(ASPECTS) (57,58). However, the score is not closely
related to infarct volume or functional eloquence and
has variable interrater agreement, particularly early
after stroke onset. In recent randomized trials, there
was clear benefit in patients with ASPECTS 6–8 and
9/10. Relatively few patients with ASPECTS 0–5 were
included in the trials. The benefit in this group
appeared to be of lesser magnitude, but a clinically
meaningful benefit could not be excluded (14).
Patients with ASPECTS 3–5 will be evaluated in a ran-
domized trial (59).
The hyperdense middle cerebral artery sign can alert
clinicians to the presence of a large vessel occlusion.
This sign has a high degree of sensitivity if thin (1-
mm) slices are reconstructed and good specificity if
clearly asymmetric compared with the contralateral
artery (60). Clot length on noncontrast CT of more
than 8 mm has been associated with lower recanaliza-
tion rates after intravenous TPA (61), but this is not
absolute (62), and none of the positive randomized
trials considered clot length in determining eligibility.
The Randomized, Concurrent Controlled Trial to
Assess the Penumbra System’s Safety and
Effectiveness in the Treatment of Acute Stroke
(THERAPY) trial that used this criterion was neutral
(18). There is evidence that occult anterograde flow can
be associated with TPA-induced recanalization even in
the presence of a long thrombus (63).
The target vessel occlusion should be established by
using noninvasive angiography (CT or magnetic reson-
ance [MR] imaging), as practiced in all the positive
randomized trials. This also provides information on
proximal arterial pathology and catheter access. CT
angiography has also been used to grade the quality
of collateral flow. However, there is potential for stand-
ard single early-phase acquisitions to underestimate
late-arriving collateral flow and therefore exclude
patients who may benefit. Dynamic angiography
derived from CT perfusion or multiphase CT angiog-
raphy acquisitions avoids this pitfall (64).
Many centers use CT perfusion to improve diagnos-
tic sensitivity and provide an estimate of tissue viability,
which is closely related to the quality of collateral blood
flow. A large volume of ischemic core (eg, >70 mL) on
CT perfusion is certainly associated with a worse prog-
nosis, but whether this alters treatment effect within 6
hours of stroke onset is yet to be clarified. Some case
series have suggested a benefit of reperfusion even in
patients with a large ischemic core >100 mL (65).
Analysis of the MR CLEAN trial did not reveal treat-
ment effect heterogeneity between cases of <70 and
>70 mL core, although the absolute probability of
independent functional outcome in those with a core
>70 mL was only 8% (66). Rather than excluding
patients from treatment as a result of a large ischemic
core, the presence of favorable imaging may be useful
in deciding to pursue treatment in patients with other-
wise less favorable clinical characteristics. Estimation
of ischemic core volume by using CT perfusion com-
bined with age and NIHSS score in clinical–core mis-
match was shown to identify patients who benefit from
International Journal of Stroke, 0(0)
Sacks et al. 5
thrombectomy in the extended time window of 6–24
hours in the DAWN trial (48).
MR imaging with diffusion imaging, with or without
perfusion imaging, is increasingly used in some centers.
There are some logistic challenges of safety screening
and rapid access to MR scanners that have to be over-
come to avoid relevant delays in treatment. However, in
the high-performing centers in the Solitaire With
the Intention For Thrombectomy as PRIMary
Endovascular treatment (SWIFT PRIME trial) (44),
there was no significant difference in arrival to random-
ization time according to image modality (ie, CT vs
MR imaging), suggesting that MR-related delay is not
inevitable. Uncertainties regarding whether core
volume is truly treatment effect–modifying or simply
prognostic apply, as discussed with CT perfusion (65).
It is also important to note that measured perfusion
lesion volumes vary between processing software pro-
grams and the thresholds used to estimate ischemic core
may vary with time (67).
Clot location.—The randomized trials demon-
strated clear benefit in internal carotid artery ter-
minus and M1 (ie, first segment of middle cerebral
artery) occlusion, with or without tandem occlusion
in the cervical carotid artery (10,14). Arterial occlu-
sions arising more proximally are associated with
poorer outcomes. Most notably, ‘‘T-lesions’’ have
the poorest outcomes among anterior-circulation
strokes (68,69). Proximal M1 occlusions have worse
outcomes than distal M1 occlusions as a result of
occlusion of lenticulostriate arteries and basal ganglia
infarction, with an increased risk of reperfusion hem-
orrhage (70). More distal M2 occlusions were less
common among trial patients, and a clear benefit
was not demonstrated, although there was no signifi-
cant heterogeneity in treatment effect observed. Many
patients with M2 occlusions were assessed as having
M1 occlusions at the site and reclassified as having
M2 occlusions by the core laboratory, leading to a
predominance of larger, more proximal occlusions.
Some case-control studies have suggested that benefit
persists in M2 occlusions, with similar safety as M1
occlusions (71). Basilar artery occlusion was not
included in the recent trials, in some cases because
of perceived lack of equipoise and in others because
of concerns regarding excessive heterogeneity. The
BASICS trial (54) is ongoing, but many sites regard
the dismal prognosis if untreated and the clear
improvement associated with recanalization as suffi-
cient grounds to treat. EVT for occlusions in the
anterior cerebral artery, M3/4 segments, and poster-
ior cerebral artery has not been systematically stu-
died. More distal vessels are smaller and more
tortuous, which potentially increases procedural risk,
and the smaller territory at risk and increased efficacy
of TPA reduces the benefit. Further device develop-
ment may alter this balance in the future.
Age.—Although increased age is associated with a
worse prognosis after stroke in general, the recent
trials have clearly demonstrated a treatment effect in
patients aged >80 years of at least the same magnitude
as in younger patients. Indeed, there is a significant
mortality benefit in elderly patients, with 20% absolute
risk reduction (number needed to treat ¼5) (10,14).
Importantly, the trials included only patients with inde-
pendent premorbid function, regardless of age, and the
potential quality-of-life benefit for patients with signifi-
cant comorbidities needs to be weighed in clinical prac-
tice. Prestroke dementia before endovascular
reperfusion has been linked with a low probability of
achieving a good clinical outcome (72). Some trials
have therefore excluded patients aged >80 years (73).
Older patients may also have tortuous arterial access,
which can complicate the procedure.
Medical comorbidities.—Most contraindications to
intravenous thrombolysis do not apply to EVT.
Overall, mechanical thrombectomy (with or without
intravenous TPA) has a similar risk of SICH compared
with TPA alone (10,14). There are relatively limited
data on the safety of EVT in patients with markedly
abnormal coagulation (eg, International Normalized
Ratio >3.0 or current use of novel or direct oral anti-
coagulant agents), and risks and benefits need to be
considered on an individual basis.
The criteria chosen to select patients for treatment
will affect outcomes. Patients at higher risk are more
likely to do poorly with or without treatment, but selec-
tion of only patients at low risk will deny clinical benefit
to a large number of severely ill patients. Because pub-
lished selection criteria vary, there is no single ‘‘correct’’
list of inclusion and exclusion criteria. The American
Heart Association has published class I recommenda-
tions for EVT patient selection (22), but 40%–50% of
patients are now being treated outside of these class I
recommendations (74,75). Based on published data and
the desired ratio of benefit to risk, each institution will
need to create and follow its own indications and
contraindications.
Metric 1: At least 90% of patients who meet the insti-
tutional selection criteria (ie, indications/contraindica-
tions) should be treated with endovascular therapy.
Process and outcomes metrics
In general, previously published endovascular stroke
therapy metrics (76) were designed to measure aggre-
gate performance of hospital or clinical outcomes. They
were neither designed nor intended to define individual
International Journal of Stroke, 0(0)
6International Journal of Stroke 0(0)
physician performance. In contrast, this document pro-
vides requirements for performance criteria for the indi-
vidual practitioner and the facility. The purpose of
these metrics is to define the minimum standards for
EVT in acute ischemic stroke patients. It is recognized
that a concerted team effort is required to ensure effi-
cient workflow, timely EVT, and safe, effective care.
The recent endovascular trials have reiterated the
importance of appropriate patient selection and pro-
cedural performance such as timely and more complete
revascularization to improve the likelihood of achieving
a good clinical outcome. This paradigm is based on
selecting patients with potentially salvageable ischemic
penumbra. A noncontrast head CT/MR study and vas-
cular imaging such as CT/MR angiography will dem-
onstrate areas of established infarct and presence of a
proximal large vessel occlusion, respectively, and pro-
vide vital information to select patients for endovascu-
lar therapy.
Data collection
From a quality-assurance perspective, endovascular
therapy for acute ischemic stroke differs slightly from
other areas in which quality initiatives, morbidity, and
mortality discussions focus on specific events in which
errors in care or complications occurred. The measure
of benefit from endovascular stroke therapy is not
based on single or isolated cases, but rather is expressed
as a percentage of aggregated patients treated who can
function independently at 3 months. This has also been
measured by using shift analysis in the recent endovas-
cular trials (77). As demonstrated in those trials, clinical
benefit from EVT is dependent on delivery of high-
quality care in a timely manner at the institutional
level by a dedicated team.
As such, performance metrics from large aggregates
of patients treated by endovascular means are com-
pared versus performance standards in clinical trials
in which benefits were demonstrated, recognizing that
patients may be treated outside trial inclusion criteria
on a case-by-case basis. This requires all patients’ pro-
cedural, process, and clinical outcomes to be entered
into a database, trial, or registry (24,76,78–80).
Without the denominator of ‘‘all patients,’’ measures
of success and percentage descriptors are
meaningless. These data allow comparison of metrics
against benchmarks for individual operator perform-
ance, risk-adjusted clinical outcomes, and individual
and institutional process measures.
As stated in a prior document concerning
Comprehensive Stroke Centers (76), it is advantageous
to collect data in a standardized fashion to avoid redun-
dant efforts. Data collection tools such as multicenter
registries will serve as useful benchmarks and will
facilitate an ongoing process of constant evaluation.
Multicenter registries are recommended over institu-
tional registries because of the ability to serve as a
benchmark against other institutions. There are numer-
ous examples of such data-collection tools for treat-
ment of acute ischemic stroke (78,80–82). A recent
publication looking at trends in endovascular therapy
and clinical outcomes within the Get With The
Guidelines–Stroke registry (83) demonstrates the utility
of strict data collection within well-maintained nation-
wide database systems.
Data collection for EVT is closely tied in with the
process already in place for patients who are eligible
for intravenous thrombolytic therapy and starts with
documentation of time of onset and the time the patient
arrives at the ‘‘door.’’ This could mean (i) the door of a
primary stroke center, where data collection should ide-
ally start, or, (ii) in cases being transferred to an endo-
vascular center, the time of registration at the center
that receives the patient. The detailed time metrics will
be discussed in the next section. Data collection, espe-
cially time points, should be as inclusive as possible,
with subsequent metrics being reported by combining
multiple elements. The mandatory threshold for collec-
tion of the minimum defined elements is 100%.
Data concerning demographic characteristics are
used to identify various patient subgroups, whereas
other data points are pertinent for risk adjustment and
are necessary for evaluation of procedural and clinical
outcomes. These would include factors specific to the
individual case, such as location of occlusion and time
from onset, as well as demographic factors specific to
patient subgroups, such as age, race, and sex. Ancillary
data such as prognostic factors pertaining to comorbid-
ity, stroke severity, and imaging parameters may help in
risk- and severity-adjusted analysis to adjust for vari-
ability in case mix. Collection of these data points is
necessary for an appropriate evaluation of patient risk
factors and also for study of institutional factors that
could influence overall patient outcomes and have a
bearing on evaluation of operator performance.
At a minimum, these data should include age, sex,
premorbid mRS score, NIHSS score, location of occlu-
sion, various time points and intervals described in the
subsequent sections, blood pressure, blood glucose level
at presentation, and presence of atrial fibrillation.
Specific data-collection metrics for EVT have already
been included in national guidelines (22,23,36). Other
data elements may be helpful and may become evident
with further research, such as radiation exposure and
contrast agent dose.
Metric 2: 100% of patients have the required minimum
process and outcomes data entered into an institutional
or national database, trial, or registry.
International Journal of Stroke, 0(0)
Sacks et al. 7
Time intervals
Emergency endovascular stroke treatment is one of the
most complex multidisciplinary functions a medical
institution chooses to undertake. Reperfusion treat-
ment (intravenous or endovascular) achieved within
the shortest period of time is widely accepted as a pre-
requisite for optimal clinical outcomes (45,84,85).
Subgroup analyses from several trials (43,86,87)
have shown that treatment delays resulted in significant
decrease in the likelihood of a good outcome of endo-
vascular stroke therapy. Analysis of the pooled data of
5 endovascular trials (46) confirmed this: every 1-hour
delay in time from onset to arterial puncture results in a
5.3% shift in the direction of more disability on
the mRS.
There are many steps from stroke onset to comple-
tion of treatment, and optimal and timely execution of
each of these steps is necessary to achieve the stated
goal. Numerous opportunities exist to minimize the
time needed for each step from the time of the acute
stroke to patient arrival to the hospital and then until
reperfusion is achieved.
Process improvement for emergency stroke treat-
ment should be an ongoing component of all stroke
systems of care and should focus on all the tasks and
activities in this complex sequence of events. These data
are then used for quality assessment/assurance and pro-
cess improvement and therefore directly relate to the
eventual clinical outcome of the patients being treated
by the team. To judge satisfaction of these performance
goals in regard to expeditious delivery of care, time
points and intervals are the units of measurement.
At a minimum, the time points and intervals speci-
fied in this document should be tracked in all cases.
Institutions may choose to measure additional time
points. The more time points that are recorded, the
more exactly deficiencies might be identified; however,
this may prove onerous to document from a resource
perspective. For instance, delays in obtaining a CT scan
may result from delay in ordering the study, delay in
response by CT staff (eg, multiple other procedures
being requested at the same time), or delay related to
transportation.
