Canadian Stroke Best Practice
Recommendations: Secondary Prevention
of Stroke Update 2020
David J. Gladstone, M. Patrice Lindsay, James Douketis, Eric E. Smith ,
Dar Dowlatshahi, Theodore Wein , Aline Bourgoin, Jafna Cox, John B. Falconer,
Brett R. Graham, Marilyn Labrie, Lena McDonald, Jennifer Mandzia, Daniel Ngui,
Paul Pageau, Amanda Rodgerson, William Semchuk, Tammy Tebbutt,
Carmen Tuchak, Stephen van Gaal , Karina Villaluna, Norine Foley,
Shelagh Coutts, Anita Mountain, Gord Gubitz, Jacob A Udell, Rebecca McGuff,
Manraj K.S. Heran, Pascale Lavoie, Alexandre Y. Poppe ; on behalf of the
Canadian Stroke Best Practice Recommendations Advisory Committee, in
collaboration with the Canadian Stroke Consortium*
Abstract The 2020 update of the Canadian Stroke Best Practice Recommendations (CSBPR) for the Secondary Prevention of Stroke
includes current evidence-based recommendations and expert opinions intended for use by clinicians across a broad range of settings.
They provide guidance for the prevention of ischemic stroke recurren ce through the identication and management of modiable vascular
risk factors. Recommendations address triage, diagnostic testing, lifestyle behaviors, vaping, hypertension, hyperlipidemia, diabetes,
atrial brillation, other cardiac conditions, antiplatelet and anticoagulant therapies, and carotid and vertebral artery disease. This update of
the previous 2017 guideline contains several new or revised recommendations. Recommendations regarding triage and initial assessment
of acute transient ischemic attack (TIA) and minor stroke have been simplied, and selected aspects of the etiological stroke workup are
revised. Updated treatment recommendations based on new evidence have been made for dual antiplatelet therapy for TIA and minor
stroke; anticoagulant therapy for atrial brillation; embolic strokes of undetermin ed source ; low-density lipoprotein lowering;
hypertriglyceridemia; diabetes treatment; and patent foramen ovale management. A new section has been added to provide practical
guidance regarding temporary interruption of antithrombotic therapy for surgical procedures. Cancer- associated ischemic stroke is
addressed. A section on virtual care delivery of secondary stroke prevention services in included to highlight a shifting paradigm of care
delivery made more urgent by the global pandemic. In addition, where appropriate, sex differences as they pertain to treatments have been
addressed. The CSBPR include supporting materials such as implementation resources to facilitate the adoption of evide nce into practice
and performance measures to enable monitoring of uptake and effectiveness of recommendations.
*Manraj K.S. Heran and Pascale Lavoie have been added as authors; their information has been added to the author afliations, Conicts of Interest, and Statement of Authorship
sections. Additionally, the supplementary material has been updated with new les. A corrigendum detailing these changes has also been published (doi: 10.1017/cjn.2022.57).
From the Division of Neurology, Department of Medicine, and Regional Stroke Centre, Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre and Sunnybrook Research
Institute, Toronto, Canada (DJG); Heart and Stroke Foundation of Canada, Toronto, Canada (MPL, RMG); Department of Medicine, McMaster University, Hamilton, Canada (JD);
University of Calgary, Cumming School of Medicine, Department of Clinical Neurosciences, Calgary, Canada (EES, SC); University of Ottawa, Department of Neurology, Ottawa,
Canada (DD); Stroke Prevention Clinic, McGill University Health Centre, Montreal, Canada (TW); Stroke Prevention Clinic, Champlain Regional Stroke Network, Ottawa, Canada (AB);
Departments of Medicine and of Community Health and Epidemiology, Dalhousie University, Halifax, Canada (JC); Division of Neurology, Faculty of Medicine, University of British
Columbia, Kelowna, Canada (JBF); Faculty of Medicine, Department of Neurology, University of Saskatchewan, Saskatoon, Canada (BRG); Faculty of Medicine (Neurology), Laval
University, Quebec City, Canada (ML); Heart Health Clinic, St. Marthas Regional Hospital, Antigonish, Canada (LMD); Department of Clinical Neurological Sciences, Western
University, London, Canada (JM); Dept of Family Medicine, University of British Columbia, Vancouver, Canada (DN); The Ottawa Hosp ital, Department of Emergency Medicine,
Ottawa, Canada (PP); Provincial Rehabilitation Unit, Queen Elizabeth Hospital, Charlottetown, Canada (AR); College of Pharmacy, University of Saskatchewan, Regina, Canada (WS);
District Stroke Centre, Waterloo Wellington, Kitchener, Canada (TT); Division of Physical Medicine and Rehabilitation, University of Alberta, Edmonton, Canada (CT); Division of
Neurology, Faculty of Medicine, University of British Columbia, Vancouver, Canada (SvG); Vancouver Stroke Program, Vancouver, Canada (KV); WorkHORSE Consulting Inc.,
London, Canada (NF); Queen Elizabeth II Health Sciences Centre, Nova Scotia Rehabilitation Centre Site, Halifax, Canada (AM); Queen Elizabeth II Health Sciences Centre, Stroke
Program, Halifax, Canada (GG); Division of Cardiology, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Canada (JAU); Department of Surgery, Laval
University; Hôpital de l'Enfant-Jésus, Quebec City, Canada (PL); Division of Neuroradiology, University of British Columbia, Vancouver, Canada (MKSH); and Department of
Neurosciences, Centre hospitalier de lUniversité de Montréal (CHUM), Montréal, Canada (AYP)
R
ECEIVED APRIL 5, 2021. FINAL REVISIONS SUBMITTED MAY 14, 2021. DATE OF ACCEPTANCE MAY 15, 2021.
Correspondence to: Dr. M. Patrice Lindsay, RN, PhD, FWSO, Senior Editor, Canadian Stroke Best Practice Recommendations, Director, Health Systems Change, Heart and Stroke
Foundation, Toronto, Canada. Email: patrice.lindsay@heartandstroke.ca
REVIEW ARTICLE
COPYRIGHT ©HEART AND STROKE FOUNDATION OF CANADA, 2021. PUBLISHED BY CAMBRIDGE UNIVERSITY PRESS ON BEHALF OF CANADIAN NEUROLOGICAL SCIENCES FEDERATION.
THIS IS AN OPEN ACCESS ARTICLE, DISTRIBUTED UNDER THE TERMS OF THE CREATIVE COMMONS ATTRIBUTION-NONCOMMERCIAL-NODERIVATIVES LICENCE (HTTP://CREATIVECOMMONS.ORG/LICENSES/BY-
NC-ND/4.0/), WHICH PERMITS NON-COMMERCIAL RE-USE, DISTRIBUTION, AND REPRODUCTION IN ANY MEDIUM, PROVIDED THE ORIGINAL WORK IS UNALTERED AND IS PROPERLY CITED.THE WRITTEN PERMISSION
OF
CAMBRIDGE UNIVERSITY PRESS MUST BE OBTAINED FOR COMMERCIAL RE-USE OR IN ORDER TO CREATE A DERIVATIVE WORK .
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES 315
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
RÉSUMÉ : La mise à jour 2020 des Recommandations canadiennes pour les pratiques optimales de soins de lAVC, relatives à la prévention
secondaire. La mise à jour 2020 des Recommandations canadiennes pour les pratiques optimales de soins de lAVC, relatives à la prévention secondaire
de ce type de trouble comprend des recommandations fondées sur les dernières données probantes ainsi que des avis dexperts courants, émis à lintention
des cliniciens pratiquant dans divers milieux de soins. Les lignes de conduite présentées dans ces recommandations visent la prévention de futurs accidents
vasculaires cérébraux (AVC) ischémiques, et ce, par la reconnaissance et la prise en charge de facteurs de risque vasculaire modiables. Léquipe de
travail sest ainsi penchée sur le triage, les examens de diagnostic, le mode de vie, le vapotage, lhypertension, lhyperlipidémie, le diabète, la brillation
auriculaire et autres troubles cardiaques, les traitements antiplaquettaire et anticoagulant ainsi que latteinte des artères carotides et vertébrales. La mise à
jour des lignes directrices de 2017 contient plusieurs recommandations nouvelles ou révisées. Celles relatives au triage et à lévaluation initiale des
accidents ischémiques transitoires (AIT) aigus et des petits AVC ont été simpliées, et certains aspects du bilan étiologique des AVC, révisés. La mise à
jour des recommandations relatives au traitement fondées sur de nouvelles données probantes touche la bithérapie antiplaquettaire pour les AIT et les petits
AVC; lanticoagulothérapie pour la brillation auriculaire; les AVC emboliques de cause indéterminée; labaissement du taux de LDL; lhypertriglycér-
idémie; le traitement du diabète; et la prise en charge de la persistance du foramen ovale. À cela sajoute une nouvelle section qui offre des conseils
pratiques sur larrêt temporaire du traitement antithrombotique en vue dune intervention chirurgicale. Il est également question des AVC ischémiques
associés au cancer. Dans une autre section portant sur la prestation virtuelle des services de prévention secondaire de lAVC, on fait ressortir un
changement de paradigme dans la prestation des soins, changement qui sest imposé plus que jamais avec la pandémie. Les auteurs ont aussi traité des
différences de traitement en lien avec le sexe lorsque cétait pertinent. Enn se greffent aux nouvelles recommandations des ressources didactiques, par
exemple de la documentation sur la mise en œuvre visant à faciliter la transposition des données probantes en des mesures de pratique et de performance
an de rendre possible la surveillance de lapplication et de lefcacité des recommandations.
Keywords: Stroke, Transient ischemic attack, Guidelines, Secondary prevention, Risk assessment, Management
doi:10.1017/cjn.2021.127 Can J Neurol Sci. 2022; 49: 315337
I
NTRODUCTION
Optimizing stroke prevention is a major public health priority.
Stroke remains a leading cause of adult neurological disability
(both physical and cognitive), dementia, and death globally. The
seventh update of the Canadian Stroke Best Practice Recom-
mendations (CSBPR) Secondary Prevention of Stroke guidelines
includes a summary of current evidence-based recommendations
for healthcare professionals. They focus on reducing the risk of
recurrent stroke following an index ischemic stroke or transient
ischemic attack (TIA) and are applicable to patients managed
across a variety of care settings. They emphasize a coordinated
and organized approach to assessment and aggressive risk factor
management. The core elements of integrated and effective
secondary stroke prevention services are included in the supple-
mental material, Appendix Four. Patient management aims to
identify treatable risk factors, apply evidence-based treatment
interventions to minimize risk , provide patient education and
shared decision-making, and encourage patient adherence and
persistence with treatment recommendations.
These recommendations have been developed in collaboration
with the Canadian Stroke Consortium. We collaborate with the
Canadian Cardiovascular Society, Thrombosis Canada, Diabetes
Canada, and Hypertension Canada to ensure alignment of recom-
mendations wherever possible. Those guidelines should be con-
sulted for additional detail and information beyond the scope of
the CSBPR. The Canadian Stroke Best Practice Recommenda-
tions (CSBPR) Secondary Prevention of Stroke 2020 Seventh
Edition module supersedes all recommendations contained in the
CSBPR Secondary Prevention of Stroke 2017 Sixth Edition
module.
G
UIDELINE DEVELOPMENT METHODOLOGY
The CSBPR development and update process follows a
rigorous framework
1,2
and addresses all criteria dened within
the AGREE trust model.
3
The methodology for development and
updates to the CSBPR has previously published
4,5
and detailed
methodology can be found on our Canadian Stroke Best Practices
website at www.strokebestpractices.ca. A broad interdisciplinary
group of experts was convened and participated in reviewing,
drafting, and revising all recommendation st atements, and a panel
of people with lived experience participated in a parallel review
process.
6
Evidence levels were assigned based on the quality of
available evidence and expert opinion. These guidelines have
undergone extensive internal and objective external review and
consensus was achieved for all content. For additional methodol-
ogy and information on these recommendations, including ratio-
nale, system implications, performance measures, knowledge
translation and implementation tools, and an extended summary
of the evidence, please visit https://www.strokebestpractices.ca/
recommendations/secondary-prevention-of-stroke.
S
ECONDARY PREVENTION OF STROKE RECOMMENDATIONS
Section 1: Triage and Initial Diagnostic Evaluation of
Transient Ischemic Attack and Non-Disabling Stroke
An acute TIA or minor stroke is a medical emergency. Initial
management aims to establish an accurate diagnosis, determine
the likely etiology, and institute secondary prevention therapy as
quickly as possible. Patients with acute TIA or minor stroke are at
risk of recurrent stroke both in the short-term (particularly within
the rst week)
7
and long-term.
8
Our triage recommendations
have been simplied to focus on patients presenting within the
rst 48 h of a suspected new acute TIA or stroke as they are at
highest risk of early recurrent stroke. For such patients, immedi-
ate assessment is recommended, with imaging of both brain (head
computerized tomography [CT] or magnetic resonance imaging
[MRI]) and vessels (ideally with a CT angiogram from aortic arch
to vertex) on an urgent basis.
9
An embolic stroke or TIA can be the rst manifestation of
previously unrecognized atrial brillation. We recommend a
tiered approach to searching for atrial brillation in patients with
a new acute embolic ischemic stroke or TIA.
10
The goal of
post-stroke electrocardiogram (ECG) monitoring is to detect
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
316
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
high-burden atrial brillation for which anticoagulation would
likely be benecial. However, ECG monitoring often reveals
brief subcl inical paroxysmal atrial brillation, and it remains
unclear what amount of device-detected atrial brillation war-
rants anticoagulation. Trials underway are evaluating this ques-
tion. The effect of post-stroke prolonged ECG monitoring on
hard clinical outcomes (i.e., recurrent stroke) remains to be
determined and is the subject of ongoing research (FIND-AF2
trial, NCT04371055).
Echocardiography can be a valuable tool in the etiological
assessment and risk stratication of patients with stroke and TIA.
However, it can be overutilized and we recommend responsible
use of this resource. Thus, the recommendations emphasize that
echocardiography is not required for all stroke patients but should
be considered for those with an embolic ischemic stroke or
TIA of undetermined source (ESUS) or when a cardioembolic
etiology or paradoxical embolism is suspected.
We have recommended against extensive thrombophilia testing
for hereditary hypercoagulable disorders in the routine investiga-
tion of adults with arterial ischemic stroke events. Such testing is
often overused in practice and should be limited to selected patients
such as those with unexplained cerebral venous thrombosis or
patent foramen ovale-related (PFO) paradoxical embolism.
An important lesson of the COVID-19 pandemic has been
how essential remote or virtual contact with patients and families
is to providing safe and timely care for stroke patients. In
particular, care for patients living in rural or remote communities
or patients for whom mobility and transport to clinic or hospital
are prohibitive, can be improved via virtual care. Home blood
pressure monitoring is encouraged in accordance with Hyperten-
sion Canada guidelines.
11
Home delivery of ECG patch monitors
that can be self-applied by patients is a welcome option in regions
where it is available. Virtual care interventions can be effective
for blood pressure lowering, improvements in diet, increased
physical activity, drug adherence, and satisfaction with access to
care,
12
reduced HgbA1c, smoki ng cessation,
13
and reduced risk
of cardiovascular events.
14
Section 1 Recommendations 2020
1.0 Patie nts with acute stroke or transie nt ischemic attack
(TIA) who present to an ambulatory setting (such as primary
care) or a hospital should undergo clinical evaluation by a
healthcare professional with expertise in stroke care to deter-
mine risk for recurrent stroke and initiate appropriate and
timely investigations and management strategies.
1.1 HIGH Risk for Recurrent Stroke (Symptom onset
within last 48 h)
i. Individuals presenting within 48 h of symptoms consistent
with a new acute stroke or TIA event (especially transient
focal motor or speech symptoms, or persistent stroke
symptoms) are at the highest risk for recurrent stroke
and should be immediately sent to an emergency department
(refer to Clinical Consideration 1.1.3) with capacity for
stroke care (including onsite brain imaging, and ideally
access to acute stroke treatments) [Evidence Level B].
ii. Urgent brain imaging (CT or MRI) with concurrent
neurovascular imaging (e.g., CT angiography [CTA])
should be completed as soon as possible and before dis-
charge from the emergency department [Evidence Level B].
iii. Patients presenting after 48 h from the onset of an acute
stroke or TIA event should receive a comprehensive
clinical evaluation and investigations as soon as possible
by a healthcare professional with stroke expertise
[Evidence Level B].