Acknowledgment of the critical importance of time to
reperfusion for obtaining favorable outcomes in myo-
cardial reperfusion treatments has led to the formation
of initiatives such as ‘‘Door to Balloon: An Alliance of
Quality’’ for patients with ST-segment elevation myo-
cardial infarction. The key was achievement of a door-
to-balloon time of <90 minutes for at least 75% of
patients presenting directly to the treating hospital by
using various strategies identified through research,
resulting in dramatic reductions in times (88,89).
The impressive results in shortening the time to myo-
cardial reperfusion for acute myocardial infarction
obtained by such initiatives provided an impetus for
launching similar initiatives related to intravenous
TPA for stroke (90). The Joint Commission has set a
more ambitious goal of 80% of patients treated within
1 hour for primary stroke centers (91). The experience
in reducing door-to-needle times reported by the group
from Helsinki (92) suggests that, with simple strategies,
median door-to-needle times of 30 minutes or even less
can be achieved. Because of the need for neurologic
assessment and imaging in addition to the emergency
medicine and interventional components, acute stroke
patients referred for EVT require more time for initi-
ation of treatment than patients with ST-segment ele-
vation myocardial infarction. Although rapid-response
mechanisms aiming to result in initiation of revascular-
ization therapies within the minimum amount of time
can be modeled according to the myocardial infarction
experience, it should be recognized that acute stroke
treatment, especially EVT, requires a far more complex
infrastructure. Notwithstanding that, it is clear that,
similar to the cardiology model, major improvements
in door-to-treatment time need to take place to increase
the proportion of favorable outcomes for patients trea-
ted with EVT for acute stroke (93).
Since the early years of endovascular stroke treat-
ment, various time metrics have been reported, with a
trend toward overall improvement in times. These were
initially reported on the basis of case series (94,95), with
newer metrics from registries (96,97), earlier device
trials (98,99), and recent randomized controlled trials
(10). These reports focused on median onset–to–groin
puncture times ranging from 200 minutes in the latest
randomized trials (10) to 277 minutes in registry data
(97). Recent trial data (10) have also reported various
components of these times, breaking them down into
intervals that include patient arrival times and imaging
times. In the ESCAPE trial (13), the authors reported a
median time from imaging to arterial puncture of 51
minutes and a median time from imaging to reperfusion
of 84 minutes. The median imaging-to-puncture time in
the SWIFT PRIME trial (20) was 57 minutes. The
Highly Effective Reperfusion evaluated in Multiple
Endovascular Stroke trials (HERMES) meta-analysis
of treatment times from 5 recent large endovascular
trials (46) reported better clinical outcomes with faster
treatment times, with median door-to-imaging time of
19 minutes, imaging-to-puncture time of 76 minutes,
and puncture-to-reperfusion time of 44 minutes in the
entire cohort.
The endovascular trials represent optimal results
based on study site and patient selection. Many of the
endovascular trials included only study sites with a
proven ability to respond rapidly, excluded patients
with carotid dissections or internal carotid artery–
origin occlusions, and excluded patients who could
International Journal of Stroke, 0(0)
8International Journal of Stroke 0(0)
not be treated rapidly. However, the reported times in
recent trials did include time taken for patient random-
ization. These rapid responses have not been uniformly
achieved in other trials, registries, or case series
(18,100–102). Nevertheless, the time intervals in this
consensus statement are intended to be achievable
with good practice as centers become proficient at rou-
tinely performing endovascular therapies, and provide
a benchmark for QI in current clinical practice.
Moreover, many of the conditions and findings that,
in the past, could have complicated decision-making,
such as older age, extracranial carotid obstruction,
vessel tortuosity, requirement for penumbral imaging,
and requirement for general anesthesia, did not nega-
tively influence treatment effect and should not delay
the decision for treatment. The treatment of more com-
plex cases than were included in the trials may prolong
treatment times but should not prolong the time to
arterial puncture.
As a general approach to setting metrics for care
processes, we used data from the HERMES collabor-
ation (46). The 75th-percentile times (ie, slowest quar-
tile) are considered minimum benchmarks, and the
25th-percentile times (ie, fastest quartile) from that
study are considered achievable by the best centers
with high volumes and good resource infrastructure.
The metrics are intended to be used for measurements
such that centers will progressively become faster and
improve times from minimum acceptable to ideal.
The times reported in the following sections apply to
anterior circulation occlusions, as vertebrobasilar
occlusions were excluded in the recent randomized
trials. These metrics should be applicable regardless
of the time of the day and regardless of whether the
patient presents on a weekday versus a weekend (103).
These metrics represent maximum recommended times.
Because of ample evidence that, the shorter the time to
reperfusion, the higher the likelihood of a favorable
outcome, all centers should strive to initiate endovas-
cular therapy within the shortest possible time frame.
Although intravenous TPA administration should not
represent a justification for excessive delays in initiation
of endovascular therapy, it is acknowledged that intra-
venous thrombolysis may be associated with some
delays in initiation of endovascular therapy.
Door to imaging.—Most hospitals will use CT-based
imaging, but some hospital protocols may use MR ima-
ging as the first imaging study. The use of CT angiog-
raphy or MR angiography for vascular imaging is
considered the standard of care for endovascular treat-
ment based on recent trials and should be incorporated
into the imaging protocol. Indeed, previously published
guidelines on imaging in acute stroke patients (104)
recommend that noninvasive vascular imaging be rou-
tinely performed, and it is recognized that the use of
advanced multimodal imaging does not delay treatment
times (105). Regardless of the choice of modality based
on institutional preferences, imaging should be started as
quickly as feasible. Because of the difficulty in defining
exactly when an order might have been entered in the
system, this document is in agreement with the
American Stroke Association recommendations that
these time intervals be measured from arrival to start of
imaging, which will also include vascular imaging.
Interpretation of imaging is done in parallel and usually
at the scanner by the treating team, and the time needed
to interpret the scans and make a decision will be part of
the overall time from the start of imaging to arterial
puncture. In the HERMES meta-analysis (46), the fastest
25% of cases had imaging initiated by 12 minutes, and
75% of patients had imaging initiated within 30 minutes.
Metric 3: 75% of patients being evaluated for revascu-
larization should have imaging initiated within 30 min-
utes from time of arrival. At the best of centers with
high volumes and an established resource infrastruc-
ture, this is expected to be achieved in 12 minutes.
Imaging to puncture.—The largest amount of time
from door to revascularization comes from the steps
from door to puncture rather than puncture to revas-
cularization, and most endovascular treatment deci-
sions are made after imaging. Therefore, the largest
opportunities to reduce delays and improve outcomes
will come from reducing imaging-to-puncture times.
The recommended time from start of imaging to arter-
ial puncture is 50 minutes or less. This is in keeping
with the time intervals reported in the recent endovas-
cular trials, which had a fastest 25th percentile of 51
minutes (46), and it is the consensus of the writing
group that this time metric is necessary, achievable,
and consistent with the improvement in door-to-bal-
loon times that have been achieved for acute myocar-
dial infarction. The recent trials also reported that 75%
of patients had an imaging-to-puncture time of no more
than 110 minutes. For patients transferred from
another site whose imaging does not need to be
repeated, it is expected that door-to-puncture times
can be reduced by 30 minutes.
Metric 4: 75% of patients treated with endovascular
therapy should have an imaging-to-puncture time of
110 minutes or less. At the best of centers with high
volumes and an established resource infrastructure, this
is expected to be achieved in 50 minutes or less.
Metric 5: For patients transferred from another site in
whom imaging is not repeated, 75% of patients being
treated should have a door-to-puncture time of 80 min-
utes or less.
International Journal of Stroke, 0(0)
Sacks et al. 9
Imaging-to-thrombus time.—Previous versions of this
document have included imaging-to-thrombus time as a
metric. This is no longer believed to be a necessary time
point for measurement as a result of inaccuracies of
measurement and inconsistent practice in documenting
the same.
Puncture time to reperfusion.—This metric assesses
the efficiency of the interventional physician and
team. Given the rapid advancements in endovascular
treatment modalities, these recommendations are
likely to change. In the Mechanical Embolus
Removal in Cerebral Ischemia registry (41), the largest
prospective endovascular database to date reflecting
procedural outcomes across a large variety of stroke
centers in the United States, the median time from
groin puncture to the end of the procedure was 90 min-
utes. Newer technologies such as ‘‘stentrievers’’ have
been noted to achieve significantly shorter procedural
times (median of approximately 50 min) (46).
Although time to final angiography is easily mea-
sured, it may be variable depending on the need to per-
form thrombolysis of peripheral-branch occlusions
after recanalization of the proximal occlusion, as
more complete revascularization is likely to lead to
improved clinical outcomes, albeit at some increased
procedural risk. The time metric described here for suc-
cessful reperfusion represents the time to first reach an
mTICI grade 2b. Additional time, if required to
achieve complete revascularization, ie, mTICI grade
3, is not reflected in this metric. Recent trials have pub-
lished their time intervals, and, by doing so, set new
expectations. Median time from groin puncture to
reperfusion in the SWIFT PRIME trial (20) was 24
minutes (interquartile range, 18–33 min). The median
puncture-to-reperfusion time in the HERMES collab-
oration (46) was 44 minutes (interquartile range, 27–
64.5 min). Generally, we recommend that procedure
times not exceed 60 minutes as in the recent trials,
and the reperfusion target should be to reach mTICI
grade 2b (20). This threshold is further clarified in the
following section on recanalization/reperfusion.
Metric 6: In 70% of patients, mTICI grade 2b should
be reached ideally within 60 minutes of arterial
puncture.
Recanalization/Reperfusion
Revascularization is key to improving outcomes with
endovascular stroke therapy. Recanalization of the
occluded vessel and reperfusion of the distal capillary
bed are measures of revascularization, and, although
intimately linked, are not necessarily interchangeable.
Of the two measures, reperfusion of the distal capillary
bed is most linked with clinical outcome (106).
Reperfusion can be assessed by using CT or MR per-
fusion imaging. On angiography, crude assessment of
reperfusion can be made by assessing blood flow into
the distal bed, but this does not necessarily correlate
with reperfusion on a microcirculatory level (106).
Although advances have been made in perfusion assess-
ment in the angiographic suite (107), this assessment is
not readily available at the present time. Therefore,
most interventionalists will rely on a combination of
recanalization and reperfusion to assess
revascularization.
Revascularization can be assessed in a number of
ways, including the thrombolysis in cerebral infarction
(TICI) scale (108), the thrombolysis in myocardial
infarction (TIMI) scale (109,110), the Mori reperfusion
scale (111), the Qureshi scale (112), and the Arterial
Occlusive Lesion score (113), among others. No direct
comparisons of the revascularization scales in terms of
their predictive ability for final infarct volume exist,
but, through expert comparison of scales, the stroke
and interventional community favors the use of the
TICI scale (114,115). In a comparison of TIMI versus
TICI scales, TICI was found to be superior to TIMI.
The mTICI scale (Table 1) shifted the definition of a
grade of 2b to reperfusion of >50% rather than >66%
of the distal territory (31), and mTICI grade 2b/3 was
used as the definition of procedural success in the
majority of the successful endovascular trials. This is
the scale recommended for future studies (30). A fur-
ther refinement to the TICI scale introduced a new cat-
egory of 2c to define angiographic revascularization
of >90% and <100% of the distal territory (116).
However, the clinical applicability of TICI grade 2c
has not been validated in larger prospective trials.
Nevertheless, the higher the recanalization and reperfu-
sion grade, the better the outcome, with particularly
improved outcomes seen with grades of 2b or higher
(116) and the best outcomes seen with TICI grade 3
revascularization (117).
Compared with earlier studies (9,17), the positive
clinical trials of endovascular stroke therapy showed
vastly improved revascularization rates, with mTICI
grade 2b/3 rates ranging from 58.7% to 88.0%
(7,11,13,15,20), and the HERMES meta-analysis (14)
found an mTICI grade 2b/3 rate of 71%. The
THERAPY trial (18) reported an mTICI grade 2b/3
rate of 73%. This was assessed with the use of core
laboratory adjudication in most studies, and it has
been shown that local sites tend to overestimate the
degree of reperfusion compared with a core laboratory
(99). Postmarket registries have found mTICI
grade 2b rates of 70.9%–73.9%, but no central adju-
dication was performed (97,118,119). Based on this, an
mTICI grade 2b rate of 70% seems a reasonable
International Journal of Stroke, 0(0)
10 International Journal of Stroke 0(0)
number for all acute ischemic strokes treated. Only
moderate agreement exists between raters for the
TICI scale, even though agreement is substantial
when the scale is dichotomized into successful (ie,
TICI grade 2b/3) or unsuccessful outcomes (ie, TICI
grade 0, 1, or 2a) (120).
In terms of technical success of procedures, it is also
important to note the presence of distal embolization
and embolization to new territory (31). The ultimate
goal of revascularization is to improve patient out-
comes. However, there is a risk that persistent attempts
to recanalize an occlusion may lead to more complica-
tions. The combined metrics for SICH, revasculariza-
tion, and mRS scores of 0–2 measure these risks and
benefits.
Metric 7: The mTICI scale should be the primary scale
used to assess angiographic reperfusion.
Metric 8: At least 70% of patients should have mTICI
grade 2b/3 (ie, >50%) reperfusion for all clot locations.
Postprocedural CT/MR Imaging
Postprocedural imaging is necessary to identify acute
SAH or parenchymal hematoma, differentiate intrapar-
enchymal hemorrhage from contrast staining, define
the overall extent of new stroke, and identify other
findings. Although there is no evidence that this
improves clinical outcomes, there is consensus based
on European guidelines that postprocedural imaging
is required (121). CT or MR imaging within 36 hours
after intervention should be performed in all stroke
patients (7,11,15,20). Although some patients may
receive CT or MR imaging immediately after the pro-
cedure, imaging performed the next day provides add-
itional valuable information. It is recognized that there
are certain circumstances that might render follow-up
imaging difficult or impossible to perform. Therefore,
the threshold for this imaging is 90%, acknowledging
that a goal of 100% is desired.
Metric 9: At least 90% of patients should have a brain
CT or MR imaging examination within 36 hours of the
end of the procedure.
SICH
The most common major risk of endovascular treat-
ment of acute ischemic stroke is SICH. As defined by
individual studies, the incidences of SICH following
endovascular revascularization range from 2% to
10% for combined intravenous and intraarterial
thrombolytic trials (9,12,39,122) and from 1% to 8%
for EVT trials (7,11,13,20,121). Several definitions have
been used, as described in the National Institute of
Neurological Disorders and Stroke trial (1), the SITS-
MOST (33) and INSTOR registries (78), and European
registries such as SITS-Thrombectomy (32,81), MR
CLEAN (82) (Netherlands), and the Heidelberg
Bleeding Classification (34).
SAH is a unique complication of endovascular ther-
apy and is not typically seen with intravenous therapy
with TPA alone. Intraprocedural SAH caused by arter-
ial perforation can be rapidly fatal, but has been
described as being asymptomatic in as many as 16%
of patients treated with mechanical thrombectomy
without perforation (123).
The definition chosen for SICH in this document is
based on that used by the SWIFT PRIME trial (20) and
includes any intracranial hemorrhage with neurologic
deterioration leading to an increase in NIHSS score >4
or leading to death within 24 hours of treatment.
SICH is not only an ‘‘end-result’’ evaluation of clin-
ical judgment in the realm of patient selection and tech-
nical skill, but also a reflection of timing, procedural
execution, and expeditious completion of the task. For
these reasons, tracking of SICH is mandatory.
Metric 10: 100% of cases with SICH are reviewed (see
Quality Improvement).
Metric 11: No more than 10% of treated patients
should develop SICH.
Embolization of New Territory
Embolization of previously unaffected territories and
embolization as a result of clot fragmentation within
the treated territory can occur during endovascular
treatment. Distal embolization within the treated terri-
tory is different from embolization of new territory and
has been reported in 16% of patients treated with endo-
vascular thrombolysis and 35% of patients treated with
thrombectomy, without decreasing the likelihood of a
favorable outcome (124,125). Embolization of new ter-
ritory has been reported in 5%–9% of patients treated
in the recent EVT trials (7,13,20) and may cause new
areas of symptomatic infarct or require additional
treatment of previously unaffected vessels.
Metric 12: No more than 10% of patients should have
embolization of new territory.
Death within 72 Hours of Treatment
Death within 72 hours of stroke is typically not a result
of the stroke itself. The authors clearly acknowledge
International Journal of Stroke, 0(0)
Sacks et al. 11
that every case is unique and that each instance needs to
be reviewed in its entirety with the understanding that
there are circumstances (eg, myocardial infarction) that
lead to death in the short term and are unrelated to
operator factors. Death soon after a procedure in and
of itself does not imply or indicate a quality problem.
However, all deaths within 72 hours are a trigger for
review.
Metric 13: 100% of cases of death within 72 hours of
the end of the procedure are reviewed.
Clinical Outcomes
Ultimately, the goal of endovascular stroke therapy is
to limit the size and extent (ie, severity) of stroke,
improve the clinical outcome of the patient, and pre-
vent long-term disability. By convention, these out-
comes are commonly assessed by using various
functional grading systems: during initial hospitaliza-
tion, stroke is commonly assessed based on changes
in the NIHSS score, and then, often at 90 days, by
using the mRS. Clinical outcomes of stroke revascular-
ization are multifactorial, depending on factors intrin-
sic to the patient such as preexisting cerebral artery
collateral vessels, procedural factors such as time to
revascularization and completeness of revasculariza-
tion, as well as the patient’s response to a host of inter-
ventions in intensive care and then rehabilitation.
Among specific patient factors, higher admission
NIHSS scores and age were shown in the HERMES
meta-analysis (14) to portend worse outcomes with
medical or endovascular therapy. Other medical
comorbidities such as underlying cardiac disease,
hypertension, and diabetes mellitus all play a role in
outcomes. From a procedural standpoint, higher rates
of recanalization are associated with improved out-
comes. A key component of any interventional stroke
program is tracking of clinical outcomes. To that end,
we propose that a discharge NIHSS score be docu-
mented on all patients, and that all patients are con-
tacted and evaluated to obtain an mRS score at 90
days. Early improvement in NIHSS score may function
as a surrogate marker of outcome in situations in which
an mRS score cannot be obtained (126,127). Although
it is ideal to assess the patient in person, this may not
always be possible, and telephone assessment of mRS
score is a reasonable alternative that is well validated
(128). We understand that some patients may be lost to
follow-up by 90 days.
Metric 14: All treated patients have a documented
NIHSS score 20–36 hours after treatment and at dis-
charge. Attempts are made to contact and document a
follow-up mRS score at 90 days (evaluated in person or
via telephone) on all treated patients. At least 90% of
treated patients have documented 90-day mRS score.
Determining a single threshold level of ‘‘good clinical
outcome’’ for all patient populations is difficult because
of the heterogeneity of treated patients and the absence
of comprehensive data. Individual centers, for example,
may have a more elderly patient population or patients
with later presentations. The incidences of patients with
an mRS score of 0–2 at 90 days in the recent rando-
mized controlled endovascular trials ranged from 33%
(MR CLEAN) (7) to 71% (EXTEND-IA) (11), with an
overall aggregate rate of 46% in the HERMES trial
(14). Similarly, the THERAPY (18) and Trial and
Cost Effectiveness Evaluation of Intra-arterial
Thrombectomy in Acute Ischemic Stroke (THRACE)
(8) trials reported 38% and 53% rates of mRS score 0–
2 at 90 days, respectively, and, in the per-protocol
population of the Pragmatic Ischaemic
Thrombectomy Evaluation (PISTE) trial (129), 57%
of the endovascular group reached an mRS score of
0–2 at 90 days.
The major trials focused on stroke patients with
large artery occlusions, specifically internal carotid ter-
minus or proximal middle cerebral (ie, M1) arteries.
However, some patients with severe stroke have occlu-
sions at other locations. Patients with isolated M2
branch occlusions may be reasonable candidates for
EVT, but, in general, the natural history of stroke in
these patients is better than those with more proximal
occlusions (130). Although there are no randomized
data showing a benefit for thrombectomy in basilar
artery occlusions, these are often treated at many cen-
ters (131). Several studies have specifically reported
worse outcomes for patients who did not meet the
trial inclusion criteria or the current American Heart
Association level IA recommendations (22). Gratz et al
(119) reported 30% versus 57% incidences of mRS
score of 0–2 for high-risk patients versus standard-
risk patients. Similarly, Goyal et al (75) reported 39%
versus 47% incidences of mRS score of 0–2 for patients
not meeting versus meeting AHA level I
recommendations.
One must take prestroke functional status into
account when setting a threshold for 90-day mRS
score for good outcome. The vast majority of patients
in the recent randomized trials had an mRS score of 0/1
at baseline. As described in the Indications section, this
is not to imply that EVT be withheld for those who do
not have an mRS score of 0/1, but that any outcome
threshold needs to account for prestroke functional
status.
Multicenter registries have reported results with
modern thrombectomy in more heterogenous groups
International Journal of Stroke, 0(0)
12 International Journal of Stroke 0(0)
of patients, including patients with vertebrobasilar and
M2 clot locations as well as tandem lesions. The regis-
tries include the German Register on Revascularization
in Ischemic Stroke Patients (REVASK) registry
(N ¼1,107 patients, 40% mRS score 0–2 at 90 d)
(132), the Catalonia stroke registry (N ¼536 patients,
43% mRS score 0–2 at 90 d) (118), the North American
Solitaire registry (N ¼354 patients, 42% mRS score 0–
2 at 90 d) (133), and the Madrid registry (N ¼479, 54%
mRS score 0–2 at 90 d) (131). The Dutch MR CLEAN
registry (134) reported a 41% incidence of mRS score
0–2 at 90 days in a group of 1,321 patients. Compared
with the randomized trials, the registries will include
some patients at higher risk (ie, basilar occlusions),
some at lower risk (ie, M2 occlusions), and some
biased data, as the data are not adjudicated, likely lead-
ing to better reported outcomes. However, this is likely
to be similar to the experiences of hospitals using the
metrics of this document.
Clinical acumen is needed to determine the risk
versus benefit of treatment based on published trial
and registry data and personal experience. The AHA
has created level I recommendations (22) for patient
selection based on current randomized trials.
However, we expect these recommendations to evolve
as results from trials address ‘‘wake-up’’ strokes, pro-
longed time from symptom onset, basilar artery occlu-
sions, large infarct size, and ‘‘mothership’’ versus ‘‘drip-
and-ship’’ cases (48,49,54,55,59,135).
We propose a single threshold for clinical outcomes
for all treated patients regardless of whether they would
have been candidates for the majority of recent trials or
meet the AHA level I recommendations. This is con-
sistent with the heterogeneity of current clinical practice
in which nearly half of treated patients do not meet the
AHA recommendations (75). This document does not
advocate for or against treating patients outside of the
randomized trial or AHA level I recommendations, but
suggests a threshold that recognizes the common prac-
tice of treating such patients. The threshold of a 30%
incidence of mRS score of 0–2 at 90 days is lower than
those of the recent randomized trials and registries
based on the experience that ‘‘off-trial’’ patients are
more likely to be at higher risk for poor outcomes
(75,119) and the belief that the published registry
results may not reflect the most current trends in
patient selection. It is important to note that, although
achieving an mRS score of 0–2 is an important goal, it
is not the only marker of a favorable outcome after
endovascular therapy. Some patients may have import-
ant clinical benefit with an mRS score shift from 4/5
down to 3. However, mRS score shift analysis requires
a control group comparison, which is not useful as a
quality metric. This suggested threshold should not dis-
suade centers from treating individual patients if they
believe there is a potential benefit from the procedure.
Given the multiple factors that influence outcomes, cen-
ters are encouraged to benchmark their outcomes
against those from a similar patient population.
The clinical outcome threshold of this document is
intended to prompt internal review of the endovascular
stroke program. It is not designed to constitute a stand-
ard for reimbursement from payers, or for accreditation
purposes. Local patient factors such as overall medical
comorbidities and time from symptom onset to treat-
ment should be taken into account when reviewing any
single institution’s performance. This is especially true
in those patients who have a greater degree of prestroke
disability or other comorbidities that may have
excluded them from the recent randomized trials, but
for whom treatment may be warranted.
Metric 15: Of all treated patients, at least 30% are inde-
pendent (ie, mRS score 0–2) at 90 days after treatment.
Quality improvement
Ongoing Quality Improvement
As EVT of acute ischemic stroke becomes a mainstream
offering at many centers, an endovascular-specific
multidisciplinary QI process should be established in
all programs offering this treatment (24,25). These
endovascular cases, similar to trauma cases, require
complex processes of care. These processes go beyond
the clinical and technical skills of the operators them-
selves and should be monitored in a continuous and
ongoing fashion.
A peer-review committee at the local hospital should
be formed that involves personnel from the several
backgrounds that have expertise in stroke care as well
as a vested interest in quality of care and outcomes.
This committee should provide an open and transpar-
ent forum for process and case review. Transparency
will optimize confidence in the process, which should
have a positive impact on patient care. Although there
may be potential for conflict or disagreement among
various participants, it is vital that the process be
viewed as a nonpolitical, nonpunitive instrument for
care process improvement.
Specifically within the United States, in keeping with
standards established under the Health Care Quality
Improvement Act of 1986 (42 USC §11101 et seq.),
peer-review meetings and minutes are generally pro-
tected from legal inquiry in most states as long as the
review is conducted under the auspices of the facility QI
program. The Health Care Quality Improvement Act
established standards for professional review actions.
Although this protection is not absolute, if a
International Journal of Stroke, 0(0)
Sacks et al. 13
professional review body meets these standards, neither
the professional review body nor any person acting as a
member or staff to the body will be liable for damages
under most federal or state laws with respect to the
action (136–139). All associated QI documents should
include routine annotation that establishes the purpose
of the document and that its content is protected under
applicable federal or state law. The program should
operate under the local facility umbrella established
for all facility QI and peer-review initiatives.
Peer Review Team
It is recommended that, under the oversight of the stroke
team medical director, a predetermined multidisciplin-
ary subgroup consisting of medical personnel with
familiarity and expertise in endovascular therapy be
established to address issues specifically relating to endo-
vascular treatment. Although a stroke neurologist is
generally in the best overall position to objectively
assess overall process deficiencies and outcomes, for
technical and procedural issues, an interventionalist per-
spective must be considered. Ideally, the endovascular
oversight team should be directed by a highly qualified
and unbiased physician such as a noninterventional vas-
cular neurologist. Depending on the institution, the
endovascular QI peer group could include a variable
combination of interventionalists, vascular neurologists,
cerebrovascular neurosurgeons, intensivists, and diag-
nostic neuroradiologists. Additional members might
include hospital representative(s) from the quality assur-
ance/improvement or risk management departments, as
well as possibly the stroke coordinator or other data
personnel and secretarial support staff.
Review Process
The endovascular QI meeting should occur at least
quarterly, and, depending on volume, may need to
occur more frequently to provide adequate assessment
and review. There should be review of every case in
centers with volumes <50 cases per year and review
of every case in which the parameters are outside the
benchmarks (eg, prolonged time to puncture, failure of
reperfusion, prolonged time to reperfusion) or in which
a complication occurs (eg, SICH, embolization of new
territory, or death within 72 h). As noted earlier in the
section on data collection, all cases should be entered
into a trial, database, or registry with national partici-
pation (24,25,76). In the United States, Medicare is
functioning under the Medicare Access and Children’s
Health Insurance Program Reauthorization Act of
2015 (140), which seeks to align disparate quality pro-
grams through Qualified Clinical Data Registries. This
approach is in keeping with our aforementioned
recommendation for data collection and quality control
(141,142).