Section 1.1 Clinical Considerations:
1. Referral to a healthcare professional with expertise in
stroke care should be considered for patients with a
suspected uncommon cause of stroke, including for
young stroke patients (e.g., <45 years);
15
family history
of young-onset stroke; suspected cerebral vasculitis or
other intracranial vasculopathy; or suspected hereditary
or acquired thrombophilia.
2. Patients presenting with symptoms of vertebrobasilar
ischemia may present with uctuating brainstem/cerebel-
lar type symptoms (e.g., diplopia, dysarthria, dysphagia,
non-positional vertigo, ataxia; rarely as isolated symp-
toms) over a longer time course (i.e., more than 48 h) and
can be mistaken for stroke mimics; however, they also
require urgent assessment, neurovascular imaging and
management as these types of strokes can have a high
morbidity. Consultation with a healthcare professional
with expertise in stroke care is strongly encouraged.
3. Setting: In some regions, urgent/rapid TIA clinics are
available that have rapid access to diagnostic services,
and they may be considered as appropriate referral
options for TIA and minor stroke patients where avail-
able and accessible.
1.2 Brain and Vascular Imaging
i. Brain imaging (CT or MRI) and non-invasive vascular
imaging (CTA or MR Angiogram [MRA] from aortic
arch to vertex) should be completed as soon as possible
following acute stroke or TIA [Evidence Level B].
a. CTA of head and neck (from aortic arch to vertex),
which can be performed at the time of initial brain CT,
is recomm ended as an ideal way to assess both the
extracranial and intracranial circulation [Evidence
Level B]. Note: Some facilities may not have CTA
readily available; the timing and type of vascular
imaging will need to be based on available resources
and local practice protocols.
b. Neurovascular imaging is recommended to identify
patients with signicant symptomatic extracranial
carotid artery stenosis (i.e., 50%99% stenosis),
which should trigger an urgent referral for potential
carotid revascularization [Evidence Level A].
c. CTA is the rst-line vascular imaging test for stroke/
TIA patients. MRA and carotid ultrasound (for extra-
cranial vascular imaging) are reasonable alternatives
to CTA as rst-line tests for assessment of carotid
vessels if CTA is not possible, and selection should be
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 317
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
based on availability and patient characteristics [Evi-
dence Level C].
Section 1.2 Clinical Considerations:
1. Brain MRI is superior to a head CT scan in terms of
diagnostic sensitivity for identifying small ischemic
lesions in patients presenting clinically with a TIA or
minor stroke event and can provide add itional informa-
tion for guiding diagnosis, prognosis, and treatment
decision-making. Decisions regarding MRI scanning
should be based on MRI access, availability, and timing
of appointments. For maximal diagnostic yield, MRI
should be completed as soon as possible after the symp-
tomatic event, ideally within 7 d of symptom onset. MRI
is particularly useful in lowe r risk patients with transient
symptoms in whom the presence of ischemia would
change their management.
2. Common scenarios where urgent brain MRI can be
valuable include:
a. Normal CT head despite symptoms persisting >24 h
(if diffusion-weighted imaging [DWI]-MRI is negative,
cerebral ischemia is unlikely).
b. Suspected brainstem or cerebellar ischemia (CT head
is insensitive for detecting strokes in the posterior
fossa due to bone artifact).
c. Foca l transient symptoms that are clinically atypical
for ischemia.
1.3 Blood Work
i. The following laboratory investigations should be rou-
tinely considered for patients with a TIA or minor ische-
mic stroke as part of the init ial evaluation:
a. Initial bloodwork: hematology (complete blood
count), el ectrolytes, coa gulation ( aPTT, INR), renal
function (creatinine, estimated glomerular ltration
rate), random glucose, ALT [Evidence Level C].
Refer to Appendix Two for full list of recommended
lab tests.
b. Additional laboratory tests may be completed during
patient encounter or as an outpatient, including a lipi d
prole (fasting or non-fasting); and screening for
diabetes with either a glycated hemoglobin (HbA1c),
fasting glucose or 75 g or al glucose tolerance test
[Evidence Level C].
c. (New for 2020): If giant cell arteritis is suspected
(e.g., retinal ischemia or headache), erythrocyte sedi-
mentation rate (ESR) and C-reactive protein (CRP)
should be measured [Evidence Level C].
ii. (New for 2020): Extensive thrombophilia testing for
hereditary hypercoagulable disorders is not recom-
mended for routine investigation of a patient with arterial
ischemic stroke and should be limited to selected situa-
tions (e.g., but not limited to, unexplained cerebral
venous thrombosis; PFO-related paradoxical embolism)
[Evidence Level C].
a. If a hypercoagulable state is suspected, consider
consultation with a healthcare professional with
hematology or thrombosis expertise [Evidence
Level C].
1.4 Cardiac Studies
1.4A Detection of Atrial Fibrillation
i. Patients with suspected ischemic stroke or TIA should
have a 12-lead ECG to assess for atrial brillation,
concurrent myocardial infarction, or structural heart dis-
ease (e.g., left ventricular hypertrophy) as potential causes
or risk factors of stroke [Evidence Level B].
ii. For patients being investigated for an acute embolic
ischemic stroke or TIA, ECG monitoring for 24 h or
more is recommended as part of the initial stroke work-up
to detect paroxysmal atrial brillation in patients who
would be potential candidates for anticoagulant therapy
[Evidence Level A].
iii. For patients being investigated for an embolic ischemic
stroke or TIA of undetermined source whose initial short-
term ECG monitoring does not reveal atrial brillation
but a cardioembolic mechanism is suspected, continuous
ECG monitoring for at least 2 weeks is recommended to
improve detection of paroxysmal atrial brillation in se-
lected patients aged 55 years who are not already receiv-
ing anticoagulant therapy but who would be potential
candidates for anticoagulant therapy [Evidence Level A].
iv. (New for 2020): For patients aged >65 years with
ischemic stroke or TIA, pulse palpation or heart auscul-
tation or ECG rhythm strip is recommended to screen for
undiagnosed atrial brillation [Evidence Level B].
1.4B Echocardiography
i. Echocardiography should be considered for patients with
an embolic ischemic stroke or TIA of undetermined
source or when a cardioembolic etiology or paradoxical
embolism is suspected [Evidence Level C]. Routine
echocardiography is not required for all stroke patients.
[Evidence Level C].
ii. (New for 2020): For patients aged 60 years or younger
who are being investigated for an embolic ischemic
stroke or TIA of undetermined source, echocardiography
with saline bubble study is recommended for detection of
a possible PFO if it may change patient management (i.e.,
in patients who would be potential candidates for PFO
closure or anticoagulant therapy if a PFO were detected)
[Evidence Level B].
a. Contrast-enhanced (agitated saline) transesophageal
echocardiography or transcranial Doppler has greater
sensitivity than transthoracic echocardiography for
detection of right-to-left cardiac and extra-cardiac
shunts [Evidence Level B].
1.5 Functional Assessment:
i. Patients with stroke should be assessed for neurological
impairments and functional limitations (e.g., cognitive
evaluation, screening for depression, screening for dys-
phagia, screening of tness to drive, need for potential
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
318
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
rehabilitation therapy, and assistance with activities of
daily living) [Evidence Level B].
ii. Patients found to have neurological impairments and
functional limitations should be considered for referral
to the appropriate rehabilitation specialist for in-depth
assessment and management [Evidence Level B].
1.6 Virtual Care for Secondary Stroke Prevention (New
2020)
i. Secondary stroke prevention services should establish
processes and technology to increase and ensure access
to services through virtual care delivery mechanisms for
patients who do not require in-person visits, and espe-
cially patients living in rural and remote settings without
local access to healthcare professionals with stroke ex-
pertise [Evidence Level C].
a. Clinicians should follow established/validated criteria
to determine the best modality for each patient at each
encounter based on the purpose and goals for each
visit [Evidence Level C].
b. Shared decision-making should also take into account
patient values, preferences, health goals, medical
complexity, social determinants of health, and health
needs [Evidence Level C].
Section 1.6 Clinical Considerations:
1. Consulting sites and individual clinicians should have
triage protocols and local intake criteria in place to ensure
patients referred for their services are seen in a timely
manner, especially high-risk patients as described in
section 1.1 of this module.
2. The use of virtual care for stroke prevention should
include decision tools to identify patients who require
in-person visits and those who can reasonably be managed
through virtual care, and a scheduling mechanism for
virtual visits that support a collaborative team approach
to care where appropriate and feasible.
3. A contingency plan should be established to have
patients seen in person in a timely way should the need
arise following a virtual care encounter.
4. Virtual care-enabled evaluations of patients for second-
ary stroke prevention should be modeled on the topics
dened in the post-stroke checklist and core elements of
stroke prev ention care.
5. Validated approaches to virtual neurological exams
should be followed.
6. Barriers to access, equity, and utilization should be
considered and work-around solutions implemented.
7. Ensure processes in place for booking follow-up tests,
referrals, and other consultations following a virtual care
visit.
8. Ensure appropriate documentation and communication
to other team members who may also be involved in care
remotely.
9. Encourage patients and their families to acquire home
blood pressure monitors where appropriate and provide
education or reliable resources on proper use. Mechan-
isms should be in place for follow-up and management of
BP for patients using home BP devices, by either primary
care provide rs or Stroke Prevention Services (SPS).
10. For timely investigations, consider use of prolonged
cardiac monitors, if available, that can be sent to patients
homes and self-applied, then returned by mail.
11. Data collection and quality improvement mechanisms
should be in place to monitor efciency, effectiveness,
and quality of virtual care encounters.
Section 2: Lifestyle Behaviors and Risk Factor Management
A healthy lifestyle, which includes a Mediterranean or Dietary
Approaches to Stop Hypertension (DASH) diet, exercise, weight
control, reduction, and avoidance of alcohol and tobacco, reduces
the risk of an initial stroke and the risk of a subsequent stroke for
patients with a prior history of stroke. Although individually,
these habits can reduce the risk of stroke, their impact is greater
when combined. Even greater impacts can be achieved with
population level interventions for physical activity include invest-
ments in health promoting infrastructure (e.g., sidewalks, walking
paths, bike lanes). At the core of these of interventions is a focus
on making the healthy choice the easy choice.
Section 2 Recommendations 2020
2.1 Risk Factor Assessment:
i. Per sons at risk of stroke and patients who have had a
stroke or TIA should be assessed for vascular disease risk
factors, lifestyle management issues (diet, sodium intake,
exercise, weight, alcohol intake, smoking), as well as use
of oral contraceptives or hormone replacement therapy
[Evidence Level B].
ii. Persons at risk of stroke or TIA and their family members
should receive individualized information and counsel-
ling about possible strategies to modify their lifestyle and
vascular risk factors [Evidence Level B].
iii. Referrals to appropriate specialists should be made to
support and manage specic vascular risk factors and
lifestyle behaviors and ch oices where required [ Evi-
dence Level B].
2.2 Healthy Balanced Diet
i. Counsel and educate individuals with TIA or stroke to
follow a healthy eating pattern and balanced diet [Evi-
dence Level B] or refer to a registered dietitian where
available [Evidence Level C].
ii. Couns el and educate individuals with TIA or stroke to
follow a Mediterranean-type or Dietary Approach to Stop
Hypertension (DASH) diet, which is high in vegetables,
fruit, whole grains, sh, nuts and olive oil and low in red
meat [Evidence Level B].
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 319
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
iii. Counselling may include:
a. consuming a variety of natural, whole, and minimally
processed foods at each meal [Evidence Level B].
b. consuming fewer highly processed foods, which in-
clude rened foods, confectionaries, sugary drinks,
processed meats and meat alternatives, and pre-pre-
pared foods [Evidence Level B].
c. consuming a diet high in vegetables and fruit; encourage
patients to choose fresh or frozen unsweetened fruit, or
fruit canned in water without added sugars and low in
sodium; fresh or frozen vegetables without added
sauces, or canned vegetables with no added salt [Evi-
dence Level B].
d. consuming lower fat and lower sugar dairy products
and unsweetened fortied soy beverages [Evidence
Level B].
e. shift to consuming more protein from plant-based
sources (legumes, nuts, and seeds) and other protein
options which are lower in saturated fats such as sh,
poultry, and lean meats [Evidence Level B].
f. consuming high ber choices such as whole grains,
beans, and legumes instead of processed or rened grains
such as white bread and pasta [Evidence Level B].
g. consuming water as the drink of choice for hydration.
Sugary drinks (such as energy drinks, fruit drinks,
juice, soft drinks, and avored coffees) add calories
and have little to no nutritional value and should be
discouraged [Evidence Level A].
h. consuming foods low in sodium [Evidence Level B].
Section 2.2 Clinical Consideration
1. Counsel and educate individuals regarding healthy eating
patterns that focus on whole, natural, minimally pro-
cessed foods, instead of specic nutrients such as dietary
cholesterol.
2.3 Sodium Intake
i. To prevent hypertension and to reduce blood pressure in
patients with hypertension, counsel and educate indivi-
duals with TIA or stroke to reduce sodium intake to a goal
of no more than 2000 mg (5 g table salt or 87 mmol
sodium, equal to less than one teaspoon) per day [Evi-
dence Level A].
Section 2.3 Clinical Consideration
i. Achieving a sodium intake of <2000 mg may be very
difcult for the general population and average daily
intake among people in Canada is 2760 m g. Enco urage
a gradual decrease in foods that are high in sodium
which will allow taste buds and behavior to adapt
appropriately.
2.4 Physical Activity
i. Counsel and educate individuals with TIA or stroke to
reduce sedentary behaviors and sedentary time, and to
work toward increased activity goals as tolerated [Evi-
dence Level B].
ii. Mo st individuals post-stroke who are medically stable
should start a regular exercise program [Evidence
Level B].
iii. Counsel and educate individuals with TIA or stroke to
participate in aerobic exercise 4 to 7 d per week, to
accumulate at least 150 min per week in episodes of
10 min or more, in addition to routine activities of daily
living [Evide nce Level B].
iv. Initiation of aerobic training should be considered after a
stroke or TIA once the patient is medically stable. To
ensure continuity of appropriate interventions, patients
should be reassessed at transition points along the con-
tinuum of care based on changing neuromotor and car-
diopulmonary capacities to participate in aerobic training
[Evidence Level B].
Section 2.4 Clinical considerations
1. Aerobic exercise intensity should be individualized.
Factors to consider include functional limitation, co-
existing medical problems such as cardiac disease, need
for an exercise stress test with electrocardiogram, and
planned exercise intensity (i.e., light, moderat e, or
vigorous).
2. Screening and supervision of adu lts with comor bid
disease such as cardiac disease which places them at
higher risk of medical comp lications should be
considered.
3. Supervision by a healthcare professional (such as a
physiotherapist) at exercise initiation should be consid-
ered in individuals with stroke at risk of falls or injury.
2.5 Weight Management
i. Counsel and educate individuals with TIA or stroke to
achieve and maintain a waist circumference of <88 cm
for women and <102 c m for men*, or a body mass
index (BM I) of 18.524.9 kg/m2 [Evidence Level B].
(*Note: these numbers a re reective of current research
based mostly on Caucasian patients. Refer to Reference
list for waist circumference values for other ethnic
groups.)
ii. Couns el and educate individuals with TIA or stroke who
are overweight to set healthy weight loss goals and
develop individualized plans to achieve goals [Evidenc e
Level B].
iii. A multi-pronged approach should be used to support
sustainable weight loss or weight gain that includes
counselling and education, increased physical activity,
and behavioral interventions [Evidence Level B].
Section 2.5 Clinical Consideration
1. When discussing weight, consider completion of a com-
prehensive history that explores root causes of weight
gain and avoids stigma and judgment.
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
320
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
2.6 Alcohol Consumption
i. Counsel and educate individuals with TIA or stroke to
avoid heavy alcohol use as excessive alcohol intake
increases the risk of hypertension, ischemic stroke and
intracerebral hemorrhage. [Evidence Level B].
ii. Counsel and educate individuals with TIA or stroke to
follow Canadas Low-Risk Alcohol Drinking Guidelines
(2018): for women, no more than 10 drinks per week,
with no more than 2 drinks per day most days, and no
more than 3 drinks on any single occasion; for men, no
more than 15 drinks per week, with no more than 3 drinks
per day most days, and no more than 4 drinks on any
single occasion [Evidence Level B].