The interventionalist who performed the specific
case under review should be present to offer his/her
observations and perspective. The focused endovascu-
lar peer review should routinely include assessment of
technical factors such as device choice, supplemental
lytic agent infusion, and equipment inventory assess-
ment. Process elements such as on-call notification,
timing (ie, door-to-imaging and imaging–to–arterial
puncture times), procedure table setup, and overall
communication should also receive routine attention.
Performance review is not limited to the treating endo-
vascular physician, but should also include the emer-
gency department, neurology and neurointensive care
personnel, interventional technologists, nursing staff,
and other related service areas as indicated.
Information concerning transfer from and communica-
tion with referring primary stroke centers before and
after return to the primary center, complications, and
90-day functional outcome should also be routinely dis-
cussed and benchmarked.
Triggers for Review
Any event that might affect quality should be reviewed.
Specific triggers for endovascular review include unmet
process benchmarks, death, and symptomatic postpro-
cedural hemorrhage. Some complications or process
delays may be unavoidable, whereas others may reflect
significant errors in judgment or process deficiencies.
A determination must be made if the patient was
harmed. Process problems such as delays or inadequate
communication increase the risk of harm. Therefore,
complications and events that increase the risk of
poor outcomes need to be reviewed as a means of
improving quality. There must also be differentiation
between clearly procedure-related complications (eg,
perforation and/or dissection, distal dislodgment of
thrombus that remains unreachable, embolization of
new territory, and immediate SICH following the pro-
cedure) and those that might be related to the primary
ischemic event itself (eg, infarction, cerebral edema, and
hemorrhagic transformation). Predisposing underlying
vascular disease and comorbidities must also be
considered.
Physicians who choose to treat sicker patients may
have poorer outcomes and may not meet established
benchmarks. These cases should not be considered in
isolation, as a poor outcome does not necessarily indi-
cate that such physicians are providing a lower quality
of care, but rather that they have a different patient mix
than the trials that were used to create the benchmarks
(7,8,10,11,13–15,18,20,143–145). Adjusting for risk and
severity may be helpful in assessing local outcomes
International Journal of Stroke, 0(0)
14 International Journal of Stroke 0(0)
Table 3. Endovascular Therapy Quality Improvement Case Review Triggers and Process Metrics
Indications for Endovascular Treatment
Metric 1: At least 90% of patients who meet the institution selection criteria (indications/contraindications) should be treated
with endovascular therapy.
Data Collection
Metric 2: 100% of patients have the required minimum process and outcomes data entered into an institutional or national
database, trial, or registry.
Key Time Intervals
Door to imaging
Metric 3: 75% of patients being evaluated for revascularization should have imaging initiated within 30 minutes from time of
arrival. At the best of centers with high volumes and an established resource infrastructure, this is expected to be achieved in
12 minutes.
Imaging to puncture
Metric 4: 75% of patients treated with endovascular therapy should have an imaging-to-puncture time of 110 minutes or less. At
the best of centers with high volumes and an established resource infrastructure, this is expected to be achieved in 50 minutes
or less.
Metric 5: For patients transferred from another site and in whom imaging is not repeated, 75% of patients being treated should
have a door-to-puncture time of 80 minutes or less.
Puncture to revascularization
Metric 6: In 70% of patients, mTICI score 2b should be reached ideally within 60 minutes of arterial puncture.
Outcome Metrics
Recanalization/reperfusion
Metric 7: The mTICI scale should be the primary scale used to assess angiographic reperfusion.
Metric 8: At least 70% of patients should have an mTICI score 2b/3 (>50% reperfusion) for all clot locations.
Postprocedure CT/MR Imaging
Metric 9: At least 90% of patients should have a brain CT or MR imaging within 36 hours of the end of the procedure.
SICH
Metric 10: 100% of cases with SICH are reviewed.
Metric 11: No more than 10% of treated patients should develop SICH.
Embolization of new territory
Metric 12: No more than 10% of patients should have embolization of new territory.
Death within 72 hours of treatment
Metric 13: 100% of cases of death within 72 hours of the end of the procedure are reviewed.
Clinical Outcomes
Metric 14: All treated patients have a documented NIHSS score at discharge. Attempts are made to contact and document a
follow-up mRS score at 90 days (evaluated in person or via telephone) on all treated patients. At least 90% of treated patients
have a documented 90-day mRS score.
Metric 15: Of all treated patients, at least 30% are independent (ie, mRS score 0–2) at 90 days after treatment.
mRS: modified Rankin scale; mTICI: modified thrombolysis in cerebral infarction; NIHSS: National Institutes of Health Stroke Scale; SICH: symptomatic
intracranial hemorrhage.
International Journal of Stroke, 0(0)
Sacks et al. 15
compared with other institutions and benchmarks.
Endovascular QI case review triggers and key process
metrics are summarized in Table 3.
In addition to these morbidity and mortality mar-
kers, it is incumbent on the institution and the quality-
assurance/improvement and peer-review committee to
also assess the ‘‘good outcomes.’’ A certain percentage
of good outcomes are necessary for there to be suffi-
cient benefit to the overall patient population. This
document also defines minimal recanalization rates as
well as improved clinical outcomes that should be
attained.
Performance and Process Improvement
The committee should be equipped to deal with poor
performance in a supportive, constructive, and collegial
manner. In cases in which negative trends and deficien-
cies become apparent, improvement may require indi-
vidual mentoring, additional education, or
supplemental training. Endovascular stroke QI review
of problematic cases should generate a specific course
of action to remedy recognized problems and prevent
future occurrences. Individual assignments should be
tracked, with accountability reports scheduled for sub-
sequent meetings. Further, process improvement is a
continuing activity that, along with individual perform-
ance improvement, will significantly impact clinical
outcomes (146).
Acknowledgments
The authors thank Zachary Zhang, MD (Rochester General
Hospital, Rochester, New York), for assistance creating the
evidence table and formatting the document; and Sean A.
Kennedy, MD (University of Toronto, Toronto, Ontario),
and Ajinkya Desai, MD (Rochester General Hospital,
Rochester, New York), for assistance creating the
evidence table.
Declaration of Conflicting Interests
The author(s) declared the following potential conflicts of
interest with respect to the research, authorship, and/or pub-
lication of this article: B.B. receives personal fees from
Penumbra (Alameda, California), Medtronic (Dublin,
Ireland), Stryker (Fremont, California), and Pulsar Vascular
(Los Gatos, California) and has a patent (US8622992) issued
to Advanced Catheter Therapies (Chattanooga, Tennessee).
C.C. receives personal fees from Microvention (Tokyo,
Japan), Stryker, Medtronic, and Balt (Montmorency,
France). D.D. receives grants from the Dutch Heart
Foundation, AngioCare (Eemnes, The Netherlands),
Medtronic/Covidien/ev3 (Dublin, Ireland), Medac/Lamepro
(Breda, The Netherlands), Penumbra, Top Medical/
Concentric (New York, New York), and Stryker. U.F.
receives grants from the SWIFT DIRECT study and personal
fees from Medtronic. J.A.H. receives personal fees from
Medtronic, Globus (Audubon, Pennsylvania), and Codman
Neuro (Raynham, Massachusetts). C.M.S. receives grants
from the National Institutes of Health/National Institute of
Neurological Disorders and Stroke, Medtronic, and
Penumbra and personal fees from Toshiba (Otawara,
Japan) and ExpertiCas (Southport, Connecticut) and is a
stockholder in Neurotechnology Investors (Palo Alto,
California). None of the other authors have identified a con-
flict of interest.
Supplementary Material
Table E1 is available online at www.jvir.org.
References
1. Tissue plasminogen activator for acute ischemic stroke.
The National Institute of Neurological Disorders and
Stroke rt-PA Stroke Study Group. N Engl J Med 1995;
333: 1581–1587.
2. Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease
and stroke statistics–2015 update: a report from the
American Heart Association. Circulation 2015; 131:
e29–e322.
3. Gibson CM, Pride YB, Frederick PD, et al. Trends in
reperfusion strategies, door-to-needle and door-to-bal-
loon times, and in-hospital mortality among patients
with ST-segment elevation myocardial infarction enrolled
in the National Registry of Myocardial Infarction from
1990 to 2006. Am Heart J 2008; 156: 1035–1044.
4. Sacks D, Black CM, Cognard C, et al. Multisociety con-
sensus quality improvement guidelines for intraarterial
catheter-directed treatment of acute ischemic stroke,
from the American Society of Neuroradiology,
Canadian Interventional Radiology Association,
Cardiovascular and Interventional Radiological Society
of Europe, Society for Cardiovascular Angiography and
Interventions, Society of Interventional Radiology,
Society of NeuroInterventional Surgery, European
Society of Minimally Invasive Neurological Therapy,
and Society of Vascular and Interventional Neurology.
J Vasc Interv Radiol 2013; 24: 151–163.
5. White PM, Bhalla A, Dinsmore J, et al. Standards for
providing safe acute ischaemic stroke thrombectomy ser-
vices (September 2015). Clin Radiol 2017; 72: 175.e1–e9.
6. Badhiwala JH, Nassiri F, Alhazzani W, et al.
Endovascular thrombectomy for acute ischemic stroke:
a meta-analysis. JAMA 2015; 314: 1832–1843.
7. Berkhemer OA, Fransen PS, Beumer D, et al. A rando-
mized trial of intraarterial treatment for acute ischemic
stroke. N Engl J Med 2015; 372: 11–20.
8. Bracard S, Ducrocq X, Mas JL, et al. Mechanical
thrombectomy after intravenous alteplase versus alte-
plase alone after stroke (THRACE): a randomised con-
trolled trial. Lancet Neurol 2016; 15: 1138–1147.
9. Broderick JP, Palesch YY, Demchuk AM, et al.
Endovascular therapy after intravenous t-PA versus t-
PA alone for stroke. N Engl J Med 2013; 368: 893–903.
10. Campbell BC, Hill MD, Rubiera M, et al. Safety and
efficacy of solitaire stent thrombectomy: individual
International Journal of Stroke, 0(0)
16 International Journal of Stroke 0(0)
patient data meta-analysis of randomized trials. Stroke
2016; 47: 798–806.
11. Campbell BC, Mitchell PJ, Kleinig TJ, et al.
Endovascular therapy for ischemic stroke with perfu-
sion-imaging selection. N Engl J Med 2015; 372:
1009–1018.
12. Ciccone A, Valvassori L, Nichelatti M, et al;
SYNTHESIS Expansion Investigators. Endovascular
treatment for acute ischemic stroke. N Engl J Med
2013; 368: 904–913.
13. Goyal M, Demchuk AM, Menon BK, et al. Randomized
assessment of rapid endovascular treatment of ischemic
stroke. N Engl J Med 2015; 372: 1019–1030.
14. Goyal M, Menon BK, van Zwam WH, et al.
Endovascular thrombectomy after large-vessel ischaemic
stroke: a meta-analysis of individual patient data from
five randomised trials. Lancet 2016; 387: 1723–1731.
15. Jovin TG, Chamorro A, Cobo E, et al. Thrombectomy
within 8 hours after symptom onset in ischemic stroke.
N Engl J Med 2015; 372: 2296–2306.
16. Kennedy SA, Baerlocher MO, Baerlocher F, et al. Meta-
analysis of local endovascular therapy for acute ischemic
stroke. J Vasc Interv Radiol 2016; 27: 307–321.
17. Kidwell CS, Jahan R, Gornbein J, et al. A trial of ima-
ging selection and endovascular treatment for ischemic
stroke. N Engl J Med 2013; 368: 914–923.
18. Mocco J, Zaidat OO, von Kummer R, et al. Aspiration
thrombectomy after intravenous alteplase versus intra-
venous alteplase alone. Stroke 2016; 47: 2331–2338.
19. Prabhakaran S, Ruff I and Bernstein RA. Acute stroke
intervention: a systematic review. JAMA 2015; 313:
1451–1462.
20. Saver JL, Goyal M, Bonafe A, et al. Stent-retriever
thrombectomy after intravenous t-PA vs. t-PA alone in
stroke. N Engl J Med 2015; 372: 2285–2295.
21. NICE Interventional Procedure Guidance. Mechanical
clot retrieval for treating acute ischaemic stroke.
Interventional procedures guidance [IPG548], February
2016. Available at: http://www.nice.org.uk/guidance/
ipg548. Accessed April 10, 2016.
22. Powers WJ, Derdeyn CP, Biller J, et al. 2015 American
Heart Association/American Stroke Association focused
update of the 2013 guidelines for the early management
of patients with acute ischemic stroke regarding endovas-
cular treatment: a guideline for healthcare professionals
from the American Heart Association/American Stroke
Association. Stroke 2015; 46: 3020–3035.
23. Wahlgren N, Moreira T, Michel P, et al. Mechanical
thrombectomy in acute ischemic stroke: consensus state-
ment by ESO-Karolinska stroke update 2014/2015, sup-
ported by ESO, ESMINT, ESNR and EAN. Int J Stroke
2016; 11: 134–147.
24. Connors JJ III, Sacks D, Black CM, et al. Training guide-
lines for intra-arterial catheter-directed treatment of
acute ischemic stroke: a statement from a special writing
group of the Society of Interventional Radiology. J Vasc
Interv Radiol 2009; 20: 1507–1522.
25. Lavine SD, Cockroft K, Hoh B, et al. Training guidelines
for endovascular ischemic stroke intervention: an
international multi-society consensus document. AJNR
Am J Neuroradiol 2016; 37: E31–E34.
26. Jovin TG, Albers GW and Liebeskind DS; STAIR IX
Consortium. Stroke treatment academic industry round-
table: the next generation of endovascular trials. Stroke
2016; 47: 2656–2665.