Note: one standard drink is considered to be approxi-
mately 44 mL (1.5 oz) of 80 proof (40%) spirits, 355 mL
(12 oz) of 5% beer or 148 mL (5 oz) of 12% wine.
2.7 Recreational Drug Use
i. Individuals with stroke and known recreational drug use
that may increase the risk of stroke (such as cocaine,
amphetamines) should be counseled to discontinu e use
[Evidence Level C]; and should be provided with appro-
priate support and referrals to services and resources for
drug addiction and rehabilitation [Evidence Level B].
ii. For cannabis, that may be prescribed for medical indica-
tions, counsel patients regarding any potential increased
risk of stroke to support informed decision-making re-
garding the use of these agents [Evidence Level B].
Section 2.7 Clinical Consideration
1. At present, there has been some association of smoking
cannabis products with possible increased stroke and
cardiovascular events. However, there is a lack of
high-quality evidence to provide clear gui dance. Individ-
ual patient factors should be considered.
2.8 Smoking Cessation
Note, the term Smoking in these recommendations
refers to tobacco and other inhaled substances.
i. In all healthcare settings along the stroke continuum
(inpatient, ambulatory, and community), patient smoking
status should be identied, assess ed, and documented
[Evidence Level A].
ii. Provide unambiguous, non-judgmental, and patient-spe-
cic advice regarding the importance of cessation to all
smokers [Evidence Level B] and others who reside with
the patient.
iii. Offer assistance with the initiation of a smoking cessa-
tion attempt either directly or through referral to
appropriate resources [Evidence Level A].
iv. A stepwise approach that starts with reduction in smok-
ing and progresses to full cessation is a valid approach
[Evidence Level B].
v. A combination of pharmacotherapy and behavioral ther-
apy should be considered in all smoking cessation pro-
grams and interventions [Evidence Level A].
vi. The three classes of pharmacological agents that should
be considered as rst-line therapy for smoking cessation
are nicotine replacement therapy, varenicline, and bupro-
pion [Evidence Level A].
a. The choice of appropriate pharmacotherapy should
take into account the patients medical stability, clini-
cal needs, other medical factors, patient preferences,
and patients ability to afford the therapy in those
cases in which it is not covered under a provincial
drug formulary [Evidence Level C].
b. The initiation of pharmacotherapy for smoking cessa-
tion should begin as soon as possible and supported
while in hospital for index stroke-related event
[Evidence Level C]. Earlier initiation of smoking cessa-
tion discussions may be benecial [Evidence Level C].
vii. For stroke patients in hospital who are current smokers,
protocols should be in place to manage nicotine with-
drawal during hospitalization [Evidence Level B].
viii. Interdisciplinary team members should counsel
patients, family members, and caregivers about the
harmful effects of exposure to environmental (second
hand) smoke [Evidence Level B].
ix. A referral to virtual smoking cessation services, smoking
cessation programs, supportive resources, and clinics
should be considered depending on regional availability
to optimize the success of smoking cessation [Evidenc e
Level B]
x. People who are not ready to quit should be offered a
motivational intervention to help enhance their readiness
to quit [Evidence Level B].
Section 2.8 Clinical Considerations
Use of E-Cigarettes
1. Although some individuals may nd vape products
helpful in smoking cessation, the evidence base around
their population-based effe ctiveness is not clear.
2. There is some evidence that shows people who use
vaping as a mechanism to quit cigarettes may continue
to vape even after cessation of cigarette use, in contrast to
use of nicotine replacement therapy which has not been
found to be continued in an ongoing basis.
16
3. Emerging evidence indicates an association between
vaping and elevated blood pressure; the strength of the
association is not clear at this time.
4. Th e most common pattern of u se in Canada is dua l
use of both vape and combustible tobacco products
and therefore smoking cessation strategies should
include consideration for both method s of n icotine
consumption.
5. Education and counselling should be provided regarding
the risks versus benets of e-cigarettes in people with
stroke, including in younger age groups who have expe-
rienced stroke.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 321
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
2.9 Pregnancy, Oral Contraceptives, and Hormone
Replacement Therapy
i. Discussions of pregnancy and implications for stroke
recurrence should be included as a routine part of post-
stroke management for all female stroke survivors of
reproductive age [Evidence Level C].
ii. Contraception should be addressed base d upon the
patients fertility and pregnancy plans as well as the
stroke mechanism and type [Evidence Level C].
iii. In cases of ischemic strok e, systemic estrogen-contain-
ing contraceptives or hormone replacement therapy
that can increase the risk of thrombosis should be
carefully considered and, in most cases, should be
avoided due to an increased risk of stroke [Evidence
Level B].
iv. Management alternatives, including progesterone-only
oral contraceptives, progesterone-only or non-hormonal
intrauterine devices, or barrier contraception can be
considered in consultation with a provider experienced
with contraceptive methods [Evidence Level C].
v. Estrogen -containing oral contraceptives or hormo ne
replacement therapy should be discouraged or
discontinued in female patients with TIA or ischemic
stroke [Evidence Level B]. Management alterna-
tives should be considered in these patients [Evidence
Level C].
vi. (New for 2020) Contraceptive management alternatives
to estrogen containing hormonal contraceptives should
be considered for women with a history of migraine with
aura [Evidence Leve l C], especially if they are also
current tobacco smokers [Evidence Level B].
17
vii. Hypertensive Disorders of Pregnancy: Discussion
on the use and dose of acetylsalicylic acid (ASA) to
reduce the risk of a hypertensive disorder of pregnan-
cy (HDP) should be individualized based upon a
womans risk of HDP (i.e., women with a prior
ischemic stroke, prior HDP or other risk factors) and
in consultation with obstetrical care providers [Evi-
dence Level C]. Refer to CSBPR Stroke during Preg-
nancy recommendations for additional information.
viii. Invi tro Fert ilization: For women who have had a
cerebral event and are considering invitro fertiliza-
tion, provide counselling and education about risks
of fertility t herapy including the potential risk of
hyperstimulation, and monitor for complications
assuming all other stroke in the young management
plans followed and optimized [Evidence Level C].
2.10 Adherence to individual prevention plans
i. At each healthcare encounter, discuss and document
patient adherence to their prescribed secondary preven-
tion treatment plans (pharmacotherapy and lifestyle
changes), explore and address non-adherence, and pro-
vide counselling and engage in joint goal setting to
encourage adherence and persistence with treatment
[Evidence Level C].
2.11 Emerging Risk Factors
Inuenza infection, vaccination, and stroke risk
i. Inuenza vaccination is recomm ended as it has been
shown to be associated with a decreased risk of stroke
or cardiovascular events, particularly in patients with pre-
existing cardiovascular risk factors [Evidence Level B].
Air pollution and stroke risk
i. Counsel individuals regarding long-term exposure to air
pollutants, particularly avoiding or minimizing exposure
to particulate matter 2.5 μm in diameter, which may be
associated with an increased risk of stroke and cardiovas-
cular disease [Evidence Level B].
Section 3: Blood Pressur e and Stroke Prevention
Hypertension is the major modiable risk factor for stroke. In
Canada, systolic hypertension is estimated to account for about
45% of the total stroke burden.
18
Although the optimal target
blood pressure to prevent a rst or recurrent stroke has not been
formally established, the current treatment recommendation to
attain a blood pressure of consistently lower than 140/90 mm Hg
for people who have had an ischemic stroke or TIA, can help to
reduce recurrent events. Using the results from a subset of 13
randomized controlled trials (RCTs) that included persons with a
previous history of stroke, Law et al.
19
reported that blood
pressure treatment resulting in a reduction of 10 mm Hg systolic
and 5 mm Hg diastolic was associated with a 34% reduced risk of
recurrent stroke (RR = 0.66, 95% CI 0.560.79). In the RE-
SPECT trial,
20
persons with a history of stroke within the
previous 30 d to three years who were randomized to a standard
treatment group with a target of <140/90 mm Hg or an intensive
treatment group with a target of <120/80 mm Hg, did not have a
signicantly reduced risk of recurrent stroke (HR = 0.73, 95% CI
0.491.11, p = 0.15); however, when these results were incorpo-
rated into an updated meta-analysis, the risk was reduced signi-
cantly with intensive therapy. The number needed to treat to
prevent one recurrent stroke was 67, with an absolute risk
reduction of 1.5%.
Section 3 Recommendations 2020
3.0 Blood pressure should be assessed and managed in all
persons with stroke or TIA [Evidence Level A].
3.1 Blood pressure assessment
i. All persons at risk of recurrent stroke should have their
blood pressure measured routinely [Evidence Level A],
no less than once annually and more frequently based on
individual clinical circumstances [Evidence Level C].
ii. Proper standardized techniques should be followed for
initial and subsequent blood pressure measurement includ-
ing ofce, home, and community testing [Evidence Level
B] as outlined by the Hypertension Canada Guidelines.
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
322
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
iii. Patients found to have an automated ofce measured
resting elevated blood pressure (systolic greater than
135 mm Hg and/or diastolic greater than 85 mm Hg)
should undergo thorough assessment for the diagnosis of
hypertension [Evidence Level C].
a. During an ofce visit for assessment of hypertension
consider taking the average of three blood pressure
measurements conducted in accordance with the
current Hypertension Canada Guidelines [Evidence
Level C]. Refer to Hypertension Canada Algorithm
for Diagnosis of Hypertension, including Home Blood
Pressure Monitoring Targets.
iv. Patients with refractory hypertension should have com-
prehensive investigations for secondary causes of hyper-
tension [Evidence Level B].
v. Patients with hypertension or at risk for hypertension
(in pre-hypertension state or other risk factors) should
receive aggressive risk factor modication, lifestyle
counselling, and lifestyle modication interventions
[Evidence Level B].
3.2 Blood pressure management
i. Strong consideration should be given to the initiation of
antihypertensive therapy after the acute phase of a stroke
or TIA [Evidence Level A].
ii. For patients who have had an ischemic stroke or TIA,
blood pressure lowering treatment is recommended to
achieve a target of consistently lower than 140/90 mm Hg
[Evidence Level B]; this includes individuals with chron-
ic kidney disease.
iii. For patients who have had a small subcortical stroke
(i.e., lacunar stroke), aggr essive blood pressure lower-
ing treatment is reasonable to achieve a systolic target of
consistently lower than 130 mm Hg [Evidence Level B].
iv. In patients with intracerebral hemorrhage, blood
pressure should be aggressively monitored, treated, and
controlled [Evidence Level A] to sustain a target blood
pressure consistently lower than 130/80 mm Hg [Evi-
dence Level B]. Refer to Canadian Stroke Best Practice
Recommendations: Management of Intracerebral Hem-
orrhage module.
v. In patients with stroke and diabetes, blood pressure
lowering treatment is recommended for the prevention of
rst or recurrent stroke to attain a target systolic blood
pressure consistently lower than 130 mm Hg [Evi-
dence Level C] and a target diastolic blood pressure
consistently lower than 80 mm Hg [Evidence Level A].
vi. Randomized controlled trials have not dened the opti-
mal time to initiate blood pressure lowering therapy after
an acute stroke or TIA. Blood pressure lowering treat-
ment should be initiated or modied before discharge
from hospital [Evidence Level B].
vii. Treatment with an angiotensin-converting enzyme
(ACE) inhibitor and thiazide/thiazide-like diuretic com-
bination is recommended [Evidence Level A]. Long-
acting diuretics may be considered over short-acting
[Evidence Level B].*
viii. The use of an ACE inhibitor combined with an angio-
tensin II receptor blockers (ARB) is not recommended
[Evidence Level B].*
ix. Patients who are not started on antihypertensive therapy in
acute care should have arrangements made for follow-up
with primary care or stroke prevention service for ongoing
evaluation and management [Evidence Level C]. Note:
Blood pressure management is the responsibility of all
healthcare team members, and initially stroke patients
may require frequent monitoring (e.g., monthly) until they
achieve target blood pressure levels and optimal therapy
has been established.
Notes: *For recommendation s on specic agents and
sequence of agents in blood pressure management for
the secondary prevention of i schemic stroke, refer to
the current Hypertensio n Ca nada treatment guide-
lines
11
Section 3 Clinical Considerations
1. (New for 2020) For patients wi th a non-revascu lari zed
critical intracranial or extracranial arterial stenosis
who are experiencing neurological symptoms attribut-
ed to hemodynamic (low o w) cerebr al or retin al
ischemia (e.g., orthostatic TIAs), it is reasonable to
aim for higher than usual blood pressure targets ( i.e.,
permissive hypertension), and avoidance of hypoten-
sion, fo r prevention of hemodynamic stroke; if such
patients are asymptomatic, then usual blood pressure
targets should be followed in the post-acute phase of
stroke.
Section 4: Lipid Management
New evidence supports more aggressive lipid management for
secondary stroke prevention. The recommended target low-den-
sity lipoprotein (LDL) cholesterol level has been lowered to
<1.8 mmol/L, from previously recommended targets of LDL <
2.0 mmol/L or 50% LDL reduction. If this target cannot be
achieved with maximum tolerated statin therapy, ezetimibe or a
PCSK9 inhibitor may be added for ischemic stroke patients with
atherosclerotic disease. Clinicians are reminded that lipid lower-
ing therapies are not recommended for secondary prevention of
intracerebral hemorrhage, or for patients with cardioembolic
ischemic stroke (e.g., atrial brillation) in the absence of athero-
sclerotic disease.
The Treat Stroke to Targ et trial studied 2860 patients with
atherosclerotic disease who had an ischemic stroke within the
previous 3 months or a TIA within the previous 15 d. Treatment to
an LDL cholesterol target <1.8 mmol/L, as compared to a target
of 2.32.8 mmol/L, was associated with a lower risk of major
cardiovascular eve nts over a median of 3.5 years (8.5% vs.
10.9%, HR = 0.78, 95% CI 0.610.98; p = 0.04).
21
About a
third of patients in this study required t he addition of ezetimibe
to their high-dose statin to achieve the more aggressive LDL
target.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 323
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
Treatment of hypertriglyceridemia with icosapent ethyl 2 g
bid may be considered for patients with ischemic stroke who have
established atherosclerotic cardiovascular disease, or diabetes
plus additional vascular risk factors, and elevated serum trigly-
cerides (1.5 mmol/L) despite statin therapy.
Section 4 Recommendations 2020
4.0 Individuals who have had an ischemic stroke or TIA
should have their serum lipid levels assessed and optimally
managed [Evidence level A].
4.1 Lipid Assessment
i. Lipid levels, including total cholesterol, triglycerides,
low-density lipoprotein [LDL] cholesterol, and high-den-
sity lipoprotein [HDL] cholesterol, should be measured in
patients presenting with ischemic stroke or TIA [Evi-
dence Level B]. Refer to Appendix Two for more infor-
mation on laboratory tests.
4.2 Lipid Management
i. Individuals with ischemic stroke or TIA should be man-
aged with aggressive lifestyle changes to lower lipid
levels, including dietary modication and exercise, as
part of a comprehensive approach to lower risk of recur-
rent stroke and other vascular events unless contraindi-
cated [Evidence Level B].
ii. Statin pharmacotherapy should be prescribed for
second-
ary prevention of stroke in individuals who have had a
non-cardioembolic ischemic stroke or TIA, [Evidence
Level A].
a. A target LDL cholesterol level of <1.8 mmol/L is
recommended [Evidence Level B].
iii. Statin therapy should not be initiated for secondary pre-
vention of intracerebral hemorrhage [Evidence Level C].
22
iv. Add-on therapies for LDL-Lowering (New 2020):
a. For individuals with ischemic stroke and atheroscle-
rotic cardiovascular disease with an LDL > 1.8 mmol/
L in spite of maximal tolerated statin therapy, ezeti-
mibe may be considered for additional LDL lowering
[Evidence Level B].
b. For individuals wi th concomitant atherosclerotic
cardiovascular dise ase in which target LDL level is
not achievable, consider referral to a health profes-
sional with expert ise in metabolic a nd lipid manage-
ment, or stroke expertise for consideration of adding
PCSK9 inhibitor [Evidence Level A].
v. Add-on therapies for hypertriglyceridemia (New
2020) For ischemic stroke patients with established
atherosclerotic cardiovascul ar disease or diabetes plus
additional vascular risk factors, who have elevated serum
triglyceride levels (1.5 mmol/L) despite statin therapy,
icosapent ethyl 2 g bid may be considered to decre ase the
risk of vascular events [Evidence Level B].
4.3 Statin Intolerance (New 2020)
i. For patients with an intolerance to statins (including
persistent myalgias, persistent signicant liver enzyme
abnormalities or rarely, myopathy or rhabdomyolysis),
the indication for statin therapy should be conrmed and
in general, systematic evaluation of the contribution of
statins to the patients symptoms should be considered
(including temporary statin cessation with observation of
symptoms, dose-adjustment, use of alternate agents)
[Evidence Level C]
Section 5: Diabetes Management in Stroke
In Canada, almost 2.5 million people have type 1 or 2
diabetes.
23
Diabetes is known to increase the risk of ischemic
stroke by 227%.
24
Although tighter glycemic control along with
other risk factor reduction strategies, can collectively help to
reduce stroke risk, on its own, aggressive glycemic control does
not reduce stroke risk.
25,26
However, trials of newer antihyper-
glycemic agents, including SGLT-2 and GLP-1 receptor agonists,
have demonstrated benet for major cardiovascular outcomes,
including stroke.
2731
Section 5 Recommendations 2020
5.0 Patients with diabetes who have had an ischemic stroke or
TIA should have their diabetes assessed and optimally man-
aged [Evide nce Level A].
5.1 Diabetes Screening and Assessment
i. Patients with ischemic stroke or TIA should be
screened
for diabetes with either a fasting plasma glucose, or 2-h
plasma glucose, or glycated hemoglobin (A1C), or 75 g
oral glucose tolerance test in either an inpatient or outpa-
tient setting [Evidence Level C].
ii. For
patients with diabetes and either ischemic stroke or
TIA, glycat ed hemoglobin (A1C) should be considered as
part of a comprehensive stroke assessment [Evidence
Level B].
5.2 Diabetes Management
i. Glyc emic targets should be individualized to achieve:
a. In general, A1C values should be targeted to 7.0% in
patients with either type 1 or type 2 diabetes (and
stroke or TIA), as this target provides strong benets
for the prevention of microvascular complications
[Evidence Level A].
b. To achieve a target of A1C 7.0%, most patients with
type 1 or type 2 diabetes should aim for a fasting
plasma glucose or pre-prandial plasma glucose target
of 4.07.0 mmol/L [Evidence Level B].
c. The 2-h postprandial plasma glucose target is 5.0
10.0 mmol/L [Evidence Level B].
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
324
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
d. If A1C targets cannot be achieved with a postprandial
target of 5.010.0 mmol/L, further postprandial blood
glucose lowering , to 5.08.0 mmol/L, should be con-
sidered [Evidence Level C].
ii. (New 2020) In patients with stroke and type 2 diabetes in
whom glycemic targets are not achieved with standard
oral antihyperglycemic medications, an antihyperglyce-
mic agent with demonstrated benet on major cardiovas-
cular outcomes (e.g., SGLT-2 inhibitors or GLP-1 recep-
tor agonists) should be considered [Evidence Level B].
Section 5.2 Clinical Consideration (New 2020):
1. The Pioglitazone after Ischemic Stroke or TIA trial
32
suggested that while there is a benet of pioglitazone for
stroke prevention in patients with positive insulin resis-
tance, it is offset by the increased risk of fractures and
bladder cancer. A post-hoc analysis of patients in the trial
with prediabetes and good drug adherence suggested a
benet of pioglitazone over placebo with regards to
stroke, acute coronary syndrome, stroke/myocardial in-
farction (MI)/hospitalization for heart failure, and pro-
gression to diabetes. The decision to use this agent could
be considered based on the spec ic risk prole for each
patient.
Section 6.0: Antiplatelet Therapy for Individuals with
Ischemic Stroke or TIA
Short-term administ ration of dual antiplatelet therapy with
aspirin and clopidogrel is recommended for secondary stroke
prevention, starting within 24 h for eligible patients with acute
non-hemorrhagic high-risk TIA or minor ischemic stroke based
on the POINT,
33
CHANCE,
34
and FASTER
35
trials. The optimal
duration of dual antiplatelet therapy has been claried by addi-
tional analyses
36,37
with net benet of dual antiplat elet therapy
over aspirin alone likely conned to the rst 21 d post-TIA/stroke
(maximal within the rst 10 d). Compared with aspirin, the short-
term dual antiplatelet therapy protocol prevents 20 more strokes
(and causes two maj or bleeds) for every 1000 patients treated.
Pharmacogenetic testing can identify patients with clopidogrel
resistance, however, its clinical implications for stroke prevention
practice are unclear at this time.
3840
Another short-term dual antiplatelet treatment option is the
combination of daily low-dose aspirin and ticagrelor, a P2Y12
antagonist most often used in coronary artery disease. The
Acute Stroke or Transient Ischemic Attack Treated With Tica-
grelor and ASA for Prevention of Stroke and Death (THALES)
trial tested a 30-d course of the aspirinticagrelor combination
starting within 24 h of a high-risk TIA or minor ischemic
stroke.
41
Ticagrelor was administered as a 180 mg loading dose
followed by 90 mg twice daily, along with aspirin 75100 mg
daily. This combination reduced the risk of recurrent stroke or
death compared with aspirin alone, although the risk of severe
bleeding, intracranial bleeding, and fatal bleeding was higher in
the ticagreloraspirin group. Maximum benet was observed in
patients with ipsilateral large vessel atherosclerotic disease.
42
The dening features of ESUS are an acute brain infarct
visualized on neuroimaging (not a subcortical lacune <1.5 cm);
absence of proximal atherosclerotic vessel stenosis >50%; no
atrial brillation or other major risk cardioembolic source; and no
other likely cause for the stroke.
43
Patients with ESUS have an
average annual stroke recurrence risk of approximately 5%. Two
trials published since the last edition investigated whether
patients with ESUS would benet more from anticoagulation
than aspirin. Neither trial showed found a signicant reduction in
recurrent stroke risk and therefore anticoagulation is not recom-
mended for patients with ESUS.
44,60
The lack of an overall
benet of anticoagulation likely reects that ESUS comprises
a heterogeneous group of many etiologies, with atherosclerotic or
other mechanisms likely predominating over occult atrial bril-
lation in the patients enrolled in these trials. The ARCADIA trial
(NCT03192215) is testing apixaban versus aspirin in a subset of
ESUS patients who have markers of atrial myopathy.
Section 6 Recommendations 2020
6.1 Acute Antiplatelet Therapy
i. All patients with acute ischemic stroke or TIA not already
on an antiplatelet agent should be treated with at least
160 mg of acetylsalicylic acid immediately as a one-time
loading dose after brain imaging has excluded intracranial
hemorrhage [Evidence Level A].
ii. For patients with dysphagia, acetylsalicylic acid (80 mg
daily) or clopidogrel (75 mg daily) may be administered
by enteral tube or acetylsalicylic acid by rectal supposi-
tory (325 mg daily) [Evidence Level A]. Note: acetylsa -
licylic acid should only be administered orally once
dysphagia screening has been performed and indicates
absence of potential dysphag ia.
iii. Antiplatelet therapy should be started as soon as possible
after brain imaging has excluded hemorrhage, within
24 h of symptom onset (ideally within 12 h) [Evidence
Level B] .
iv. For patients receiving intravenous thrombolysis therapy,
avoid antiplatelet therapy within the rst 24 h; antiplate-
let therapy could then be initiated after brain imaging has
excluded secondary hemorrha ge [Evidence Level B].
v. For TIA or minor ischemic stroke patients who are being
discharged from the emergency department, antiplatelet
therapy should be started prior to discharge [Evidence
Level C].
6.2 Antiplatelet Therapy for Secondary Stroke Prevention
i. For patients with ischemic stroke or TIA, antiplatelet
therapy is recomm ended for long-term secondary stroke
prevention to reduce the risk of recurrent stroke and other
vascular events unless there is an indication for anticoag-
ulant therapy [Evidence Level A].
ii. Antiplatelet therapy should be started as soon as possible
after brain imaging has excluded hemorrhage, within
24 h of symptom onset (ideally within 12 h) [Evidence
Level B].
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 325
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
iii. For long-term secondary stroke prevention, either acet-
ylsalicylic acid (80325 mg daily), or clopidogrel (75 mg
daily), or combined acetylsalicylic acid and extended-
release dipyridamole (25mg/200 mg BID), are all appro-
priate treatment options and selection depends on patient
factors or clinical circumstances [Evidence Level A]
6.2.1 Short-Term Dual Antiplatelet Therapy for Second-
ary Stroke Prevention
iv. For patients with an acute high-risk TIA or minor
ischemic stroke of non-cardioembolic origin (NIHSS
03), who are not at high bleeding risk, dual antiplatelet
therapy is recommended with clopidogrel 75 mg daily
plus acetylsalicylic acid 81 mg daily for a duration of
21 d after the event, followed by antiplatelet monother-
apy thereafter (acetylsalicylic acid or clopidogrel alone)
[Evidence Level A].
v. (Revised for 2020): Dual antiplatelet therapy for longer
than the rst 21 d following a TIA or minor stroke is not
recommended unless there is a specic indication (e.g.,
arterial stent; symptomatic intracranial artery stenosis), due
to an increased risk of bleeding without clear benetbeyond
21 d [Evidence Level B]. Patients should be counseled that
dual antiplatelet therapy with acetylsalicylic acid and clo-
pidogrel should continue for only 21 d, followed by anti-
platelet monotherapy to be continued indenitely.
vi. A single loading dose of clopidogrel (either 300 mg
(CHANCE trial) or 600 mg (POINT trial)) and acetylsa-
licylic acid (160325 mg) should be administered at the
start of treatment [Evidence Level A].
vii. (New for 2020): Another reasonable short-term dual
antiplatelet treatment option is the combination of daily
low-dose acetylsalicylic acid plus ticagrelor (180 mg
loading dose, followed by 90 mg bid) for 30 d [Evidence
Level B].
viii. (New for 2020): For patients with a recent stroke or TIA
due to symptomatic intracranial atherosclerotic stenosis
of 70%99%, and a low estimated bleeding risk, the
SAMMPRIS protocol should be considered, which
includes dual antiplatelet therapy (acetylsalicylic acid
and clopidogrel) for the rst 3 months, typically fol-
lowed by antiplatelet monotherapy thereafter, in addi-
tion to intensive lipid-lowering therapy with high-dose
statin, blood pressure treatment, and structured lifestyle
modication addressing smoking cessation, exercise and
diet [Evidence Level B].
6.2.2 Specic Clinical Situations
ix. (New for 2020): For patients with an embolic stroke of
undetermined source, and no known atrial brillation,
anticoagulant therapy is not currently recommended
over low-dose acetylsalicylic acid for secondary stroke
prevention [Evidence Level A]. Additional trials are
ongoing to investigate this issue.
Section 6.2 Clinical Considerations
1. For patients who experience a stroke while receiving one
antiplatelet agent, stroke etiology should be reassessed
and addressed, and all other vascular risk factors aggres-
sively managed. Either continuing the current agent or
switching to a different antiplatelet agent are reasonable
options. At the present time, evidence is lacking to make
more specic recommendations.
2. (New for 2020): Pharmacogenetic testing can identify
patients with clopidogrel resistance, however, its clinical
implications for stroke prevention treatment are unclear at
this time.
3. (New for 2020): For carefully selected patients with coro-
nary artery disease or peripheral vascular disease meeting
the eligibility criteria of the COMPASS trial, including a
low-estimated bleeding risk and no history of lacunar stroke
or hemorrhagic stroke, the combination of rivaroxaban
2.5 mg BID plus daily low-dose acetylsalicylic acid is a
reasonable treatment option. It should not be used within
the rst month after a stroke event.
Section 7: Anticoagulant Therapy for Atrial Fibrillation
Oral anticoagulant therapy is strongl y recommended for
secondary stroke prevention in patients with atrial brillation.
Anticoagulation for AF has been associated with a 66% relative
risk reduction o f recurrent stroke, with an absolute risk reduc-
tion of 7.3% .
45
Direct oral anticoagulants (DOACs) are gener-
ally preferred over warfarin for most pat ients with non-valvular
atrial brillation (non-valvula r is now d ened as atrial brilla-
tion without moderatesevere mitral stenosis or mechanical
heart valves).
46
A recent trial supports the use of rivaroxaban
over warfarin for patients with atrial brillation and a biopros-
thetic mitral valve.
47
Clinicians are reminded to avoid inappropriate underdosing of
DOACs, a practice that is associated with increased stroke risk.
For patients with atrial brillation and chronic stable coronary
artery disease (or >1-year post-percutaneous coronary interven-
tion [PCI] or coronary artery bypass graft [CABG]), the addition
of an antiplatelet agent to chronic DOAC therapy is not recom-
mended as it increases bleeding risk without providing additional
benet in reducing ischemic events (cardiac or cerebral). The
Atrial Fibrillation and Ischemic Events With Rivaroxaban in
Patients With Stable Coronary Artery Disease Study (AFIRE)
trial showed that rivaroxaban alone was as effective as the
combination of rivaroxaban and aspirin in this patient population,
with a lower incidence of bleeding.
48
Section 7 Recommendations
7.1 Detection of Atrial Fibrillation following Stroke
i. Patients with suspected ischemic stroke or TIA should
have a 12-lead ECG to assess for atrial brillation,
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
326
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
myocardial infarction, or structural heart disease (e.g., left
ventricular hypertrophy) as potential causes or risk factors
of stroke [Evidence Level B].
ii. For patients being investigated for an acute embolic
ischemic stroke or TIA, ECG monitoring for 24 h or
more is recommended as part of the init ial stroke work-
up to detect paro xysmal atrial brillation in patients who
would be potential candidates for anticoagulant thera py
[Evidence Level A].
iii. For patients being investigated for an embolic ischemic
stroke or TIA of undetermined source whose initial short-
term ECG monitoring does not reveal atrial brillation
but a cardioembolic mechanism is suspected, prolonged
ECG monitoring for at least 2 weeks is recommended to
improve detection of paroxysmal atrial brillation in
selected patients aged 55 years who are not already
receiving anticoagulant therapy but would be potential
anticoagulant candidates [Evidence Level A].
iv. (New for 2020): For patients aged >65 years with
ischemic stroke or TIA, routine pulse palpation is
recommended to screen for undiagnosed atrial brilla-
tion [Evidenc e Level C].
7.2 Secondary Stroke Prevention in Patients with Atrial
Fibrillation
i. Patients with ischemic stroke or TIA and atrial brillation
should receive oral anticoagulant therapy for secondary
stroke prevention [Evidence Level A].
a. (New for 2020): For patients with an ischemic stroke
or TIA and atrial brillation, oral anticoagulant therapy
is strongly recommended [Evidence Level A]. It is
recommended over acetylsalicylic acid [Evidence
Level A] and dual antiplatelet therapy [Evidence
level B].
b. For most patients requiring anticoagulants for non-
valvular atrial brillation, a direc t oral anticoagulant
(DOAC) such as apixaban, dabigatran, edoxaban , or
rivaroxaban should be prescribed in preference over
warfarin [Evidence Level A].
c. For patients already receiving warfarin with good
International Normalized Ratio (INR) control (range
2.03.0, wit h time in therapeutic ran ge (TTR) of
>70%) and w ithout adverse effects, continuing war-
farin, rather than switching to a DOAC, is a reason-
able anticoagulant option [Evidence Level B].