27. Fink A, Kosecoff J, Chassin M and Brook RH.
Consensus methods: characteristics and guidelines for
use. Am J Public Health 1984; 74: 979–983.
28. Institute of Medicine (U.S.) Committee on Standards for
Developing Trustworthy Clinical Practice Guidelines.
Clinical Practice Guidelines We Can Trust. Graham R,
Mancher M, Wolman DM, Greenfield S, Steinberg E,
eds. Washington, DC: National Academies Press, 2011.
xxxiv.
29. Sacco RL, Kasner SE, Broderick JP, et al. An updated
definition of stroke for the 21st century: a statement for
healthcare professionals from the American Heart
Association/American Stroke Association. Stroke 2013;
44: 2064–2089.
30. Wintermark M, Albers GW, Broderick JP, et al. Acute
stroke imaging research roadmap II. Stroke 2013; 44:
2628–2639.
31. Zaidat OO, Yoo AJ, Khatri P, et al. Recommendations
on angiographic revascularization grading standards for
acute ischemic stroke: a consensus statement. Stroke
2013; 44: 2650–2663.
32. Wahlgren N, Ahmed N, Eriksson N, et al. Multivariable
analysis of outcome predictors and adjustment of main
outcome results to baseline data profile in randomized
controlled trials: safe implementation of thrombolysis in
stroke-monitoring study (SITS-MOST). Stroke 2008; 39:
3316–3322.
33. Wahlgren N, Ahmed N, Davalos A, et al. Thrombolysis
with alteplase for acute ischaemic stroke in the safe imple-
mentation of thrombolysis in stroke-monitoring study
(SITS-MOST): an observational study. Lancet 2007;
369: 275–282.
34. von Kummer R, Broderick JP, Campbell BC, et al. The
Heidelberg Bleeding Classification: classification of
bleeding events after ischemic stroke and reperfusion
therapy. Stroke 2015; 46: 2981–2986.
35. van Swieten JC, Koudstaal PJ, Visser MC, Schouten HJ
and van Gijn J. Interobserver agreement for the assess-
ment of handicap in stroke patients. Stroke 1988; 19:
604–607.
36. Casaubon LK, Boulanger JM, Blacquiere D, et al.
Canadian stroke best practice recommendations: hyper-
acute stroke care guidelines, update 2015. Int J Stroke
2015; 10: 924–940.
37. Campbell BC, Donnan GA, Lees KR, et al.
Endovascular stent thrombectomy: the new standard of
care for large vessel ischaemic stroke. Lancet Neurol 2015;
14: 846–854.
38. Coutts SB, Modi J, Patel SK, et al. CT/CT angiography
and MRI findings predict recurrent stroke after transient
ischemic attack and minor stroke: results of the prospect-
ive CATCH study. Stroke 2012; 43: 1013–1017.
39. Furlan A, Higashida R, Wechsler L, et al. Intra-arterial
prourokinase for acute ischemic stroke. The PROACT II
International Journal of Stroke, 0(0)
Sacks et al. 17
study: a randomized controlled trial. Prolyse in acute
cerebral thromboembolism. JAMA 1999; 282: 2003–2011.
40. Penumbra Pivotal Stroke Trial I. The penumbra pivotal
stroke trial: safety and effectiveness of a new generation
of mechanical devices for clot removal in intracranial
large vessel occlusive disease. Stroke 2009; 40: 2761–2768.
41. Smith WS, Sung G, Saver J, et al. Mechanical thrombec-
tomy for acute ischemic stroke: final results of the Multi
MERCI trial. Stroke 2008; 39: 1205–1212.
42. Tarr R, Hsu D, Kulcsar Z, et al. The POST trial: initial
post-market experience of the Penumbra system: revascu-
larization of large vessel occlusion in acute ischemic
stroke in the United States and Europe. J Neurointerv
Surg 2010; 2: 341–344.
43. Fransen PS, Berkhemer OA, Lingsma HF, et al. Time to
reperfusion and treatment effect for acute ischemic
stroke: a randomized clinical trial. JAMA Neurol 2016;
73: 190–196.
44. Goyal M, Jadhav AP, Bonafe A, et al. Analysis of work-
flow and time to treatment and the effects on outcome in
endovascular treatment of acute ischemic stroke: results
from the SWIFT PRIME randomized controlled trial.
Radiology 2016; 279: 888–897.
45. Khatri P, Abruzzo T, Yeatts SD, et al. Good clinical
outcome after ischemic stroke with successful revascular-
ization is time-dependent. Neurology 2009; 73:
1066–1072.
46. Saver JL, Goyal M, van der Lugt A, et al. Time to treat-
ment with endovascular thrombectomy and outcomes
from ischemic stroke: a meta-analysis. JAMA 2016;
316: 1279–1288.
47. Lansberg MG, Cereda CW, Mlynash M, et al. Response
to endovascular reperfusion is not time-dependent in
patients with salvageable tissue. Neurology 2015; 85:
708–714.
48. Nogueira R and Jovin T. Clinical mismatch in the triage
of wake up and late presenting strokes undergoing neu-
rointervention with trevo (DAWN). N Engl J Med 2018;
378: 11–21.
49. POSITIVE stroke clinical trial. Available at: https://clin-
icaltrials.gov/ct2/show/NCT01852201?term¼posi-
tiveþstrokeþtrial&rank¼1. Accessed February 12, 2016.
50. Endovascular therapy following imaging evaluation
for ischemic stroke 3 (DEFUSE 3). Available at:
https://clinicaltrials.gov/ct2/show/NCT02586415.
Accessed February 12, 2016.
51. Brandt T, von Kummer R, Muller-Kuppers M and
Hacke W. Thrombolytic therapy of acute basilar artery
occlusion. Variables affecting recanalization and out-
come. Stroke 1996; 27: 875–881.
52. Lin DD, Gailloud P, Beauchamp NJ, Aldrich EM, Wityk
RJ and Murphy KJ. Combined stent placement and
thrombolysis in acute vertebrobasilar ischemic stroke.
AJNR Am J Neuroradiol 2003; 24: 1827–1833.
53. Schonewille WJ, Wijman CA, Michel P, et al. Treatment
and outcomes of acute basilar artery occlusion in the
basilar artery international cooperation study
(BASICS): a prospective registry study. Lancet Neurol
2009; 8: 724–730.
54. Basilar artery international cooperation study (BASICS).
Available at: https://clinicaltrials.gov/ct2/show/NCT017
17755?term¼basicsþstrokeþtrial&rank¼1. Accessed
April 11, 2017.
55. Basilar artery occlusion Chinese endovascular trial.
Available at: https://clinicaltrials.gov/ct2/show/NCT027
37189?term¼bestþstrokeþtrial&rank¼2. Accessed April
11, 2017.
56. Hacke W, Kaste M, Fieschi C, et al. Randomised double-
blind placebo-controlled trial of thrombolytic therapy
with intravenous alteplase in acute ischaemic stroke
(ECASS II). Second European-Australasian Acute
Stroke Study Investigators. Lancet 1998; 352: 1245–1251.
57. Barber PA, Demchuk AM, Zhang J and Buchan AM.
Validity and reliability of a quantitative computed tom-
ography score in predicting outcome of hyperacute stroke
before thrombolytic therapy. ASPECTS Study Group.
Alberta Stroke Programme early CT score. Lancet
2000; 355: 1670–1674.
58. Pexman JH, Barber PA, Hill MD, et al. Use of the
Alberta Stroke Program early CT score (ASPECTS) for
assessing CT scans in patients with acute stroke. AJNR
Am J Neuroradiol 2001; 22: 1534–1542.
59. Efficacy and safety of thrombectomy in stroke with
extended lesion and extended time window (tension).
Available at: https://clinicaltrials.gov/show/
NCT03094715. Accessed April 11, 2017.
60. Riedel CH, Zoubie J, Ulmer S, Gierthmuehlen J and
Jansen O. Thin-slice reconstructions of nonenhanced
CT images allow for detection of thrombus in acute
stroke. Stroke 2012; 43: 2319–2323.
61. Riedel CH, Zimmermann P, Jensen-Kondering U,
Stingele R, Deuschl G and Jansen O. The importance
of size: successful recanalization by intravenous thromb-
olysis in acute anterior stroke depends on thrombus
length. Stroke 2011; 42: 1775–1777.
62. Rohan V, Baxa J, Tupy R, et al. Length of occlusion
predicts recanalization and outcome after intravenous
thrombolysis in middle cerebral artery stroke. Stroke
2014; 45: 2010–2017.
63. Ahn SH, d’Esterre CD, Qazi EM, et al. Occult antero-
grade flow is an under-recognized but crucial predictor of
early recanalization with intravenous tissue-type plas-
minogen activator. Stroke 2015; 46: 968–975.
64. Menon BK, d’Esterre CD, Qazi EM, et al. Multiphase
CT angiography: a new tool for the imaging triage of
patients with acute ischemic stroke. Radiology 2015;
275: 510–520.
65. Gilgen MD, Klimek D, Liesirova KT, et al. Younger
stroke patients with large pretreatment diffusion-
weighted imaging lesions may benefit from endovascular
treatment. Stroke 2015; 46: 2510–2516.
66. Borst J, Berkhemer OA, Roos YB, et al. Value of com-
puted tomographic perfusion-based patient selection for
intra-arterial acute ischemic stroke treatment. Stroke
2015; 46: 3375–3382.
67. d’Esterre CD, Boesen ME, Ahn SH, et al. Time-depen-
dent computed tomographic perfusion thresholds for
patients with acute ischemic stroke. Stroke 2015; 46:
3390–3397.
International Journal of Stroke, 0(0)
18 International Journal of Stroke 0(0)
68. Barreto AD, Albright KC, Hallevi H, et al. Thrombus
burden is associated with clinical outcome after intra-
arterial therapy for acute ischemic stroke. Stroke 2008;
39: 3231–3235.
69. Lin R, Vora N, Zaidi S, et al. Mechanical approaches
combined with intra-arterial pharmacological therapy
are associated with higher recanalization rates than
either intervention alone in revascularization of acute car-
otid terminus occlusion. Stroke 2009; 40: 2092–2097.
70. Shi ZS, Loh Y, Walker G and Duckwiler GR; MERCI
and Multi-MERCI Investigators. Clinical outcomes in
middle cerebral artery trunk occlusions versus secondary
division occlusions after mechanical thrombectomy:
pooled analysis of the mechanical embolus removal in
cerebral ischemia (MERCI) and multi MERCI trials.
Stroke 2010; 41: 953–960.
71. Coutinho JM, Liebeskind DS, Slater LA, et al.
Mechanical thrombectomy for isolated M2 occlusions: a
post hoc analysis of the STAR, SWIFT, and SWIFT
PRIME studies. AJNR Am J Neuroradiol 2016; 37:
667–672.
72. Busl KM, Nogueira RG, Yoo AJ, Hirsch JA, Schwamm
LH and Rost NS. Prestroke dementia is associated with
poor outcomes after reperfusion therapy among elderly
stroke patients. J Stroke Cerebrovasc Dis 2013; 22:
718–724.
73. Khatri P, Hill MD, Palesch YY, et al. Methodology of
the Interventional Management of Stroke III Trial. Int J
Stroke 2008; 3: 130–137.
74. Bhole R, Goyal N, Nearing K, et al. Implications of
limiting mechanical thrombectomy to patients with emer-
gent large vessel occlusion meeting top tier evidence cri-
teria. J Neurointerv Surg 2017; 9: 225–228.
75. Goyal N, Tsivgoulis G, Frei D, et al. A multicenter study
of the safety and effectiveness of mechanical thrombec-
tomy for patients with acute ischemic stroke not meeting
top-tier evidence criteria. J Neurointerv Surg 2018; 10:
10–16.
76. Leifer D, Bravata DM, Connors JJ III, et al. Metrics for
measuring quality of care in comprehensive stroke cen-
ters: detailed follow-up to Brain Attack Coalition com-
prehensive stroke center recommendations: a statement
for healthcare professionals from the American Heart
Association/American Stroke Association. Stroke 2011;
42: 849–877.
77. Saver JL and Gornbein J. Treatment effects for which
shift or binary analyses are advantageous in acute
stroke trials. Neurology 2009; 72: 1310–1315.
78. Interventional stroke therapy outcomes registry
(INSTOR). Available at: http://www.strokeregistry.org.
Accessed July 19, 2016.
79. Meyers PM, Schumacher HC, Alexander MJ, et al.
Performance and training standards for endovascular
ischemic stroke treatment. J Stroke Cerebrovasc Dis
2009; 18: 411–415.
80. Schwamm LH, Fonarow GC, Reeves MJ, et al. Get with
the guidelines—stroke is associated with sustained
improvement in care for patients hospitalized with acute
stroke or transient ischemic attack. Circulation 2009; 119:
107–115.
81. SITS–thrombectomy registry. Available at: http://www.
sitsinternational.org/registries/sits-thrombectomy/.
Accessed August 12, 2016.
82. MR CLEAN registry. Available at: http://www.mrclean-
trial.org.
83. Menon BK, Saver JL, Goyal M, et al. Trends in endo-
vascular therapy and clinical outcomes within the nation-
wide get with the guidelines–stroke registry. Stroke 2015;
46: 989–995.
84. Hacke W, Donnan G, Fieschi C, et al. Association of
outcome with early stroke treatment: pooled analysis of
ATLANTIS, ECASS, and NINDS rt-PA stroke trials.
Lancet 2004; 363: 768–774.
85. Lansberg MG, Schrooten M, Bluhmki E, Thijs VN and
Saver JL. Treatment time-specific number needed to treat
estimates for tissue plasminogen activator therapy in
acute stroke based on shifts over the entire range of the
modified Rankin scale. Stroke 2009; 40: 2079–2084.
86. Menon BK, Sajobi TT, Zhang Y, et al. Analysis of work-
flow and time to treatment on thrombectomy outcome in
the endovascular treatment for small core and proximal
occlusion ischemic stroke (ESCAPE) randomized, con-
trolled trial. Circulation 2016; 133: 2279–2286.
87. Ribo M, Molina CA, Cobo E, et al. Association between
time to reperfusion and outcome is primarily driven by
the time from imaging to reperfusion. Stroke 2016; 47:
999–1004.
88. Bradley EH, Curry LA, Webster TR, et al. Achieving
rapid door-to-balloon times: how top hospitals improve
complex clinical systems. Circulation 2006; 113:
1079–1085.
89. Krumholz HM, Bradley EH, Nallamothu BK, et al. A
campaign to improve the timeliness of primary percutan-
eous coronary intervention: door-to-balloon: an alliance
for quality. JACC Cardiovasc Interv 2008; 1: 97–104.
90. Fonarow GC, Smith EE, Saver JL, et al. Improving door-
to-needle times in acute ischemic stroke: the design and
rationale for the American Heart Association/American
Stroke Association’s target: stroke initiative. Stroke 2011;
42: 2983–2989.
91. Mitka M. Reducing door-to-needle time for tPA use
remains an elusive goal in stroke care. JAMA 2011;
305: 1288–1289.
92. Meretoja A, Strbian D, Mustanoja S, Tatlisumak T,
Lindsberg PJ and Kaste M. Reducing in-hospital delay
to 20 minutes in stroke thrombolysis. Neurology 2012; 79:
306–313.
93. Rai AT, Smith MS, Boo S, Tarabishy AR, Hobbs GR
and Carpenter JS. The ‘pit-crew’ model for improving
door-to-needle times in endovascular stroke therapy: a
Six-Sigma project. J Neurointerv Surg 2016; 8: 447–452.
94. Costalat V, Machi P, Lobotesis K, et al. Rescue, com-
bined, and stand-alone thrombectomy in the manage-
ment of large vessel occlusion stroke using the solitaire
device: a prospective 50-patient single-center study:
timing, safety, and efficacy. Stroke 2011; 42: 1929–1935.
95. Miley JT, Memon MZ, Hussein HM, et al. A multicenter
analysis of ‘‘time to microcatheter’’ for endovascular
therapy in acute ischemic stroke. J Neuroimaging 2011;
21: 159–164.
International Journal of Stroke, 0(0)
Sacks et al. 19
96. Sun CH, Ribo M, Goyal M, et al. Door-to-puncture: a
practical metric for capturing and enhancing system
processes associated with endovascular stroke care, pre-
liminary results from the rapid reperfusion registry.
J Am Heart Assoc 2014; 3: e000859.
97. Zaidat OO, Castonguay AC, Gupta R, et al. North
American solitaire stent retriever acute stroke registry:
post-marketing revascularization and clinical outcome
results. J Neurointerv Surg 2014; 6: 584–588.
98. Nogueira RG, Lutsep HL, Gupta R, et al. Trevo versus
Merci retrievers for thrombectomy revascularisation of
large vessel occlusions in acute ischaemic stroke
(TREVO 2): a randomised trial. Lancet 2012; 380:
1231–1240.
99. Saver JL, Jahan R, Levy EI, et al. Solitaire flow restor-
ation device versus the Merci Retriever in patients with
acute ischaemic stroke (SWIFT): a randomised, parallel-
group, non-inferiority trial. Lancet 2012; 380:
1241–1249.
100. Abilleira S, Ribera A, Davalos A, et al. Functional out-
come after primary endovascular therapy or IV thromb-
olysis alone for stroke. An observational, comparative
effectiveness study. Cerebrovasc Dis 2014; 38: 328–336.
101. Goyal M, Almekhlafi MA, Fan L, et al. Evaluation of
interval times from onset to reperfusion in patients
undergoing endovascular therapy in the interventional
management of stroke III trial. Circulation 2014; 130:
265–272.
102. van Heerden J, Yan B, Churilov L, Dowling RJ and
Mitchell PJ. Picture-to-puncture time in acute stroke
endovascular intervention: are we getting faster?
J Neurointerv Surg 2015; 7: 564–568.
103. Nikoubashman O, Probst T, Schurmann K, et al.
Weekend effect in endovascular stroke treatment: do
treatment decisions, procedural times, and outcome
depend on time of admission? J Neurointerv Surg
2017; 9: 336–339.
104. Latchaw RE, Alberts MJ, Lev MH, et al.
Recommendations for imaging of acute ischemic
stroke: a scientific statement from the American Heart
Association. Stroke 2009; 40: 3646–3678.
105. McTaggart RA, Ansari SA, Goyal M, et al. Initial hos-
pital management of patients with emergent large vessel
occlusion (ELVO): report of the standards and guide-
lines committee of the Society of NeuroInterventional
Surgery. J Neurointerv Surg 2017; 9: 316–323.
106. Cho TH, Nighoghossian N, Mikkelsen IK, et al.
Reperfusion within 6 hours outperforms recanalization
in predicting penumbra salvage, lesion growth, final
infarct, and clinical outcome. Stroke 2015; 46:
1582–1589.
107. Struffert T, Deuerling-Zheng Y, Engelhorn T, et al.
Feasibility of cerebral blood volume mapping by flat
panel detector CT in the angiography suite: first experi-
ence in patients with acute middle cerebral artery occlu-
sions. AJNR Am J Neuroradiol 2012; 33: 618–625.
108. Higashida RT, Furlan AJ, Roberts H, et al. Trial design
and reporting standards for intra-arterial cerebral
thrombolysis for acute ischemic stroke. Stroke 2003;
34: e109–e137.
109. The Thrombolysis in Myocardial Infarction (TIMI)
trial. Phase I findings. TIMI Study Group. N Engl J
Med 1985; 312: 932–936.
110. Chesebro JH, Knatterud G, Roberts R, et al.
Thrombolysis in myocardial infarction (TIMI) Trial,
phase I: a comparison between intravenous tissue plas-
minogen activator and intravenous streptokinase.
Clinical findings through hospital discharge.
Circulation 1987; 76: 142–154.
111. Mori E, Yoneda Y, Tabuchi M, et al. Intravenous
recombinant tissue plasminogen activator in acute car-
otid artery territory stroke. Neurology 1992; 42:
976–982.
112. Qureshi AI. New grading system for angiographic evalu-
ation of arterial occlusions and recanalization response
to intra-arterial thrombolysis in acute ischemic stroke.
Neurosurgery 2002; 50: 1405–1414.
113. Khatri P, Neff J, Broderick JP, et al. Revascularization
end points in stroke interventional trials: recanalization
versus reperfusion in IMS-I. Stroke 2005; 36: 2400–2403.
114. Gerber JC, Miaux YJ and von Kummer R. Scoring flow
restoration in cerebral angiograms after endovascular
revascularization in acute ischemic stroke patients.
Neuroradiology 2015; 57: 227–240.
115. Zaidat OO, Lazzaro MA, Liebeskind DS, et al.
Revascularization grading in endovascular acute ische-
mic stroke therapy. Neurology 2012; 79(suppl 1):
S110–S116.
116. Almekhlafi MA, Mishra S, Desai JA, et al. Not all ‘‘suc-
cessful’’ angiographic reperfusion patients are an equal
validation of a modified TICI scoring system. Interv
Neuroradiol 2014; 20: 21–27.
117. Kleine JF, Wunderlich S, Zimmer C and Kaesmacher J.
Time to redefine success? TICI 3 versus TICI 2b reca-
nalization in middle cerebral artery occlusion treated
with thrombectomy. J Neurointerv Surg 2017; 9:
117–121.
118. Abilleira S, Cardona P, Ribo M, et al. Outcomes of a
contemporary cohort of 536 consecutive patients with
acute ischemic stroke treated with endovascular therapy.
Stroke 2014; 45: 1046–1052.
119. Gratz PP, Jung S, Schroth G, et al. Outcome of stand-
ard and high-risk patients with acute anterior circulation
stroke after stent retriever thrombectomy. Stroke 2014;
45: 152–158.
120. Gaha M, Roy C, Estrade L, et al. Inter- and intraobser-
ver agreement in scoring angiographic results of intra-
arterial stroke therapy. AJNR Am J Neuroradiol 2014;
35: 1163–1169.
121. Fiehler J, Cognard C, Gallitelli M, et al. European rec-
ommendations on organisation of interventional care in
acute stroke (EROICAS). Int J Stroke 2016; 11:
701–716.
122. Ogawa A, Mori E, Minematsu K, et al. Randomized
trial of intraarterial infusion of urokinase within 6
hours of middle cerebral artery stroke: the middle cere-
bral artery embolism local fibrinolytic intervention trial
(MELT) Japan. Stroke 2007; 38: 2633–2639.
123. Yoon W, Jung MY, Jung SH, Park MS, Kim JT and
Kang HK. Subarachnoid hemorrhage in a multimodal
International Journal of Stroke, 0(0)
20 International Journal of Stroke 0(0)
approach heavily weighted toward mechanical thromb-
ectomy with solitaire stent in acute stroke. Stroke 2013;
44: 414–419.
124. Herial N, Qureshi M, Khan A, et al. Arterial reocclusion
and distal embolization during endovascular treatment
using new generation stent retrievers in acute ischemic
stroke patients (P4.299). Neurology 2015; 84(14 suppl
P4): 299.
125. Janjua N, Alkawi A, Suri MF and Qureshi AI. Impact
of arterial reocclusion and distal fragmentation during
thrombolysis among patients with acute ischemic stroke.
AJNR Am J Neuroradiol 2008; 29: 253–8.
126. Kerr DM, Fulton RL and Lees KR; VISTA
Collaborators. Seven-day NIHSS is a sensitive outcome
measure for exploratory clinical trials in acute stroke:
evidence from the Virtual International Stroke Trials
Archive. Stroke 2012; 43: 1401–1403.
127. Sajobi TT, Menon BK, Wang M, et al. Early trajectory
of stroke severity predicts long-term functional out-
comes in ischemic stroke subjects: results from the
ESCAPE trial (endovascular treatment for small core
and anterior circulation proximal occlusion with empha-
sis on minimizing CT to recanalization times). Stroke
2017; 48: 105–110.
128. Janssen PM, Visser NA, Dorhout Mees SM, Klijn CJ,
Algra A and Rinkel GJ. Comparison of telephone and
face-to-face assessment of the modified Rankin scale.
Cerebrovasc Dis 2010; 29: 137–139.
129. Muir KW, Ford GA, Messow CM, et al. Endovascular
therapy for acute ischaemic stroke: the pragmatic
ischaemic stroke thrombectomy evaluation (PISTE)
randomised, controlled trial. J Neurol Neurosurg
Psychiatry 2017; 88: 38–44.
130. Khan M, Goddeau RP Jr, Zhang J, Moonis M and
Henninger N. Predictors of outcome following stroke
due to isolated M2 occlusions. Cerebrovasc Dis Extra
2014; 4: 52–60.
131. Alonso de Lecinana M, Kawiorski MM, Ximenez-
Carrillo A, et al. Mechanical thrombectomy for basilar
artery thrombosis: a comparison of outcomes with
anterior circulation occlusions. J Neurointerv Surg
2017; 60: 635–643.
132. Minnerup J, Wersching H, Teuber A, et al. Outcome
after thrombectomy and intravenous thrombolysis in
patients with acute ischemic stroke: a prospective obser-
vational study. Stroke 2016; 47: 1584–1592.
133. Nogueira RG, Zaidat OO, Castonguay AC, et al.
Rescue thrombectomy in large vessel occlusion strokes
leads to better outcomes than intravenous thrombolysis
alone: a ‘real world’ applicability of the recent trials.
Interv Neurol 2016; 5: 101–110.
134. Jansen I, Mulder M, Goldhoorn RJ, et al; on behalf of
the MRCLEAN-Registry investigators (www.mrclean-
trial.org). MR CLEAN registry: a post-trial multicenter
registry of intra-arterial treatment for acute ischemic
stroke in The Netherlands. Presented at the
International Stroke Conference; February 21–24,
2017; Houston, TX.
135. Direct transfer to an endovascular center compared to
transfer to the closest stroke center in acute stroke
patients with suspected large vessel occlusion
(RACECAT). Available at: https://clinicaltrials.gov/
ct2/show/NCT02795962. Accessed April 11, 2017.
136. Legal Protections for Peer Review H-375.962: American
Medical Association; Available at: https://searchpf.ama-
assn.org/SearchML/searchDetails.action?uri¼%
2FAMADoc%2FHOD.xml-0-3167.xml. Accessed May
31, 2012.
137. The Health Care Quality Improvement Act of 1986
(HCQIA), 42 USC §11101 et seq.
138. Suydam S, Liang BA, Anderson S, Weinger MB. Patient
safety data sharing and protection from legal discovery.
In: Henriksen K, Battles JB, Marks ES, Lewin DI, edi-
tors. Advances in Patient Safety: From Research to
Implementation.Volume 3: Implementation Issues.
Advances in Patient Safety. Rockville, MD: Agency for
Healthcare Research and Quality, 2005. Available at:
https://www.ncbi.nlm.nih.gov/books/NBK20558/.
Accessed January 20, 2018.
139. Willner RB. Health Care Quality Improvement Act
(HCQIA) of 1986. What is it? Why was it established?
Is it working? Available at: http://www.peerreview.org/
articles/HCQIQ.htm. Accessed May 31, 2012.
140. H.R. 2—114th Congress: Medicare Access and CHIP
Reauthorization Act of 2015. Available at: http://
www.congress.gov/bill/114th-congress/house-bill/2.
Accessed April 24, 2017.
141. Hirsch JA, Harvey HB, Barr RM, et al. Sustainable
growth rate repealed, MACRA revealed: historical con-
text and analysis of recent changes in Medicare phys-
ician payment methodologies. AJNR Am J Neuroradiol
2016; 37: 210–214.