Patient preferences should be considered in deci-
sion-making [Evidence Level C].
d. When selecting an oral anticoagulant, patient specic
criteria shou ld be considered [Evidence Leve l C].
ii. For patients with acute ischemic stroke and atrial brillation
who are being started on warfarin, routine use of bridging
with heparin is not recommended [Evidence Level B].
a. Bridging with antiplatelet therapy (e.g., low-dose
acetylsalicylic acid) is suggested until the patient is
anticoagulated within therapeutic range [Evidence
Level C].
iii. For patients with ischemic stroke or TIA and atrial
brillation who are unable to take oral anticoagulant
therapy (DOAC or warfarin), acetylsalicylic acid alone
is recommended unless also contraindicated [Evidence
Level A].
a. For patients at high risk of bleeding, dual antiplatelet
therapy is not recommended in preference to antic-
oagulation as the risks of bleeding are comparable,
and dual antiplatelet therapy is less effective for stroke
prevention [Evidence Level B].
iv. For ischemic stroke or TIA in patients with atrial bril-
lation who cannot receive long-term oral anticoagulant
therapy, a left atrial appendage occlusion procedure may
be considered [Evidence Level B].
v. For patients with a mechanical heart valve, warfarin is
recommended for stroke prevention with carefu l INR
monitoring; direct oral anticoagulants (DOACs) are con-
traindicated [Evidence Level B]. Note, patients with
bioprosthetic heart valves do not routinely require
long-term anticoagulation.
vi. (New for 2020): For patients with atrial brillation who
experience ischemic stroke or TIA in spite of anticoag-
ulant therapy, we recommend the following: (1) identify
and address medication non-adherence; (2) ensure cor-
rect DOAC dosing or warfarin INR control; (3) avoid
DOACs drugdrug int eractions; (4) investigate for and
treat other p otential stroke etiologies; and (5) promote
general vascular risk factor modication [Evidence
Level C].
Section 7.2 Clinical Considerations Revi sed for 2020
Timing of Initiation of Oral Anticoagulant Therapy
following Acute Stroke:
1. The optimal timing to start anticoagulant therapy after
an ischemic stroke has not yet been well de
ned by
clinical trial evidence and should be based on individual
benet/risk assessment taking into account the clinical
circumstances, stroke severity, infarct size, imaging
appearances, risk of hemorrhagic transformation, age,
comorbidities, and estimated stroke recurrence risk.
2. There is a lack of randomized evidence to guide specic
timing. According to expert consensus, a general ap-
proach to the target timing of initiation of DOAC therapy
post-stroke is as follows:
a. For patients with a brief TIA and no visible infar ct or
hemorrhage on imaging, anticoagulation may be
started within the rst 24 h post-TIA.
b. For patients with a minor clinical stroke/small non-
hemorrhagic infarct on imaging, anticoagulation may
be start ed 3 d post-stroke.
c. For patients with a moderate clinical stroke/moderate-
sized infarct on imaging (without hemorrhage on CT),
anticoagulation may be started 67 d post-stroke.
d. For patients with a severe clinical stroke/large-sized
infarct on imaging (wi thout hemorrhage on CT),
anticoagulation may be started 1214 d post-stroke.
3. If anticoagulation is delayed beyond 24 h, it is recom-
mended to obtain repeat brain imaging for reassessment
prior to initiation of anticoagulation to exclude the
presence of asympto matic hemorrhagic transformation
of the index infarct.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 327
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
4. It is reasonable to delay the initiation of anticoagulation
for more than 2 weeks post-stroke if in the judgement of
the clinician the risk of intracranial bleeding is felt to be
high, for example, for some patients with large infarcts
and those with hemorrhagic transformation.
Stroke while on DOAC Therapy
1. (New for 2020): For patients with atrial brillation who
experience ischemic stroke or TIA despite anticoagulant
therapy, either continuing the current agent or switching
to a different anticoagulant agent are reasonable options.
At the present tim e, evidence is lacking to make more
specic recommendations.
2. The routine addition of acetylsalicylic acid to chronic antico-
agulant therapy is not recommended because of increased
bleeding risk without clear evidence of benet and potential
for harm unless there is a specic medical indi cation.
7.3 Enhancing anticoagulant therapy effectiveness in
practice and minimizing bleeding complications.
i. Medication adherence should be continually assessed and
reinforced for patients on all oral anticoagulants at each
follow-up visit [Evidence Level B].
a. Patients who are prescribed a DOAC should be
reassessed at intervals and educated regarding the
short h alf-life o f this class of drugs, the importance
of daily medication adherence and the dangers of
missed doses o r prolonged interruptions of therapy
[Evidence Level C].
b. For patients with atrial brillation taking warfarin,
careful dosing and consistent INR mo nitoring is
recommended to minimize adverse events; warfarin
efcacy is dependent on maintaining therapeu tic INR
control and declines signicantly when the INR falls
below 2.0 [Evidence Level A].
c. Patients and family members should be provided
education, resources, and ongoing monitoring
regarding atrial brillation and adherence to en-
hance compliance and address potential barriers in
a timely way to facilitate self-management [Evi-
dence Level C].
ii. (New for 2020): For patients prescribed DOAC therapy,
avoid inappropriate underdosing as it is associated with
increased stroke risk [Evidence Level C].
iii. For patients prescribed DOACs, creatinine clearance
should be routinely monitored at least once annually, and
when there is a change in health status [Evidence Level C].
a. Dose adjustments or a change in selected agent may
be requ ired based on changes in renal function if
detected [Evidence Level C].
b. More frequent monitoring of renal function (every 6
months or more frequently) may be considered for
patients with renal impairment or a dehydrating illness
for medication adjustment if required, particularly for
patients receiving dabigatran [Evidence Level C].
iv. For patients taking chronic oral anticoagulant therapy
for non-val vular atrial brillation, the addition of
antiplatelet therapy is not recommended due to increased
bleeding risk unless there is a specic medical indication
for antiplatelet therapy (e.g., recent vascular stent; cer-
tain mechanical heart valves) [Evidence Level B].
v. (New for 2020): For patien ts with atrial brillation and
chronic stable coronary artery disease (and >1-year post-
PCI or CABG), the addition of an antiplatelet agent to
DOAC therapy is not recommended as it increases
bleeding risk without providing any signicant benet
in reducing ischemic events (cardiac or cerebral) [Evi-
dence Level B].
Section 8: Perioperative Management of Anticoagulant and
Antiplatelet Therapy (New for 2020)
This edition features a new section on perioperative antithrom-
botic management a commonly encountered issue in the stroke
population and one in which practice variations abound. Our
recommendations are aligned with Thrombosis Canada.
49
For stroke
or TIA patients who require temporary interruption of chronic
antiplatelet or anticoagulant therapy for an upcoming elective
surgery, decisions regarding the duration of therapy interruption
depend on the agent and the estimated bleeding risk associated with
the surgery or procedure. The goal is to minimize the risk of
ischemic stroke while simultaneously minimizing the risk of clini-
cally important (major) bleeding. Patients should avoid unnecessary
or prolonged interruptions of their antithrombotic therapy. Clinicians
should communicate clear instructions to patients regarding their
perioperative management plan before an elective procedure.
Because DOACs have a rapid offset (average half-life of
approximately 12 h) and a rapid onset of action, the duration of
DOAC interruption can be kept short to minimize the risk of
ischemic stroke. This approach of standardized DOAC interruption
and resumption appeared safe in the Perioperative Anticoagulation
Use for Surgery Evaluation (PAUSE) study of 3007 DOAC-
treated patients; the 30-d post-operative rates of arterial thrombo-
embolism and major bleeding were <1% and <2%, respectively.
50
For patients undergoing a minimal-bleed-risk procedure, antic-
oagulants can generally be continued without interruption, with
some caveats; for DOACs, it is reasonable to omit the morning
DOAC dose before the procedure to reduce bleeding risk.
Descriptions of type of surgery or procedure and bleeding
risk category:
A high-bleed-risk surgery or procedure incl udes major
abdominal surgery (e.g., cancer resection), major thoracic
surgery, major orthopedic surg ery, and any c ardiac,
spinal, or intracranial surgery. Any patient having neur-
axial anesthesia is classied as high-bleed-risk because of
the risk for spinal epidural hematomas which could cause
limb paralysis.
A low-to-moderate-bleed-risk surgery or procedure
includes most surgeries that are <1-h duration and proce-
dures that do not involve neuraxial anesthesia.
A minimal-bleed-risk surgery or procedure includes tooth
extractions, root canal, skin biopsies, cataract surgery, and
selected colonoscopies, for which anticoagulants can be
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
328
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
continued without interruption. Permanent pacemaker and
internal cardiac debrillator implantation, as well as cardiac
catheterization, also can be done without stopping
anticoagulants.
Section 8 Recommendations 2020
i. Patients with atrial brillation or a mechanical heart
valve who are receiving oral anticoagulant therapy and
require a procedure associated with a minimal risk of
bleeding (e.g., tooth extraction, skin biopsy, cataract
removal, cardiac pacemaker) should not have anticoa-
gulation interrupted around the time of the procedure
[Evidence Level B].
ii. For patients with atrial brillation receiving a DOAC
for stroke prevention who require temporary DOAC
interruption for an elective surgery or procedure,
the following approach is recommended [Evidence
Level B]:
a. For a low-to-moderate-bleed-risk surgery or proce-
dure, stop the DOAC the day before the procedure and
the day of the procedure (i.e., skip 2 d total), and
restart the day after the procedure.
b. For a high-bleed-risk surgery or procedure, stop the
DOAC 2 d before the procedure, the day of the
procedure, and one day after the procedure (i.e., skip
4 d total).
Note: An exception involves patients on dabigatran with
impaired renal function (CrCl <50 mL/min) in whom an
additional 12 d of interruption is suggested before
surgery or procedure. Refer to clinical considerations
for additional information.
iii. For pati ents with atrial brillation receiving warfarin
for stroke prevention who require temporary warfarin
interruption for an elective surgery or procedure:
a. For patients at low-to-moderate stroke risk (e.g.,
CHADS2 score 04), warfarin should be stopped
for 5 d pre-procedure, and r esumed within 24 h
post-procedure, without heparin bridging [Evidence
Level A].
b. For patients at high-stroke risk (e.g., CHADS2 score
56 or prior perioperative stroke), heparin bridging is
suggested during warfarin interruption, typically with
twice-daily subcutaneous injections of low-molecu-
lar-weight heparin for 3 d before and 3 d after the
surgery or procedure [Evidence Level B]. If bridging
is used pre-operatively, it is recommended to forego
post-operative bridging in selected patients, especially
those undergoing high-bleed-risk procedures [Evi-
dence Level B].
iv. For patients with a mechanical heart valve who are
receiving warfarin for stroke prevention and require
temporary warfarin interruption for elective surgery or
procedure, stopping warfarin 5 d pre-procedure is
recommended and should be resum ed within 24 h
post-procedure [Evidence Level A].
Heparin bridging is recommended for selected patients
with a mitral valve prosthesis and for high-risk patients
with an aortic valve prosthesis (e.g., with additional
risk factors for stroke) [Evidence Level B].
If bridging is used pre-operatively, it is recommended
to forego post-operative bridging in selected patients,
especially those undergoing high-bleed-risk proce-
dures [Evidence Level B].
v. For patients receiving acetylsalicylic acid for stroke
prevention who require an elective or urgent (within
7 d) carotid endarterectomy (CEA) or coronary artery
bypass surgery, acetylsalicylic acid should be continued
without interruption [Evidence Level B].
vi. For patients who are receiving dual antiplatelet therapy
with acetylsalicylic acid and a P2Y12 inhibitor (e.g.,
clopidogrel, ticagrelor) for secondary stroke prevention
who require urgent CEA (within 7 d), acetylsalicylic acid
and a P2Y12 inhibitor should be continued periopera-
tively [Evidence Level C].
vii. For patients undergoing other types of surgery, continu-
ing acetylsalicylic acid could be considered before a
low/moderate-bleed-risk surgery or procedure. Interrupt-
ing acetylsalicylic acid before a high-bleed-risk surgery
or procedure could be considered for 710 d [Evidence
Level C].
Section 8 Clinical Considerations
Perioperative management of patients undergoing a mini-
mal-bleed-risk procedure
1. For patients undergoing minor procedures that are con-
sidered minimal-bleed-risk (refer to denition above), it
is not routinely necessary to stop anticoagulants. How-
ever, there are some caveats to the management of such
patients:
a. Any of the minimal-bleed-risk procedures could be
considered as having a higher bleed risk warranting
anticoagulant interruption (e.g., tooth extraction in a
patient with poor dentition or cataract surgery with
retrobulbar anesthesia) based on individual patient
circumstances.
b. In patients receiving a DOAC who are undergoing a
minimal bleed-risk procedure, it is prudent to omit the
morning DOAC dose just before the procedure be-
cause the peak anticoagulant effect, occurring 13h
after intake, may coincide with the timing of the
procedure and may increase the risk for bleeding.
c. For pacemaker or ICD implantation, patients can
continue warfarin, but the INR should be <3.0 at the
time of the procedure.
d. For coronary angiography, continuing anticoagulants
if a femoral artery approach is used may not be
advisable as such patients are at increased risk for
developing a hematoma or false aneurysm.
e. For colonoscopy, anticoagulation can be continued in
selected patients in which the likelihood of polypect-
omy or multiple biopsies is low.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 329
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
f. For dental procedures, oral tranexamic acid mouth-
wash can be used before and 23 times daily after the
procedure to reduce bleeding since such oral bleeding,
although not clinically important, may cause distress
to patients.
Perioperative management of patients undergoing a mod-
erate-to-high-risk procedure
1. Patients having a high-bleed-risk surgery or procedure
only need to be off DOACs for 2 d before the procedure,
corresponding to a 6068-h interval between the last
DOAC dose and the time of surgery, which means there
is little to no residual anticoagu lant effect at surgery
given the 1215-h half-life of DOACs.
2. Patients having a low/moderate-bleed-risk surgery or
procedure only need to be off DOACs for 1 d before the
procedure, corresponding to a 3642-h interval between
the last dose and the surgery.
3. For all patients, no DOAC should be taken on the day of
surgery/procedure.
4. The exception to this approach is patients on dabigatran
with impaired renal function (creatinine clearance
<50 mL/minute). Because dabigatran is cleared primarily
by the kidneys, a longer interruption interval is needed
(4 d before a high-bleed-risk surgery: 2 d before a low/
moderate-bleed-risk surgery).
5. Post-operative resumption of DOACs should wait at least
24 h after a low/moderate-bleed-risk surgery or procedure
and 4872 h after a high-bleed-risk surgery or procedure.
6. There are caveats to post-operative DOAC management:
First, the 4872-h resumption interval can be extended if
there is greater than expected post-operative bleeding,
which is important because the full anticoagulant effect
of DOAC is almost immediate after oral intake. Second,
in patients who are unable to take medications by mouth
and who are at high risk for venous thromboembolism,
low-dose low molecular weight heparin (LMWH) can be
given for the initial 13 post-operative days
Section 9: Management of Extracranial Carotid Disease and
Intracranial Atherosclerosis
Carotid endarterectomy (CEA) has been shown to prevent
stroke recurrence in patients who have sustained a minor stroke or
TIA with ipsilateral high-grade carotid stenosis. For those with
50%99% stenosis, the number of persons needed to undergo
surgery to prevent one ipsilateral stroke in ve years was
estimated to be 9 for men versus 36 for women. Women with
symptomatic disease had signicantly higher odds of 30-d mor-
tality following CEA compared with men. (adjusted OR = 1.4,
95% CI 1.021.94).
51
The use of CEA for asymptomatic carotid artery disease is
controversial. One-year results from the recent SPACE-2 trial,
52
indicated there were no signicant differences between groups (CEA
vs. best medical management) in the occurrences of any stroke after
day 30, up to one-year, ipsilateral stroke, disabling stroke, any death,
myocardial infarction, restenosis or TIA. The trial was terminated
early due to low recruitment. In this same trial, there were no
signicant differences in the same outcomes for the comparison of
best medical management versus carotid-artery angioplasty.
Section 9 Recommendations 2020
9.1 Symptomatic Carotid Artery Stenosis
9.1.1 Imaging
i. If revascularization is being considered for carotid steno-
sis based only on carotid ultrasound, then CTA or contrast
enhanced MRA is recommended to conrm the degree of
stenosis and guide surgical decision-making, as well as to
assess fo r tandem disease [Evidence Level C].
a. Conversely, carotid ultrasound may be required after
initial diagnosis of carotid stenosis using CTA or
contrast-enhanced MRA if heavily calcied plaque
or other features make quantication of stenosis less
reliable [Evidence Level C].