142. Hirsch JA, Leslie-Mazwi TM, Patel AB, et al. MACRA:
background, opportunities and challenges for the neu-
rointerventional specialist. J Neurointerv Surg 2016; 8:
868–874.
143. Berkhemer OA, Jansen IG, Beumer D, et al. Collateral
status on baseline computed tomographic angiography
and intra-arterial treatment effect in patients with prox-
imal anterior circulation stroke. Stroke 2016; 47:
768–776.
144. Hussein HM, Georgiadis AL, Vazquez G, et al.
Occurrence and predictors of futile recanalization fol-
lowing endovascular treatment among patients with
acute ischemic stroke: a multicenter study. AJNR Am
J Neuroradiol 2010; 31: 454–458.
145. Kim JT, Jahan R and Saver JL.; SWIFT Investigators.
Impact of glucose on outcomes in patients treated with
mechanical thrombectomy: a post hoc analysis of the
solitaire flow restoration with the intention for thromb-
ectomy study. Stroke 2016; 47: 120–127.
146. Institute of Medicine (US) Committee on Quality of
Health Care in America; Kohn LT, Corrigan JM,
Donaldson MS, editors. To Err is Human: Building a
Safer Health System. Washington DC: National
Academies Press, 2000.
International Journal of Stroke, 0(0)
Sacks et al. 21
... Triglycerides are the main component of lipid droplets (LD) stored in hepatocytes and represent "safe" and "inert" storage molecules together with cholesterol esters [20,21]. Conversely, toxic lipids, such as free unesterified cholesterol (FC) along with free fa y acids [22], diacylglycerols [23], and ceramides [24], are toxic to various cellular processes and organelles, even with modest quantitative level increases. Some authors propose hepatic FC as the primary trigger of inflammatory response in a subgroup of MASLD patients, introducing the term "cholesterol-associated steatohepatitis" (CASH) [25]. ...
... Moreover, recent findings reveal the presence of cholesterol crystals within the LD of fa y hepatocytes in individuals diagnosed with MASH induced by a high cholesterol diet [24]. Interestingly, these crystals were absent in patients with simple steatosis despite exhibiting a comparable level of fat accumulation, characterized by similar LD numbers and sizes. ...
Article
Full-text available
Metabolic dysfunction-associated steatotic liver disease (MASLD) involves excessive lipid accumulation in hepatocytes, impacting global healthcare due to its high prevalence and risk of progression to severe liver conditions. Its pathogenesis involves genetic, metabolic, and inflammatory factors, with cardiovascular events as the leading cause of mortality. This review examines the role of lipid-lowering therapies in MASLD, with a particular focus on bempedoic acid, a recently approved cholesterol-lowering agent for hypercholesterolemia and high cardiovascular-risk patients. It explores its potential in liver disease by modulating lipid metabolism and inflammatory pathways based on the most recent studies available. Bempedoic acid inhibits ATP-citrate lyase, reducing cholesterol and fatty acid synthesis while activating AMP-activated protein kinase to suppress gluconeogenesis and lipogenesis. Animal studies indicate its efficacy in reducing hepatic steatosis, inflammation, and fibrosis. Bempedoic acid holds promise as a therapeutic for MASLD, offering dual benefits in lipid metabolism and inflammation. Further clinical trials are required to confirm its efficacy and safety in MASLD patients, potentially addressing the multifaceted nature of this disease.
... Previous experiments have indicated that activated endothelial cells release pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, IL-1β, and IL-8. These cytokines can Zhang attract neutrophils and stimulate the release of NETs [96,97]. Exposing brain endothelial cells to Aβ peptides in a laboratory setting leads to an increase in the expression of cytokine genes, particularly IL-1β. ...
... Depleting B lymphocytes has been shown to lower TGF-β + microglia levels and reduce Trem2, Clec7a, and Itgax expression in the hippocampus, indicating that B lymphocytes may drive chronic microglial dysfunction and amyloid plaque buildup in disease conditions [199]. Tfh cells significantly promote the isotype conversion and antibody secretion of B lymphocytes [96]. The cytokine IL-21, characteristic of Tfh cells, shows a marked elevation in AD patients [200] and has a positive correlation with IgG levels [201]. ...
Article
Full-text available
Alzheimer’s disease (AD) is a serious brain disorder characterized by the presence of beta-amyloid plaques, tau pathology, inflammation, neurodegeneration, and cerebrovascular dysfunction. The presence of chronic neuroinflammation, breaches in the blood-brain barrier (BBB), and increased levels of inflammatory mediators are central to the pathogenesis of AD. These factors promote the penetration of immune cells into the brain, potentially exacerbating clinical symptoms and neuronal death in AD patients. While microglia, the resident immune cells of the central nervous system (CNS), play a crucial role in AD, recent evidence suggests the infiltration of cerebral vessels and parenchyma by peripheral immune cells, including neutrophils, T lymphocytes, B lymphocytes, NK cells, and monocytes in AD. These cells participate in the regulation of immunity and inflammation, which is expected to play a huge role in future immunotherapy. Given the crucial role of peripheral immune cells in AD, this article seeks to offer a comprehensive overview of their contributions to neuroinflammation in the disease. Understanding the role of these cells in the neuroinflammatory response is vital for developing new diagnostic markers and therapeutic targets to enhance the diagnosis and treatment of AD patients.
... The PI3K/Akt signaling pathway is widely present in cells and plays an important role in cell survival, metastasis, metabolism, and angiogenesis [60][61][62]. Its dysregulation is involved in the pathogenesis of various diseases, such as osteoarthritis [63], neurological dysfunction [64,65], diabetes [66], male infertility [67], cancer and others [68][69][70][71][72][73]. Several abnormal cellular behaviors were observed in trophoblasts following the inhibition of PI3K/Akt signaling [74][75][76][77][78][79]. ...
Article
Full-text available
Preeclampsia is a multisystem progressive condition and is one of the most serious complications of pregnancy. Owing to its unclear pathogenesis, there are no precise and effective therapeutic targets for preeclampsia, and the only available treatment strategy is to terminate the pregnancy and eliminate the clinical symptoms. In recent years, non-coding RNAs have become a hotspot in preeclampsia research and have shown promise as effective biomarkers for the early diagnosis of preeclampsia over conventional biochemical markers. PIWI-interacting RNAs, novel small non-coding RNA that interact with PIWI proteins, are involved in the pathogenesis of various diseases at the transcriptional or post-transcriptional level. However, the mechanisms underlying the role of PIWI-interacting RNAs in the pathogenesis of preeclampsia remain unclear. In this review, we discuss the findings of existing studies on PIWI-interacting RNA biogenesis, functions, and their possible roles in preeclampsia, providing novel insights into the potential application of PIWI-interacting RNAs in the early diagnosis and clinical treatment of preeclampsia.
... Considering these circumstances, there is a pressing need for innovative therapeutic approaches for seizure management. Following the emergence of the epilepsy-epileptogenesis paradigm, the focus has shifted toward understanding the mechanistic links between inflammation and epilepsy [16][17][18]. In this context, our research aims to investigate the efficacy of natural products in trimethyltin (TMT)-induced epilepsy models. ...
... The potential for harm from PUCFA supplementation in pregnancy did not become apparent until children were 10 years old. 56 Our efforts need to focus on pursuing the efficacy of currently available interventions for disease modification, understanding the mechanisms of preschool wheezing better to identify additional, novel interventions and ensuring we do not shy away from building in mechanism of action and long-term efficacy into trial designs. ...
Article
Full-text available
Recurrent wheezing in preschool children is heterogeneous and results from numerous genetic and environmental risk factors, which result in the same final clinical manifestation of acute episodes of wheezing but have distinct underlying mechanisms. Effective disease‐modifying approaches, therefore, need to target the pathways driving the symptoms. We have good evidence to show that targeting airway eosinophilia alone in early‐life preschool wheezing and using inhaled corticosteroids is not disease‐modifying. Although airway remodelling develops early in preschool wheezing, the challenge is identifying suitable treatments for structural airway changes. There is increasing evidence for the role of lower airway bacterial infection contributing to wheeze episodes, but clinical trials investigating the impact of targeted antibiotic treatment on disease modification are needed. There is also increasing data supporting an association between lower airway neutrophilia and wheezing in a subgroup of preschool children, but direct causation and the role of neutrophil function remain unknown. Finally, there is encouraging preliminary data for the role of inactivated mixed bacterial lysates in children with non‐allergic, infection‐associated wheeze episodes, but the impact on longer‐term outcomes and their mechanism of action is unknown. This review outlines a range of potential novel targets and approaches that may enable secondary prevention of asthma from preschool wheezing. In parallel, the potential for harm when interventions are introduced indiscriminately is highlighted. Some of the challenges that need to be addressed, including trial designs allowing tailored interventions, the need for non‐invasive biomarkers for targeted interventions, and ensuring extended and long‐term follow‐up after intervention, are highlighted.
... 10 In particular, the L925I mutation has been functionally confirmed in Xenopus oocytes. 47 Our results revealed a high frequency of the L925V mutation in the domain IIS4-IIS5 of the VGSC gene in pyrethroid resistant PRM populations. The L925V/M mutation has been found to be associated with pyrethroid resistance in multiple European PRM populations. ...
Article
Full-text available
BACKGROUND Dermanyssus gallinae, the poultry red mite (PRM), is a worldwide ectoparasite posing significant economic challenges in poultry farming. The extensive use of pyrethroids for PRM control has led to the emergence of pyrethroid resistance. The objective of this study is to detect the pyrethroid resistance and explore its associated point mutations in the voltage‐gated sodium channel (VGSC) gene among PRM populations in China. RESULTS Several populations of D. gallinae, namely CJF‐1, CJP‐2, CJP‐3, CSD‐4 and CLD‐5, displayed varying degrees of resistance to beta‐cypermethrin compared to a susceptible field population (CBP‐5). Mutations of VGSC gene in populations of PRMs associated with pyrethroid resistance were identified through sequencing its fragments IIS4–IIS5 and IIIS6. The mutations I917V, M918T/L, A924G and L925V were present in multiple populations, while no mutations were found at positions T929, I936, F1534 and F1538. CONCLUSION The present study confirmed the presence of extremely high levels of pyrethroid resistance in PRM populations in China, and for the first time detected four pyrethroid resistance mutations in the VGSC gene. Identifying pyrethroid resistance in the field population of PRM in China can be achieved through screening for VGSC gene mutations as an early detection method. Our findings underscore the importance of implementing chemical PRM control strategies based on resistance evidence, while also considering the management of acaricide resistance in the control of PRMs. © 2024 Society of Chemical Industry.
Article
BACKGROUND Early diagnosis of pancreatic ductal adenocarcinoma (PDAC) has been a longstanding challenge. The prognosis of patients with PDAC depends on the stage at diagnosis. It is necessary to identify biomarkers for the detection and differentiation of pancreatic tumors and optimize PDAC sample preparation procedures for DNA and RNA analysis. Most molecular studies are done using paraffin-embedded blocks; however, the integrity of DNA and RNA is often compromised in this format. Moreover, RNA isolated from human pancreatic tissue samples is generally of low quality, in part, because of the high concentration of endogenous pancreatic RNAse activity present. AIM To assess the potential of endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) to obtain specimens from pancreatic neoplasms for subsequent RNA molecular profiling, including next-generation sequencing (NGS). METHODS Thirty-four EUS-FNA samples were included in this study: PDAC (n = 15), chronic pancreatitis (n = 5), pancreatic cysts (n = 14), mucinous cysts (mucinous cystic neoplasia/intraductal papillary mucinous neoplasia) n = 7, serous cystic neoplasms n = 5, and pseudocysts n = 2. Cyst material consisted of cyst fluid and cyst wall samples obtained by through-the-needle biopsy (TTNB). Samples were stored at -80 °C until analysis. RNA purity (A260/230, A260/280 ratios), concentration, and integrity (RIN) were assessed. Real-time polymerase chain reaction was conducted on all samples, and small RNA libraries were prepared from solid mass samples. RESULTS RNA was successfully extracted from 29/34 (85%) EUS-FNA samples: 100% pancreatic adenocarcinoma samples, 100% chronic pancreatitis samples, 70% pancreatic fluid cyst samples, and 50% TTNB samples. The relative expression of GAPDH and HPRT were obtained for all successfully extracted RNA samples (n = 29) including low-quality RNA specimens. Low concentration and nonoptimal RIN values (no less than 3) of RNA extracted from EUS-FNA samples did not prevent NGS library preparation. The suitability of cyst fluid samples for RNA profiling varied. The quality of RNA extracted from mucinous cyst fluid had a median RIN of 7.7 (5.0-8.2), which was compatible with that from solid neoplasms [6.2 (0-7.8)], whereas the quality of the RNA extracted from all fluids of serous cystic neoplasms and TTNB samples had a RIN of 0. CONCLUSION The results demonstrate the high potential of EUS-FNA material for RNA profiling of various pancreatic lesions, including low-quality RNA specimens.
Article
Cancer treatment with anti–PD-1 immunotherapy can cause central nervous system immune-related adverse events (CNS-irAEs). The role of microglia in anti–PD-1 immunotherapy–induced CNS-irAEs is unclear. We found that anti–PD-1 treatment of mice caused morphological signs of activation and major histocompatibility complex (MHC) class II up-regulation on microglia. Functionally, anti–PD-1 treatment induced neurocognitive deficits in mice, independent of T cells, B cells, and natural killer cells. Instead, we found that microglia mediated these CNS-irAEs. Single-cell RNA sequencing revealed major transcriptional changes in microglia upon anti–PD-1 treatment. The anti–PD-1 effects were mediated by anti–PD-1 antibodies interacting directly with microglia and were not secondary to peripheral T cell activation. Using a proteomics approach, we identified spleen tyrosine kinase (Syk) as a potential target in activated microglia upon anti–PD-1 treatment. Syk inhibition reduced microglia activation and improved neurocognitive function without impairing anti-melanoma effects. Moreover, we analyzed CNS tissue from a patient cohort that had received anti–PD-1 treatment. Imaging mass cytometry revealed that anti–PD-1 treatment of patients was associated with increased surface marker expression indicative of microglia activation. In summary, we identified a disease-promoting role for microglia in CNS-irAEs driven by Syk and provide an inhibitor-based approach to interfere with this complication after anti–PD-1 immunotherapy.