9.1.2 Indications for carotid revascularization
i. Patients with a symptomatic event attributed to an ipsi-
lateral 50%99% carotid artery stenosis should be evaluat-
ed without delay for potential carotid revascularizati on by a
health professional with stroke expertise [Evidence Level B].
a. In men with 50%99% and women with 70%99%
symptomatic carotid artery stenosis, CEA is recom-
mended and should be performed as soon as possible
following the qualifying event [Evidence Level A].
b. In women with 50 to 69 percent symptomatic carotid
stenosis, CEA may be considered in those at hig hest
risk of stroke recurrence and upon consideration of
other patient factors [Evidence Level B].
9.1.3 Procedures
i. Caro tid revascularization (CEA or Carotid artery stenting
[CAS]) should be performed by a proceduralist/centre
that routinely audits their performance results, espe-
cially perioperative stroke, and death rates [Evidence
Level B].
a. For CEA, the randomized trials upon which these
recommendations are based (benets accrued for
patients undergoing surgery within 6 months of symp-
toms) involved combined perioperative stroke and
death rates of 6%7% [Evidence Level A].
b. For CAS, the rand omized trial upon which these
recommendations are based involved combined peri-
procedural stroke and death rates of 5% [Evidence
Level B].
ii. CEA is generally more appropriate than CAS for patients
over age 70 years who are otherwise t for surgery as current
evidence indicates stenting carries a higher peri-procedural
risk of stroke and death in older patients [Evidence Level A].
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
330
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
iii. Carotid stenting may be considered for patients who are
not operative candidates for technical, anatomic, or
medical reasons [Evidence Level A].
9.1.4 Timing
i. In clinically stable patients (men and women), CEA
should be performed as early as possible following a
qualifying event [Evidence Level B] and ideally within
14 d [Evide nce Level A].
ii. In men with 50%69% stenosis, the benet of CEA is
greatest when performed within 14 d of the qualifying
event [Evidence Level A] and is attenuated when per-
formed beyond 14 d of the qualifying event (Refer to
Appendix Three below for summary of recurrent stroke
risk at various time points).
Section 9.1 Clinical Considerations
1. Most data regarding optimal timing of carotid revascu-
larization for symptomatic carotid stenosis are derived
from studies of CEA and not CAS. However, it may be
reasonable to consider that similar recommendations
regarding timing also apply to CAS.
2. In exceptional situations, if local system barriers preclude
timely access to CEA while CAS is more rapidly accessi-
ble, this latter revascularization procedure may be consid-
ered in patients otherwise considered eligible for CAS.
However, every effort must be made to enable local
systems of care to ensure timely access to CEA.
3. It may be reasonab le to consider delaying CEA beyond
48 h of the qualifying event as surgery before this time
may be associated with a higher risk of perioperative
complications, particularly when the qualifying event
was a stroke and not a TIA.
4. For patients with moderate or severe stroke due to
symptomatic carotid stenosis, the benet of carotid
revascularization is uncertain and should be considered
on an individual basis, as such patients were excluded
from trials of CEA and CAS.
5. In acute stroke patients with tandem lesions (cervical carotid
stenosis or occlusion and ipsilateral intracranial large vessel
occlusion) who have undergone endovascular thrombectomy
(EVT) but in whom no acute CAS has been performed
during the EVT procedure, subsequent carotid revasculari-
zation by CAS and CEA should be considered if the patient
otherwise remains a candidate for either procedure (as
determined by residual degree of carotid stenosis, stroke
severity, patient recovery, infarct size, reperfusion and bleed-
ing risk, and other factors).
9.2 Asymptomatic and Remotely Symptomatic Carotid
Artery Stenosis
i. Individuals with asymptomatic carotid artery stenosis
should receive aggressive medical management of risk
factors as dened throughout the Secondary Prevention of
Stroke Module (e.g., blood pressure, diabetes, cholesterol,
antiplatelet therapy, smoking cessation, and lifestyle
changes) [Evidence Level B].
ii. CEA may be considered for highly selected patients
with 60%99% carotid stenosis who are asymptomatic
or were remotely symptomatic (i.e., greater than six
months prior to presentation) [Evidence Level A].
a. The benet of CEA for women with 60%99%
asymptomatic carotid artery steno sis is not clear and
should only be considered in highly selected patients
[Evidence Level B] in consultation with a health
professional with stroke expertise.
b. Patients should be evaluated to determine eligibility
for CEA, such as a life expectancy of more than ve
years, and an acceptable risk of surgical complications
[Evidence Level A].
c. In carefully
selected patients, CEA should be performed
by a surgeon who routinely audits their performance
results and demonstrates a less than 3% risk of
peri-operative morbidity and mortality [Evidence
Level B].
d. Important improvements in best medical therapy
(control of blood pressure, lipids, diabetes, and smok-
ing) since the major trials of endarterectomy for
asymptomatic stenosis possibly make their results
less applicable to contemporary management practise
(Evidence Level C).
iii. Carotid stenting may be considered in patients wi th
60%99% asymptomatic carotid stenosis who are not
operative candidates for technical, anatomic, or medical
reasons provided there is a less than 3% risk of peri-
procedural morbidity and mortality [Evidence Level A].
Section 9.2 Clinical Considerations:
1. Although their impact on clinical decision-making re-
garding revascularization of asymptomatic patients is
uncertain, several factors may confer a higher risk of
stroke in patients with asymptomatic stenosis, including:
a. Progression of stenosis over time.
b. Ipsilateral covert brain infarcts on imaging.
c. Ipsilateral intracranial embolizat ion detected on tran-
scranial Doppler.
d. Plaque morphology on non-invasive imaging (e.g.,
volume, echolucency, intraplaque hemorrhage).
9.3 Symptomatic Vertebral Artery Stenosis
i. (New for 2020): For patients with symptomatic vertebral
artery stenosis (extracrani al or intracranial), medical ther-
apy is recommended over stenting for secondary stroke
prevention [Evidence Level B].
9.4 Symptomatic Intracranial Artery Stenosis
i. For patients with a recent ischemic stroke or TIA due to
symptomatic intracranial artery stenosis of 70%99%,
medical therapy is recommended over stenting for sec-
ondary stroke prevention [Evidence Level B].
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 331
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
Note: The SAMMPRIS protocol consist ed of 3 months of
dual antiplatelet therapy with acetylsalicylic acid and
clopidogrel (excluding high-bleeding risk patients), and
is typically followed by antiplatelet monotherapy there-
after, plus intensive lipid-lowering therapy with high-
dose statin, blood pressure treatment, and structured
lifestyle modication addressing smoking cessation, exer-
cise, and diet.
ii. In patients who have been managed with maximal
medical therapy in the presence of intracranial stenosis
and experience a recurrent stroke, there is lack of evi-
dence to guide management decisions; intracranial an-
gioplasty (with or without stenting) may be reasonable in
carefully selected patients [Evidence Level C].
9.5 Cervicocephalic Artery Dissection
i. (New for 2020): For patients with ischemic stroke or TIA
that is preceded by head/neck trauma, cervical spine
mechanical trigger event, or prominent head/neck pain,
a diagnosis of carotid or vertebral artery dissection should
be suspected [Evidence Level C].
ii. For patients with ischemic stroke or TIA in whom a
carotid or vertebral artery dissection is suspected, CTA
or MRA of the head and neck (or catheter angiogram) is
recommended as the diagnostic neurovascular i maging
test rather than ultrasound [Evidence Level C].
Note: CTA or MRA are the preferred non-invasive
diagnostic imaging tests for patients with a suspected
cervicocephalic artery d issection, as neck ultrasound
does not fully visualize the vertebral arteries and can
miss distal carotid artery dissections originating above
the angle of the jaw.
iii. Antithrombotic therapy for stroke prevention is recom-
mended for individuals with a diagnosis of an acute or
recent extracranial carotid or vertebral artery dissection
[Evidence Level B].
a. (New for 2020): There is an uncertainty about the
comparative efcacy of antiplatelet therapy versus
anticoagulation with heparin or warfarin; either treat-
ment is considered reasonable based on current evi-
dence [Evidence Level B]; decisions should be based
on individual risk/benet analysis taking into consid-
eration the imaging features of the dissection (pres-
ence and degree of stenosis, intraluminal thrombus,
vessel occlusion, pseudoaneurysm), brain imaging,
patient characteristics, and estimated bleeding risk
[Evidence Level C].
b. The optimal duration of antithrombotic therapy post-
dissection is uncertain; decisions may be based on
individual clinical factors and imaging appearances
on follow-up vascular imaging [Evidence Level C].
iv. There is a lack of evidence regarding the safety and
efcacy of anticoagulation for intracranial arterial dis-
sections and treatment decisions shou ld be individualized
[Evidence Level C].
Section 9.5 Clinical Considerations
1. There is an insufcient evidence at this time to make a
recommendation regarding the use of DOACs in patients
with arterial dissections [Evidence Level C].
Section 10: Other Cardiac Issues in Individuals with Stroke
Since the last edition, a new rand omized trial
53
and additional
meta-analyses and other reports further support PFO closure for
secondary stroke prevention in selected patients.
5456
Given that
TIA can be difcult to differentiate from mimics and the fact that
only one of the PFO trials enrolled patients with TIA as an index
event, clinicians should be cautious when contemplating PFO
closure for TIA unless there is a high certainty of ischemia;
accordingly, these 2021 recommendations no longer indicate TIA
as an unqualied indication for closure. There is now moderate-
strength evidence that PFO closure may be targe ted to patient
groups with higher risk echocardiographic features.
For patients with heart failure and without atrial brillation,
the COMMANDER-HF trial,
57
which compared rivaroxaban to
standard care, found no signicant difference in the frequency of
the primary outcome (a composite of death from any cause, MI,
or stroke) between groups. The risks of the individual compo-
nents of the primary outcome did not differ between groups with
the exception of the risk of stroke, which was reduced si gni-
cantly with rivaroxaban (1.08 vs. 1.63 events/100-person years;
HR = 0.66, 95% CI 0.470.95). In the Warfarin versus Aspirin in
Reduced Cardiac Ejection Fraction (WARCEF) trial,
58
which
compared the effectiveness of anticoagulation compared with
antiplatelet therapy for stroke prevention in patients with heart
failure in sinus rhythm, warfarin was associated with a signi-
cantly reduced risk of ischemic stroke (HR = 0.52, 95% CI
0.330.82, p = 0.005); however, the risks of major and minor
hemorrhages were signicantly increased.
59
Section 10 Recommendations
10.1 Patent Foramen Ovale (PFO)
i. Patients with a recent ischemic stroke suspected to be
related to a PFO should have an evaluation by healthcare
professionals with stroke and cardiovascular expertise
[Evidence Level C].
ii. For carefully selected patients with a recent ischemic
stroke attributed to a PFO, PFO device closure plus long-
term antiplatelet therapy is recommended over long-term
antithrombotic therapy alone
provided all the following
criteria are met [Evidence Level A]:
a. Age 1860 years.
b. The diagnosis of the index stroke event is conrmed
by imaging as a non-lacunar embolic ischemic stroke.
c. The p atient has been evaluated by a neurologist or
healthcare professional with stroke expertise, and the
PFO is felt to be the most likely cause for the index
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
332
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
stroke event following a thorough etiological evaluation
that has excluded alternate likely etiologies.
iii. (New for 2020): It is reasonable to recommend against
PFO closure for patients who have none of the following
higher-risk anatomical features on echocardiography: (a)
atrial septal aneurysm; (b) large right-to-left shunt (e.g.,
>20 microbubbles); and (c) large diameter PFO (e.g.,
2 mm) [Evidence Level B].
iv. For patients requiring long-term anticoagulation for other
reasons, the benet of PFO closure is uncertain, and
treatment decisions should be based on individual patient
characteristics and risk versus benet prole [Evidence
Level C].
v. For patients with a recent ischemic stroke attributed to a
PFO who do not undergo PFO closure and are aged 60
years or younger, either antiplatelet or anticoagulant
therapy is recommended for secondary stroke prevention,
unless there is a separate evidence-based indication for
chronic anticoagulant therapy [Evidence Level B].
Section 10.1 Clinical Considerations
1. Warfarin can reduce recurrent stroke; however, this
benet may be outweighed by the increased risk of major
hemorrhage.
2. The role of DOACs is unknown in this population.
10.2 Aortic Arch Ather oma:
i. Aortic arch atheroma should be managed according to the
stroke prevention recommendations included in all rele-
vant sections of the Secondary Prevention of Stroke
Module [Evidence Level C].
ii. In the Aortic Arch Related Cerebral Hazard (ARCH) trial,
no signicant difference was found in individuals treated
with dual antiplatelet therapy (acetylsalicylic acid plus
clopidogrel) as compared with warfarin; the effectiveness
of anticoagulant therapy compared with antiplatelet therapy
in this context is uncertain and the choice should be
individualized [Evidence Level B].
10.3 Heart Failure, Decreased Left Ventricular Ejection
Fraction, Cardiac Thrombus
i. For patients with ischemic stroke or TIA who are in sinus
rhythm and have a left atrial or left ventricular thrombus
demonstrated by echocardiography or other imaging
modality, anticoagulant therapy is recommen ded for
greater than 3 months [Evidence Level C].
ii. For patients with ischemic stroke or TIA who are in sinus
rhythm and have severe left ventricular dysfunction
(ejection fraction 35%) without evidence of left atrial
or left ventricular thrombus, the net benet of anticoagu-
lant therapy (with either vitamin K antagonists or
DOACs) compared with antiplatelet therapy is uncertain,
and the choice of management strategies should be
individualized [Evidence Level B].
Section 11: Cancer-Associated Ischemic Stroke
A diagnosis of cancer can increase the risk of stroke in the
months or years following the diagnosis , particularly among
persons with lung cancer or with more advanced cancers.
60,61
Thrombosis is a common comp lication of malignancy and
represents a frequent cause of death in cancer patients with
a history of stroke.
Section 11 Recommendations
11.1 Cancer-Associated Ischemic Stroke
i. Patients with active malignancy who experience an arte-
rial ischemic stroke or TIA should undergo a standard
etiological work-up for their stroke, including vascular
imaging and cardiac rhythm monitoring [Evidence Level
C]. Refer to Section 1 on Stroke Investigations for addi-
tional information.
ii. Stroke mechanisms associated with malignancy may be
considered when determining etiological investigations,
including non-bacterial (marantic) endocarditis, hyperco-
agulability, paradoxical embolism due to venous throm-
bosis, tumor-related vascular compression, and stroke
related to anti-cancer treatments [Evidence Level C].
iii. In patients with active malignancy and arterial ischemic
stroke or TIA in whom a cancer-associated hypercoagu-
lable state may have contributed to the stroke, antic-
oagulation could be considered over antiplatelet therapy
[Evidence Level C].
a. When anticoagulation is used, low-molecular weight
heparin therapy is preferred [Evidence Level C]. The
role of DOACs is unkno wn but under study and may
be reasonable after consideration of patient prefer-
ence.
Section 11 Clinical considerations
1. Management decisions for these patients should be made
in collaboration with a health professional with expertise
in Hematology, Oncology or Thrombosis, and should
take into account the type of underlying cancer, the risk
of bleeding, the extent of neoplastic disease, the patients
overall prognosis and expressed goals of care.
2. In patients with active malignancy and arterial ischemic
stroke or TIA with a concurrent venous thromboembo-
lism (deep vein thrombosis [DVT] or pulmonary
embolism [PE]) in whom the stroke is presumed to be
due to a paradoxical embolus, anticoagulation for sec-
ondary prevention should follow guidelines for the man-
agement of DVT and PE in cancer patients which
includes LMWH and selected DOACs (Refer to
www.thrombosiscanada.ca).
C
HALLENGES AND FUTURE DIRECTIONS
Advances in stroke prevention, driven by high-quality clinical
studies, continue to inform each new edition of these guidelines.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 333
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
However, we are still far from adequately addressing, at a global
level, the 10 modiable risk factors that account for 90% of the
population attributable risk of stroke.
62
The largest impact on
stroke prevention globally will likely be achieved by continued
large-scale efforts to address hypertension, diabetes, diet, exer-
cise, smoking, in addition to atrial brillation at both policy and
individual levels.