Article
Full-text available
Rationale: The benefit of thrombectomy in patients with intracranial large vessel occlusion of the anterior circulation has been shown in selected patients in previous randomized controlled trials, but patients with extended ischemic lesions were excluded in the majority of these trials. TENSION aims to demonstrate efficacy and safety of thrombectomy in patients with extended lesions in an extended time window (up to 12 h from onset or from last seen well). Design: TENSION is an investigator-initiated, randomized controlled, open label, blinded endpoint, European, two-arm, postmarket study to compare the safety and effectiveness of thrombectomy as compared to best medical care alone in stroke patients with extended stroke lesions defined by an Alberta Stroke Program Early Computed Tomography Scan score of 3-5 and in an extended time window. In an adaptive design study, up to 665 patients will be randomized. Outcomes: Primary efficacy endpoint will be clinical outcome defined by the modified Rankin Scale at 90-day poststroke. The main safety endpoint will be death and dependency (modified Rankin Scale 4-6) at 90 days. Additional effect measures include adverse events, health-related quality of life, poststroke depression, and costs utility assessment. Discussion: TENSION may make effective treatment available for patients with severe stroke in an extended time window, thereby improving functional outcome and quality of life of thousands of stroke patients and reducing the individual, societal, and economic burden of death and disability resulting from severe stroke. TENSION is registered at ClinicalTrials.gov (ClinicalTrials.gov Identifier NCT03094715).
Article
BACKGROUND AND PURPOSE: Arterial reocclusion and distal embolization have been identified in previous studies of endovascular treatment in patients with acute ischemic stroke. We determined the rates of reocclusion and distal embolization during endovascular treatment using new generation stent retrievers. MATERIALS AND METHODS: Acute ischemic stroke patients underwent mechanical thrombectomy using new generation stent retrievers without intra-procedural heparin or proximal flow arrest for angiographically demonstrated arterial occlusion. "Distal embolization" was defined qualitatively as appearance of an occlusion on a downstream vessel. "Arterial reocclusion" was defined as subsequent reocclusion of the target vessel after initial recanalization had been achieved. The rates were compared with data from 4 previous prospective acute stroke protocols prior to availability of stent retrievers. RESULTS: The median initial National Institutes of Health Scale Score (NIHSS) for these patients was 14 (range 34-0); mean time from symptom onset to treatment was 5.2 +/- 2.6 hr. Arterial reocclusion occurred in 2.3% of these patients, whereas distal embolization occurred in 24% of the 86 patients. The rates of arterial reocclusion (3.0% versus 1.9%) and distal fragmentation (21.2% versus 30.8%) were similar in patients who did or did not receive IV alteplase prior to thrombectomy. Arterial reocclusion, but not distal embolization, was associated with a lower likelihood of favorable outcome at 1-3 months (P = .05; odds ratio, 3.9; 95% confidence interval, 0.01-0.98) after adjusting for age, initial NIHSS, sex, time to treatment, initial angiographic grade, symptomatic intracranial hemorrhage, and final recanalization. The rates of arterial reocclusion (2.1% versus 2.6% of 86 patients, p=0.8) and distal embolization (35.4% versus 18.4% of 86 patients, p=0.8) were not similar in patients treated with and without stent retrievers. CONCLUSIONS: Arterial reocclusion and distal embolization occur in 16%-18% of acute ischemic stroke patients undergoing mechanical thrombectomy with new stent retrievers without IV heparin or proximal flow arrest. The rates are no higher than those observed in cohorts treated without stent retrievers.
Article
Background & Purpose: Intra-arterial therapy (IAT) is being implemented worldwide as the main treatment option for acute ischemic stroke (AIS). We wondered whether effectiveness and safety results that have been reported in randomized clinical trials can be reproduced in everyday clinical practice. We will report results of the Dutch National post MR CLEAN IAT registry including work flow parameters, primary and secondary outcomes, as well as serious adverse events. Methods: The MR CLEAN Registry is a prospective registry of all patients undergoing IAT for AIS in the Netherlands, started after completion of the MR CLEAN trial in March 2014. Registration was required for reimbursement. A core set was defined, with inclusion criteria similar to those of the MR CLEAN trial, including a proven anterior circulation occlusion and treatment possible withing 6 hours from onset. The primary study outcome is the score on the modified Rankin Scale (mRS) at 90 days. The secondary clinical outcome is NIHSS after 24 to 48 hours. Secondary radiological outcomes include the mTICI score on DSA and final infarct volume and major bleeding on follow up NCCT. We used a propensity weighted and an unadjusted ordinal logistic regression model to compare outcomes in the MR CLEAN Registry core and total dataset with the treatment arm of MR CLEAN. Results: Between March 2014 and August 2016 the inclusion rate of the MR CLEAN Registry has been increasing steadily to an average of 79 (SD 22) per month for a cumulative inclusion of 1548 patients in July 2016 (Figure 1). Conclusions: The MR CLEAN registry data is now being analyzed. Results will be reported at the conference.
Article
Background The effect of endovascular thrombectomy that is performed more than 6 hours after the onset of ischemic stroke is uncertain. Patients with a clinical deficit that is disproportionately severe relative to the infarct volume may benefit from late thrombectomy. Methods We enrolled patients with occlusion of the intracranial internal carotid artery or proximal middle cerebral artery who had last been known to be well 6 to 24 hours earlier and who had a mismatch between the severity of the clinical deficit and the infarct volume, with mismatch criteria defined according to age (<80 years or ≥80 years). Patients were randomly assigned to thrombectomy plus standard care (the thrombectomy group) or to standard care alone (the control group). The coprimary end points were the mean score for disability on the utility-weighted modified Rankin scale (which ranges from 0 [death] to 10 [no symptoms or disability]) and the rate of functional independence (a score of 0, 1, or 2 on the modified Rankin scale, which ranges from 0 to 6, with higher scores indicating more severe disability) at 90 days. Results A total of 206 patients were enrolled; 107 were assigned to the thrombectomy group and 99 to the control group. At 31 months, enrollment in the trial was stopped because of the results of a prespecified interim analysis. The mean score on the utility-weighted modified Rankin scale at 90 days was 5.5 in the thrombectomy group as compared with 3.4 in the control group (adjusted difference [Bayesian analysis], 2.0 points; 95% credible interval, 1.1 to 3.0; posterior probability of superiority, >0.999), and the rate of functional independence at 90 days was 49% in the thrombectomy group as compared with 13% in the control group (adjusted difference, 33 percentage points; 95% credible interval, 24 to 44; posterior probability of superiority, >0.999). The rate of symptomatic intracranial hemorrhage did not differ significantly between the two groups (6% in the thrombectomy group and 3% in the control group, P=0.50), nor did 90-day mortality (19% and 18%, respectively; P=1.00). Conclusions Among patients with acute stroke who had last been known to be well 6 to 24 hours earlier and who had a mismatch between clinical deficit and infarct, outcomes for disability at 90 days were better with thrombectomy plus standard care than with standard care alone. (Funded by Stryker Neurovascular; DAWN ClinicalTrials.gov number, NCT02142283.)
Article
Rationale Early reperfusion in patients experiencing acute ischemic stroke is effective in patients with large vessel occlusion. No randomized data are available regarding the safety and efficacy of endovascular therapy beyond 6 h from symptom onset. Aim The aim of the study is to demonstrate that, among patients with large vessel anterior circulation occlusion who have a favorable imaging profile on computed tomography perfusion or magnetic resonance imaging, endovascular therapy with a Food and Drug Administration 510 K-cleared mechanical thrombectomy device reduces the degree of disability three months post stroke. Design The study is a prospective, randomized, multicenter, phase III, adaptive, blinded endpoint, controlled trial. A maximum of 476 patients will be randomized and treated between 6 and 16 h of symptom onset. Procedures Patients undergo imaging with computed tomography perfusion or magnetic resonance diffusion/perfusion, and automated software (RAPID) determines if the Target Mismatch Profile is present. Patients who meet both clinical and imaging selection criteria are randomized 1:1 to endovascular therapy plus medical management or medical management alone. The individual endovascular therapist chooses the specific device (or devices) employed. Study outcomes The primary endpoint is the distribution of scores on the modified Rankin Scale at day 90. The secondary endpoint is the proportion of patients with modified Rankin Scale 0–2 at day 90 (indicating functional independence). Analysis Statistical analysis for the primary endpoint will be conducted using a normal approximation of the Wilcoxon–Mann–Whitney test (the generalized likelihood ratio test).
Article
Background: While mechanical thrombectomy (MT) has become the standard of care for patients with acute ischemic stroke (AIS) with emergent large-vessel occlusions (ELVO), recently published guidelines appropriately award top-tier evidence to the same selective criteria that were employed in completed clinical trials. We sought to evaluate the safety and effectiveness of MT in patients with AIS with ELVO who do not meet top-tier evidence criteria (TTEC). Methods: We conducted an observational study on consecutive patients with AIS with ELVO who underwent MT at six high-volume endovascular centers. Standard safety outcomes (3-month mortality, symptomatic intracranial hemorrhage) and effectiveness outcomes (3-month functional independence: modified Rankin Scale scores of 0-2) were compared between patients meeting and failing TTEC. Results: The sample consisted of 349 (60%) controls fulfilling TTEC and 234 (40%) non-TTEC patients. Control patients meeting TTEC for MT tended to have higher functional independence rates at 3 months (47% vs 39%; p=0.055), while the rates of symptomatic intracerebral hemorrhage (sICH) were similar (9%) in both groups (p=0.983). In multivariable logistic regression models, adherence to TTEC for MT was not independently related to any safety outcome (sICH: OR 0.71, 95% CI 0.30 to 1.68, p=0.434; 3-month mortality: OR 1.27, 95% CI 0.69 to 2.33, p=0.448) or effectiveness outcome (3-month functional independence: OR 0.81, 95% CI 0.48 to 1.37, p=0.434; 3-month functional improvement: OR 0.73, 95% CI 0.48 to 1.11, p=0.138) after adjusting for potential confounders. Conclusions: Approximately 40% of patients with AIS with ELVO offered MT do not fulfill TTEC for MT. Patients who did not meet TTEC had high rates of good clinical outcome and low complication rates.
Article
Background and purpose: The benefits of mechanical thrombectomy (MT) in basilar artery occlusions (BAO) have not been explored in recent clinical trials. We compared outcomes and procedural complications of MT in BAO with anterior circulation occlusions. Methods: Data from the Madrid Stroke Network multicenter prospective registry were analyzed, including baseline characteristics, procedure times, procedural complications, symptomatic intracranial hemorrhage (SICH), modified Rankin Scale (mRS), and mortality at 3 months. Results: Of 479 patients treated with MT, 52 (11%) had BAO. The onset to reperfusion time lapse was longer in patients with BAO (median (IQR) 385 min (320-540) vs 315 min (240-415), p<0.001), as was the duration of the procedures (100 min (40-130) vs 60 min (39-90), p=0.006). Moreover, the recanalization rate was lower (75% vs 84%, p=0.01). A trend toward more procedural complications was observed in patients with BAO (32% vs 21%, p=0.075). The frequency of SICH was 2% vs 5% (p=0.25). At 3 months, patients with BAO had a lower rate of independence (mRS 0-2) (40% vs 58%, p=0.016) and higher mortality (33% vs 12%, p<0.001). The rate of futile recanalization was 50% in BAO versus 35% in anterior circulation occlusions (p=0.05). Age and duration of the procedure were significant predictors of futile recanalization in BAO. Conclusions: MT is more laborious and shows more procedural complications in BAO than in anterior circulation strokes. The likelihood of futile recanalization is higher in BAO and is associated with greater age and longer procedure duration. A refinement of endovascular procedures for BAO might help optimize the results.
Article
Background and purpose: The trajectory of neurological improvement after stroke treatment is clinically likely to be an important prognostic signal. We compared the accuracy of early longitudinal National Institutes of Health Stroke Scale (NIHSS) measurement versus other early markers of stroke severity post treatment in predicting subjects' 90-day stroke outcome. Methods: Data are from the Endovascular treatment for Small Core and Anterior circulation Proximal occlusion with ESCAPE trial (Endovascular Treatment for Small Core and Anterior Circulation Proximal Occlusion With Emphasis on Minimizing CT to Recanalization Times). Stroke severity was assessed at baseline, 1, 2, 5, 30, and 90 days. Subjects' functional outcome was assessed using the modified Rankin Scale at baseline, 30 days, and 90 days. Group-based trajectory model was used to identify distinct subgroups of longitudinal trajectories of NIHSS measured over the first 2, 5, and 30 days. The accuracy of baseline NIHSS, infarct volume, 24-hour change in NIHSS, infarct volume, and disease severity trajectory subgroups in predicting 90-day stroke outcome were assessed using logistic regression analysis. Results: Group-based trajectory model of the 2-day longitudinal NIHSS data revealed 3 distinct subgroups of NIHSS trajectories-large improvement (41.6%), minimal improvement (31.1%), and no improvement (27.3%) subgroups. Individuals in the large improvement group were more likely were more likely to exhibit good outcomes after 90 days than those in the minimal improvement or no improvement subgroup. Among candidate predictors, the 2-day trajectory subgroup variable was the most accurate in predicting 90-day modified Rankin Scale at 84.5%. Conclusions: Early trajectory of neurological improvement defined by 2-day longitudinal NIHSS data predicts functional outcomes with greater accuracy than other common variables. Clinical trial registration: URL: https://www.clinicaltrials.gov. Unique Identifier: NCT01778335.