A key tenet of secondary stroke prevention remains the
importance of identifying the most likely stroke etiology and
tailoring therapy accordingly. Although the completed ESUS
trials found no overall benet of anticoagulation, further research
aims to identify whether specic subgroups may benet. Dual
pathway inhibition is a promising strateg y.
63
Newer anticoagu-
lants targeting factor XI represent promising future treatments for
stroke prevention. Studies are ongoing (NCT02604667) and
others are needed to better dene when and how occult cancer
should be investigated in cryptogenic stroke patients, and if
found, what antithrombotic regimen best protects these patients
from recurrent arterial strokes.
64
Immediate challenges to optimal secondary stroke prevention
would therefore include the need to develop, grow, and maintain
systems for virtual delivery of care to patients through telemedi-
cine.
65,66
The SARS-CoV2 virus represents a well-documented
challenge to acute stroke care
67
but its impact on the risk of stroke
recurrence, either directly among patients having been infected
with the virus, or on other patients who have suffered collateral
damage from diminished access to stroke care, will be important
to now study.
A challenge that concerns research in all elds of medicine
including stroke
68
is the need to ensure adequate sex and gender
representation in therapeutic trials to ensure generalizab ility of
results to both men and women. This edition is the rst of our
guidelines to start incorporating a sex and gender descriptive
analysis into the literature review for eac h recommendation, and
future editions will strive to include gender and sex-based
recommendations where appropriate.
S
UMMARY
The 2020 update of the Canadian Stroke Best Practice
Secondary Prevention of Stroke Recommendations provide a
common set of guiding principles for important aspects of
secondary stroke prevention, emphasizing that individuals who
have experienced a stroke or TIA require access expert preven-
tion care in a timely way. In Canada, coordinated systems have
evolved over time, growing the number of stroke prevention
services and protocols to increase access in many under-serviced
areas. In the age of Covid-19, there are new opportunities to
provide preventi on interventions remotely to narrow the inequi-
ties in access to care.
A
CKNOWLEDGEMENTS
Heart &Stroke gratefully acknowledges the Secondary Pre-
vention of Stroke writing group leaders and members all of whom
have volunteered their time and expertise to the update of these
recommendations. Members of the Canadian Stroke Consortium
were involved in all aspects of the development of these recom-
mendations. These recommendations (in whole or specic parts)
underwent external review by: Jason Andrade, Rohit Bhatia,
Margie Burns, Elena Adela Cora, Roxanne Cournoyer, Laurent
Derex, Paul Dorian, Charles Duffy, Eric Ehrensperger, Yuriy
Flomin, Kirsten George-Phillips, Sarah Grant, Milan Gupta,
Rahul Jain, Shirin Jalani, Glen Jickling, Hooman Kamel, Hong
Kao, Puneet Kapur, Lisa Keon, Lisa Korec, Catherine Legault,
Gerald MacDonald, GB John Mancini, Michael MacDonald,
Kaylee Murphy, Kelvin Kuan Huei Ng, Darlene Peacock, Andre
Roussin, Joanna D Schaafsma, Peter Senior, Aleksander Tkach,
Sean Virani, Elissa Weinberg, Heather Williams, and Janice
Williams. We thank the Canadian Stroke Best Practices and
Quality Advisory Committee members, including Eric Smith
(Co-Chair), Anita Mountain (Co-Chair), Leanne Casaubon, Gord
Gubitz, Dar Dowlatshahi, Dylan Blacquiere, Louise Clement,
Thalia Field, Farrell Leibovitch, Christine Papoushek, Jeffrey
Habert, Barbara Campbell, Joyce Fung, Michael Hill, Tim
Hillier, Thomas Jeerakath il, Eddy Lang, Pascale Lavoie, Beth
Linkewich, Colleen OConnell, Melanie Penn, Jai Shankar,
Debbie Timpson, Theodore Wein, and Katie White. We acknowl-
edge and thank Norine Foley and the evidence analysis team at
workHORSE; Laurie Charest of Heart & Stroke for her coordi-
nation of the Canadian Stroke Best Practice Recommendations
(CSBPR) teams and processes; Andrea deJong, Francine Forget
Marin, and the Heart & Stroke internal teams who contributed to
the development of these recommendations and publication:
Communications, Translation, Knowledge Translation, Engage-
ment, Health Policy, and Digital Solutions. Heart & Stroke is
especially grateful to the members of the Community Consulta-
tion and Review Panel who reviewed all sections of this module,
shared their personal experiences and insights on what did or
would have made their journey optimal. The members of the
Secondary Prevention of Stroke Community Consultation and
Review Panel (CCRP) included: Cheryl Beattie, Jennifer Bogart,
Dan Dobbin, Glen Hilton, Judy Hilton, Allan Morrison, and
additional volunteers who reviewed all sections and provided
input and developed patient infographic resources.
F
UNDING
The development of the CSBPR is funde d in its entirety by
Heart & Stroke. No funds for the development of these guidelines
come from commercial interests, including pharmaceutical and
device companies. All members of the recommendation writing
groups, and external reviewers are volunteers and do not receive
any remuneration for participation in guideline development,
updates, and reviews. All participants complete a conict of
interest declaration prior to participation.
C
ONFLICTS OF INTEREST
The following authors have identied actual or potential
conicts of interest which have been mitigated through the design
of a multidisciplinary writing group model and additional mea-
sures by the advisory committee as required. David J. Gladstone
received a Mid-Career Investigator Award from the Heart and
Stroke Foundation, a peer-reviewed provincial operating grant
from Ontario Genomics; all funds paid to his institution to
support the project (no personal fees); Independent Medical
Safety Monitor for the NINDS-sponsored ARCADIA trial
(uncompensated), and local site PI for the NASPAF-ICH and
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
334
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
ENRICH-AF trials (with all site fees paid to his institution; no
personal fees); served as PI of the SCREEN-AF trial (uncom-
pensated); operating grant from the Canadian Stroke Prevention
Intervention Network (C-SPIN), a peer-reviewed Canadian Insti-
tutes of Health Research [CIHR]) national network grant; Adju-
dication Committee SAFE-HD trial (uncompensated); and is the
Site Investigator for NAVIGATE ESUS trial and NASPAF-ICH
trial (all site fees paid to my institution); co-leader of NAVI-
GATE ESUS atrial myopathy/atrial brillation working group
(uncompensated). Alexandre Y. Poppe is Site PI and Site
co-investigator for ESCAPE-NA1 (NoNo), NAVIGATE-ESUS
(Bayer), RESPECT-ESUS (Boehringer-Ingelheim), POINT
(NIH); DSMB for FLOW; Canadian Stroke Trials for Optimized
Results (CaSTOR) networking grant; Chair, Canadian Stroke
Consortium National Stroke Fellowship Program; receives sup-
port for fellowship program from Servier; and research grant
support from Stryker. Jafna Cox is a Medical Consultant
(received payment) for Bayer, HLS Therapeutics, Novartis;
Lecture Series (received payment) with Bayer; holds an Investi-
gator-initiated grant from Bayer; and participating in a Phase II
study of a Factor XI inhibitor, funded by Bayer. James Douketis
is an Advisory Board Consultant for BMS Pzer, Servier,
Leo Pharma, Sano, Bayer; holds a grant/Honorarium from
Thrombosis Canada (non-prot); participation in PAUSE trial.
Monies received as personal fees from Janssen, Pzer, Bayer,
Bristol Myers Squibb, Sano, Servier Canada, Portola are de-
posited in hospital-based (St. Josephs Healthcare Hamilton) and
university-based (McMaster University) research accounts and/or
charitable foundations. Brett R. Graham holds a Canadian Stroke
Consortium Catalytic Research Capacity Generation Grant; is site
PI University of British Columbia, University of Calgary -
SECRET and TOPSECRET; site sub-I for TEMPO-2 an
ESCAPE NA1. Honorarium received from Servier Canada (to
give a talk on anticoagulation in ab to family physicians.
Marilyn Labrie is an advisory board member for Teva Canada
Fremanezumab 20200823. Jennifer Mandzia is an advisory
board with Bayer; and is clinical trial Site PI for several studies.
Daniel Ngui is a member of the Advisory Board for Amgen,
Astra Zeneca, BMS, BI, Lilly, Novonordisk; moderating and
speaking engagements for Amgen, Astra Zeneca, BMS, BI, Lilly,
Novonordisk; EMR grants and audits for Amgen, Astra Zeneca,
BI, Novartis; holds research grants from Simple Trial, Amgen
20170191 trial, IHE eCare CV Risk, CHRC EMR Registry
Trials: AF OAC, Advantage CV, and Advantage OP-Phase 4
and EMR audits; Health Choices First Video Education shares;
Investments in communications companies including CHRC,
CCRN, MD Briefcase, Medplan, Liv Agency, Four Health; board
membership on CCS Lipid guideline Panel, CCS A. b guideline
second panel, SPH Hospital CME Committee, BC Guidelines,
UBC CPD CME This changed my practice, Alliance for Best
Practices in Health Education. William Semchuk is an Advisory
Board Member for BMS Pzer; Speaker Honorarium from BMS,
Pzer, Astra Zeneca, Sano, Servier, Bayer, BI. Jacob A Udell is
an advisory board member for Boehringer Ingelheim, Novartis,
Sano; Secondary analysis of banked biospecimens from a
completed RCT for Janssen; Consultant on clinical research
development, (no involvement in marketing) with Boehringer
Ingelheim, Janssen, Sano, Amgen, Merck, Novartis; received
grant to University Health Network for clinical trial from Astra-
Zeneca; grant to University Health Network for clinical trial and
honorarium for leadership of a multicenter RCT from Boehringer
Ingelheim; grant to Womens College Hospi tal for clinical
research study from Janssen; grant to Womens College Hospital
to be a site in a multicenter RCT and honorarium for steering
committee membership in cohort study by Novartis; grant to
Womens College Hospital for site participation in a multicenter
RCT and honorarium for national co-PI role in multicenter RCT
for Sano; grants to his institutions for clinical trial participation
from Boehringer Ingelheim, Novartis, Sano; grant from Bayer.
Stephen van Gaal is a site investigator who enrolls patients for
Portola, Bayer; advisory board member for Servier (edoxaban);
support for conference attendance from Bayer (rivaroxaban);
Canadian Stroke Cons ortium committee member. Karina Villa-
luna is a Clinical Research Coordinator participating in research
for NoNO Inc, Portola and BMS. Eric E. Smith participates in
consulting for clinical trials in cerebral amyloid angiopathy,
vascular cognitive impairment, and preventing atrial brilla-
tion-related stroke with Bayer, Biogen, Javelin; Royalties from
UpToDate for chapter on diagnosing vascular dementia; and is
Study site for Biogen study on adacanumab for Alzheimers
disease. Dar Dowlatshahi holds a Heart & Stroke Foundation of
Canada Research Grant and Salary Award; a Patent for CARL fo r
detection of contrast extravasation; and is involved in several
funded clinical trials; member, Canadian Stroke Consortium
board of directors. Theodore Wein is a consultant for Servier,
Allergan Inc, Ipsen Inc; a speaker for Servier; receives research
funding from Allergan and Servier; and is PI on a Servier funded
study. Shelagh Coutts holds a current CIHR grant. Gord Gubitz is
an Advisory Board member for Bayer, BI, Pzer; Member,
DSMB CATIS-ICAD trial; Member, Steering Committee. HSF
Canada Stroke Best Practices; Co-Chair, World Stroke Organiza-
tion Education Committee; Atlantic Canada Together Enhancing
Acute Stroke Treatment (ACTEAST): Improving Access and
Efciency of Treatment. Co-Investigator. Canadian Institutes of
Health Research (CIHR) Project Grant; Optimization and Valida-
tion of a Novel Emergency Department Point-of-Care MRI. Nova
Scotia Health Research Foundation. Research Nova Scotia Trust,
Industry collaborator; the effects of prism adaptation training on
visual attention and functional activities in stroke patients with
neglect. Nova Scotia Health Research Foundation Establishment
Grant. Paul Pageau is a past board member, Canadian Association
of Emergency Physicians. Pascale Lavoie holds investments in
Johnson and Johnson/United health group. The following authors
have no conicts of interest to declare: M. Patrice Lindsay, Anita
Mountain, Aline Bourgoin, John B. Falconer, Norine Foley,
Manraj K.S. Heran, Lena McDonald, Rebecca McGuff, Amanda
Rodgerson, Tammy Tebbutt, and Carmen Tuchak.
S
TATEMENT OF AUTHORSHIP
David J Gladstone (First author) and Alexandre Y. Poppe
(Senior Author) are co-chairs of the Secondary Prevention of
Stroke expert writing group and lead authors contributing to all
aspects of the development, evidence and data analysis, writing,
editing, and nal approval of this manuscript; M. Patrice Lindsay is
corresponding author, senior editor of the Canadian Stroke Best
Practice Recommendations (CSBPR) and of this manuscript,
involved in all aspects of scientic literature review, writing group
deliberations, external review process, manuscript preparation, and
a writer of supplementary documentation. Aline Bourgoin, Jafna
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 335
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
Cox, James Douketis, John B. Falconer, Brett R. Graham, Marilyn
Labrie, Lena McDonald, Jennifer Mandzia, Daniel Ngui, Paul
Pageau, Amanda Rodgerson, William Semchuk, Tammy Tebbutt,
Carmen Tuchak, Jacob Udell, Stephen van Gaal, Karina Villaluna,
Manraj K.S. Heran, and Pascale Lavoie are all members of
Secondary Prevention of Stroke expert writing group and contrib-
uted by reviewing, analyzing and discussing the evidence and
collectively nalizing the wording of all included recommenda-
tions. Norine Foley conducted the evidence searches and complet-
ed the evidence tables and evidence summaries supporting this
guideline update and contributed to writing of this manuscript. Dar
Dowlatshahi, Theodore Wein, Eric E. Smith, Anita Mountain, and
Gord Gubitz are senior leaders of the stroke best practices advisory
committee and provided inputs throughout development of the
recommendations, and participated in development, preparation,
and editing of this manuscript. Shelagh Coutts led a subgroup
focused on updates to section one recommendations for triage.
Rebecca McGuff provided inputs to this manuscript and is respon-
sible for development of knowledge translation resources.
S
UPPLEMENTARY MATERIAL
To view supplementary material for this article, please visit
https://doi.org/10.1017/cjn.2021.127.
R
EFERENCES
1. Graham ID, Harrison MB, Brouwers M, Davies BL, Dunn S.
Facilitating the use of evidence in practice: evaluating and
adapting clinical practice guidelines for local use by health care
organizations. JOGNN. 2002;31:599611.
2. Vernooij RW, Alonso-Coello P, Brouwers M, Martinez Garcia L.
Reporting items for updated clinical guidelines: checklist for the
reporting of updated guidelines (CheckUp). PLoS Med. 2017;14:
e1002207. DOI: 10.1371/journal.pmed.1002207.
3. Brouwers MC, Kho ME, Browman GP, et al. AGREE II: advancing
guideline development, reporting and evaluation in health care.
CMAJ. 2010;182:E839842. DOI: 10.1503/cmaj.090449.
4. Coutts SB, Wein TH, Lindsay MP, et al. Canadian stroke best
practice recommendations: secondary prevention of stroke guide-
lines, update 2014. Int J Stroke. 2015;10:28291. DOI: 10.1111/
ijs.12439.
5. Wein T, Lindsay MP, Cote R, et al. Canadian stroke best practice
recommendations: secondary prevention of stroke, sixth edition
practice guidelines, update 2017. Int J Stroke. 2018;13:42043.
DOI: 10.1177/1747493017743062.
6. Lindsay MP, Gierman N, Harris JE, et al. People with lived
experience at the centre of Canadian stroke best practice recom-
mendations: a model for guideline developers. J Patient Exp.
2020;7:95156. DOI: 10.1177/2374373520956538.
7. Rothwell PM, Giles MF, Chandratheva A, et al. Effect of urgent
treatment of transient ischaemic attack and minor stroke on early
recurrent stroke (EXPRESS study): a prospective population-
based sequential comparison. Lancet. 2007;370:143242. DOI:
10.1016/s0140-6736(07)61448-2.
8. Lioutas VA, Ivan CS, Himali JJ, et al. Incidence of transient
ischemic attack and association with long-term risk of stroke.
Jama. 2021;325:37381. DOI: 10.1001/jama.2020.25071.
9. Kamal N, Hill MD, Blacquiere DP, et al. Rapid assessment and
treatment of transient ischemic attacks and minor stroke in
Canadian emergency departments: time for a paradigm shift.
Stroke. 2015;46:298790. DOI: 10.1161/strokeaha.115.010454.
10. Schnabel RB, Haeusler KG, Healey JS, et al. Searching for atrial
brillation poststroke: a white paper of the AF-SCREEN Inter-
national Collaboration. Circulation. 2019;140:183450. DOI: 10.
1161/circulationaha.119.040267.
11. Rabi DM, McBrien KA, Sapir-Pichhadze R, et al. Hypertension
Canadas 2020 comprehensive guidelines for the prevention,
diagnosis, risk assessment, and treatment of hypertension in
adults and children. Can J Cardiol. 2020;36:596624. DOI: 10.
1016/j.cjca.2020.02.086.
12. Salisbury C, O Cathain A, Thomas C, et al. Telehealth for patients at
high risk of cardiovascular disease: pragmatic randomised con-
trolled trial. BMJ. 2016;353:i2647. DOI: 10.1136/bmj.i2647.
13. Liu S, Feng W, Chhatbar PY, Liu Y, Ji X, Ovbiagele B. Mobile
health as a viable strategy to enhance stroke risk factor control: a
systematic review and meta-analysis. J Neurol Sci.
2017;378:1405. DOI: 10.1016/j.jns.2017.04.050.
14. Widmer RJ, Collins NM, Collins CS, West CP, Lerman LO, Lerman
A. Digital health interventions for the prevention of cardiovascu-
lar disease: a systematic review and meta-analysis. Mayo Clin
Proc. 2015;90:46980. DOI: 10.1016/j.mayocp.2014.12.026.
15. Kapoor A, Lindsay MP, Yu AYX, et al. Call 911: lower ambulance
utilization among young adults, especially women, with stroke.
Can J Neurol Sci. 2020;47:7649. DOI: 10.1017/cjn.2020.119.
16. Hajek P, Phillips-Waller A, Przulj D, et al. A randomized trial of e-
cigarettes versus nicotine-replacement therapy. N Engl J Med.
2019;380:62937. DOI: 10.1056/NEJMoa1808779.
17. McClester M, Mounsey A, Mackler L. Clinical inquiry: do oral
contraceptives carry a signicant risk of stroke for women with
migraines? J Fam Pract. 2013;62:6623.
18. Feigin VL, Roth GA, Naghavi M, et al. Global burden of stroke and
risk factors in 188 countries, during 19902013: a systematic
analysis for the Global Burden of Disease Study 2013. Lancet
Neurol. 2016;15:91324. DOI: 10.1016/s1474-4422(16)30073-4.
19. Law MR, Morris JK, Wald NJ. Use of blood pressure lowering
drugs in the prevention of cardiovascular disease: meta-analysis
of 147 randomised trials in the context of expectations from
prospective epidemiological studies. BMJ. 2009;338:b1665.
DOI: 10.1136/bmj.b1665.
20. Kitagawa K, Yamamoto Y, Arima H, et al. Effect of standard vs
intensive blood pressure control on the risk of recurrent stroke: a
randomized clinical trial and meta-analysis. JAMA Neurol.
2019;76:130918. DOI: 10.1001/jamaneurol.2019.2167.
21. Amarenco P, Kim JS, Labreuche J, et al. A Comparison of two LDL
cholesterol targets after ischemic stroke. N Engl J Med.
2020;382:919. DOI: 10.1056/NEJMoa1910355.
22. Shoamanesh A, Patrice Lindsay M, Castellucci LA, et al. Canadian
stroke best practice recommendations: management of spontaneous
intracerebral hemorrhage, 7th edition update 2020. Int J Stroke.
2020:1747493020968424. DOI: 10.1177/1747493020968424.
23. Global Burden of Disease Results Tool. Available at: http://
ghdx.healthdata.org/gbd-results-tool; accessed November 15,
2020.
24. Sarwar N, Gao P, Seshasai SR, et al. Diabetes mellitus, fasting blood
glucose concentration, and risk of vascular disease: a collabora-
tive meta-analysis of 102 prospective studies. Lancet.
2010;375:221522. DOI: 10.1016/s0140-6736(10)60484-9.
25. Group AtCCRiDS. Effects of intensive glucose lowering in type 2
diabetes. N Engl J Med. 2008;2008:254559.
26. Duckworth W, Abraira C, Moritz T, et al. Glucose control and
vascular complications in veterans with type 2 diabetes. N Engl J
Med. 2009;360:12939.
27. Perkovic V, Jardine MJ, Neal B, et al. Canagliozin and renal
outcomes in type 2 diabetes and nephropathy. N Engl J Med.
2019;380:2295306. DOI: 10.1056/NEJMoa1811744.
28. Cannon CP, Pratley R, Dagogo-Jack S, et al. Cardiovascular out-
comes with ertugliozin in type 2 diabetes. N Engl J Med.
2020;383:14251435. DOI: 10.1056/NEJMoa2004967.
29. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and
cardiovascular outcomes in type 2 diabetes (REWIND): a double-
blind, randomised placebo-controlled trial. Lancet.
2019;394:12130. DOI: 10.1016/s0140-6736(19)31149-3.
30. Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and
cardiovascular outcomes in patients with type 2 diabetes. N Engl J
Med. 2019;381:841
51. DOI: 10.1056/NEJMoa1901118.
31. Rosenstock J, Perkovic V, Johansen OE, et al. Effect of linagliptin
vs placebo on major cardiovascular events in adults with type 2
diabetes and high cardiovascular and renal risk: The CARME-
LINA Randomized Clinical Trial. Jama. 2019;321:6979. DOI:
10.1001/jama.2018.18269.
THE CANADIAN JOURNAL OF NEUROLOGICAL SCIENCES
336
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press
32. Kernan WN, Viscoli CM, Furie KL, et al. Pioglitazone after
ischemic stroke or transient ischemic attack. N Engl J Med.
2016;374:132131. DOI: 10.1056/NEJMoa1506930.
33. Johnston SC, Easton JD, Farrant M, et al. Clopidogrel and aspirin in
acute ischemic stroke and high-risk TIA. N Engl J Med.
2018;379:21525. DOI: 10.1056/NEJMoa1800410.
34. Wong KS, Wang Y, Leng X, et al. Early dual versus mono
antiplatelet therapy for acute non-cardioembolic ischemic stroke
or transient ischemic attack: an updated systematic review and
meta-analysis. Circulation. 2013;128:165666. DOI: 10.1161/
circulationaha.113.003187.
35. Kennedy J, Hill MD, Ryckborst KJ, Eliasziw M, Demchuk AM,
Buchan AM. Fast assessment of stroke and transient ischaemic
attack to prevent early recurrence (FASTER): a randomised
controlled pilot trial. Lancet Neurol. 2007;6:9619. DOI: 10.
1016/s1474-4422(07)70250-8.
36. Pan Y, Elm JJ, Li H, et al. Outcomes associated with clopidogrel-
aspirin use in minor stroke or transient ischemic attack: A pooled
analysis of clopidogrel in high-risk patients with acute non-
disabling cerebrovascular events (CHANCE) and platelet-orient-
ed inhibition in new TIA and minor ischemic stroke (POINT)
trials. JAMA Neurol. 2019;76:146673. DOI: 10.1001/
jamaneurol.2019.2531.
37. Hao Q, Tampi M, ODonnell M, Foroutan F, Siemieniuk RA,
Guyatt G. Clopidogrel plus aspirin versus aspirin alone for acute
minor ischaemic stroke or high risk transient ischaemic attack:
systematic review and meta-analysis. BMJ. 2018;363:k5108.
DOI: 10.1136/bmj.k5108.
38. Pereira NL, Farkouh ME, So D, et al. Effect of genotype-guided oral
P2Y12 inhibitor selection vs conventional clopidogrel therapy on
ischemic outcomes after percutaneous coronary intervention: the
TAILOR-PCI randomized clinical trial. Jama. 2020;324:76171.
DOI: 10.1001/jama.2020.12443.
39. Wang Y, Zhao X, Lin J, et al. Association Between CYP2C19 Loss-of-
Function Allele Status and Efcacy of Clopidogrel for Risk Reduc-
tion Among Patients With Minor Stroke or Transient Ischemic
Attack. Jama. 2016;316:708. DOI: 10.1001/jama.2016.8662.
40. Meschia JF, Walton RL, Farrugia LP, et al. Efcacy of clopidogrel for
prevention of stroke based on CYP2C19 allele status in the POINT
trial. Stroke. 2020;51:205865. DOI: 10.1161/strokeaha.119.028713.
41. Johnston SC, Amarenco P, Denison H, et al. Ticagrelor and aspirin
or aspirin alone in acute ischemic stroke or TIA. N Engl J Med.
2020;383:20717. DOI: 10.1056/NEJMoa1916870.
42. Amarenco P, Denison H, Evans SR, et al. Ticagrelor added to
aspirin in acute nonsevere ischemic stroke or transient ischemic
attack of atherosclerotic origin. Stroke. 2020;51:350413. DOI:
10.1161/strokeaha.120.032239.
43. Hart RG, Diener HC, Coutts SB, et al. Embolic strokes of undeter-
mined source: the case for a new clinical construct. Lancet
Neurol. 2014;13:429 38. DOI: 10.1016/s1474-4422(13)70310-7.
44. Diener HC, Sacco RL, Easton JD, et al. Dabigatran for prevention of
stroke after embolic stroke of undetermined source. N Engl J
Med. 2019;380:190617. DOI: 10.1056/NEJMoa1813959.
45. Secondary prevention in non-rheumatic atrial
brillation after tran-
sient ischaemic attack or minor stroke. EAFT (European Atrial
Fibrillation Trial) study group. Lancet. 1993;342:125562.
46. Andrade JG, Aguilar M, Atzema C, et al. The 2020 Canadian
cardiovascular society/Canadian heart rhythm society compre-
hensive guidelines for the management of atrial brillation. Can J
Cardiol. 2020;36:18471948. DOI: 10.1016/j.cjca.2020.09.001.
47. Guimarães HP, Lopes RD, de Barros ESPGM, et al. Rivaroxaban in
patients with atrial brillation and a bioprosthetic mitral valve. N
Engl J Med. 2020;383:211726. DOI: 10.1056/NEJMo
a2029603.
48. Yasuda S, Kaikita K, Akao M, et al. Antithrombotic therapy for
atrial brillation with stable coronary disease. N Engl J Med.
2019;381:110313. DOI: 10.1056/NEJMoa1904143.
49. Thrombosis Canada. NOACs DOACs: Perioperative Management
(May 2020). Available at: https://thrombosiscanada.ca/wp-
uploads/uploads/2021/01/22.-NOACs-DOACs-Perioperative-Ma
nagement-17May2020.pdf; accessed December 15, 2020.
50. Douketis JD, Spyropoulos AC, Duncan J, et al. Perioperative
management of patients with atrial brillation receiving a direct
oral anticoagulant. JAMA Intern Med. 2019;179:146978. DOI:
10.1001/jamainternmed.2019.2431.
51. Dansey KD, Pothof AB, Zettervall SL, et al. Clinical impact of sex
on carotid revascularization. J Vasc Surg. 2020;71:1587
94.e1582. DOI: 10.1016/j.jvs.2019.07.088.
52. Reiff T, Eckstein HH, Mansmann U, et al. Angioplasty in asymp-
tomatic carotid artery stenosis vs. endarterectomy compared to
best medical treatment: one-year interim results of SPACE-2. Int J
Stroke. 2019;15:1747493019833017. DOI: 10.1177/17474930
19833017.
53. Lee PH, Song JK, Kim JS, et al. Cryptogenic stroke and high-risk
patent foramen ovale: the DEFENSE-PFO trial. J Am Coll
Cardiol. 2018;71:233542. DOI: 10.1016/j.jacc.2018.02.046.
54. Saver JL, Mattle HP, Thaler D. Patent foramen ovale closure
versus medical therapy for cryptogenic ischemic stroke: a topi-
cal review. Stroke. 2018;49:15418. DOI: 10.1161/strokeaha.
117.01815 3.
55. Mas JL, Derex L, Guérin P, et al. Transcatheter closure of patent
foramen ovale to prevent stroke recurrence in patients with
otherwise unexplained ischaemic stroke: expert consensus of the
French Neurovascular Society and the French Society of Cardi-
ology. Arch Cardiovasc Dis. 2019;112:53242. DOI: 10.1016/j.
acvd.2019.06.002.
56. Turc G, Calvet D, Guerin P, Sroussi M, Chatellier G, Mas JL.
Closure, anticoagulation, or antiplatelet therapy for cryptogenic
stroke with patent foramen ovale: systematic review of random-
ized trials, sequential meta-analysis, and new insights from the
CLOSE study. J Am Heart Assoc. 2018;7:e008356. DOI: 10.
1161/jaha.117.008356.
57. Zannad F, Anker SD, Byra WM, et al. Rivaroxaban in patients with
heart failure, sinus rhythm, and coronary disease. N Engl J Med.
2018;379:133242. DOI: 10.1056/NEJMoa1808848.
58. Homma S, Thompson JL, Pullicino PM, et al. Warfarin and aspirin
in patients with heart failure and sinus rhythm. N Engl J Med.
2012;366:1859
69. DOI: 10.1056/NEJMoa1202299.
59. Homma S, Thompson JL, Sanford AR, et al. Benet of warfarin
compared with aspirin in patients with heart failure in sinus
rhythm: a subgroup analysis of WARCEF, a randomized con-
trolled trial. Circ Heart Fail. 2013;6:98897. DOI: 10.1161/
circheartfailure.113.000372.
60. Navi BB, Reiner AS, Kamel H, et al. Risk of arterial thromboem-
bolism in patients with cancer. J Am Coll Cardiol. 2017;70:926
38. DOI: 10.1016/j.jacc.2017.06.047.
61. Jang HS, Choi J, Shin J, et al. The long-term effect of cancer on
incident stroke: a nationwide population-based cohort study in
Korea. Front Neurol. 2019;10:52. DOI: 10.3389/fneur.2019.
00052.
62. ODonnell MJ, Chin SL, Rangarajan S, et al. Global and regional
effects of potentially modiable risk factors associated with acute
stroke in 32 countries (INTERSTROKE): a case-control study.
Lancet. 2016;388:76175. DOI: 10.1016/s0140-6736(16)30506-2.
63. Sharma M, Hart RG, Connolly SJ, et al. Stroke outcomes in the
cardiovascular outcomes for people using anticoagulation strate-
gies (COMPASS) trial. Circulation. 2019;139:113445. DOI: 10.
1161/circulationaha.118.035864.
64. Rioux B, Touma L, Nehme A, Gore G, Keezer MR, Gioia LC.
Frequency and predictors of occult cancer in ischemic stroke: a
systematic review and meta-analysis. Int J Stroke. 2021;16:129.
DOI: 10.1177/1747493020971104.
65. Smith EE, Mountain A, Hill MD, et al. Canadian stroke best practice
guidance during the COVID-19 pandemic. Can J Neurol Sci.
2020;47:4748. DOI: 10.1017/cjn.2020.74.
66. Iodice F, Romoli M, Giometto B, et al. Stroke and digital technolo-
gy: a wake-up call from COVID-19 pandemic. Neurol Sci.
2021;42:8059. DOI: 10.1007/s10072-020-04993-3.
67. Nogueira R, Abdalkader M, Qureshi MM, et al. EXPRESS: global
impact of the COVID-19 pandemic on stroke hospitalizations
and mechanical thrombectomy volumes. Int J Stroke. 2021:
1747493021991652. DOI: 10.1177/1747493021991652.
68. ONeill ZR, Deptuck HM, Quong L, et al. Who says no to
participating in stroke clinical trials and why: an observational
study from the Vancouver stroke program. Trials. 2019;20:313.
DOI: 10.1186/s13063-019-3434-0.
LE JOURNAL CANADIEN DES SCIENCES NEUROLOGIQUES
Volume 49, No. 3 May 2022 337
https://doi.org/10.1017/cjn.2021.127 Published online by Cambridge University Press