Download Prevention of Stroke - Canadian Stroke Best Practice

Document related concepts

Seven Countries Study wikipedia , lookup

Epidemiology of metabolic syndrome wikipedia , lookup

Transcript
CANADIAN BEST PRACTICE
RECOMMENDATIONS FOR
STROKE CARE
Fourth Edition
Lindsay MP, Gubitz G, Bayley M, Phillips S (Editors),
on Behalf of the Canadian Stroke Best Practices and Standards Working Group
CHAPTER 2
Stroke Prevention
(UPDATE September 2012)
Coutts S, Kelloway L (Co-Chairs)
on Behalf of the Prevention of Stroke
Best Practices Writing Group 2012
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Section 2: Stroke Prevention
Table of Contents
Canadian Best Practice Recommendations for Stroke Care
Prevention of Stroke Chapter ~ Fourth Edition
(Updated September 2012)
Table of Contents
Topic
Page
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE - INTRODUCTION
2
PREVENTION OF STROKE OVERVIEW
2
Highlights of Prevention of Stroke Update 2012
3
Stroke Prevention Definitions
4
Development of the CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
5
STROKE PREVENTION BEST PRACTICES TASK GROUP 2012
6
STROKE PREVENTION EXTERNAL REVIEWERS 2012
7
CANADIAN STROKE BEST PRACTICES AND STANDARDS WORKING GROUP
7
PREVENTION OF STROKE BEST PRACTICE RECOMMENDATIONS
8
2.1
Lifestyle And Risk Factor Management
8
2.1.1
Healthy Balanced Diet
8
2.1.2
Sodium
8
2.1.3
Exercise
8
2.1.4
Weight
8
2.1.5
Alcohol Consumption
8
2.1.6
Oral Contraceptives and Hormone Replacement Therapy
8
2.2
Blood Pressure Management
13
Figure 2.2: CHEP Expedited Assessment and Diagnosis of Patients with Hypertension
18
Table 2.2: CHEP Hypertension Pharmacomanagement Table (2012)
19
2.3
Lipid Management
20
2.4
Diabetes Management
23
2.5
Antiplatelet Therapy
26
2.6
Antithrombotic Therapy for Atrial Fibrillation
31
Figure 2.6: CCS Algorithm for Stroke Risk and Decisions in Patients with Atrial Fibrillation
37
Table 2.6: Oral Anticoagulants for the Prevention of Stroke in Atrial Fibrillation Patients
38
2.7 Management of Extracranial Carotid Disease and Intracranial Atherosclerosis
42
2.8
Sleep Apnea Screening and Management
47
2.9
Smoking Cessation Management
52
Table 2.9: Pharmacotherapy for Smoking Cessation in Patients with Stroke and TIA
56
Reference List
Fourth Edition FINAL
59
Update: September 2012
Page 1 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
The Canadian Best Practice Recommendations for Stroke Care are intended to provide up-to-date evidence-based
guidelines for the prevention and management of stroke. The goal of disseminating and implementing these
recommendations is to reduce practice variations in the care of stroke patients across Canada, and to reduce the gap
between knowledge and practice. Recommendations are updated every two years to ensure they continue to reflect
contemporary stroke research evidence and leading expert opinion. Each update involves critical review of the current
medical literature, which informs decisions regarding modification of the recommendations and the performance measures
used to assess their impact. Every attempt is made to coordinate with other Canadian groups who are developing guidelines
that relate to stroke, such as hypertension, atrial fibrillation and diabetes.
This is the fourth edition of the Canadian Best Practice Recommendations for Stroke Care, which was first released in 2006.
The theme of the 2012 – 2013 update is TAKING ACTION, and stresses the critical role and responsibility of healthcare
providers at every stage of the continuum of care to ensure that best practice recommendations are implemented and
adhered to. TAKING ACTION will lead to optimal outcomes for each stroke patient by providing the best care within the most
appropriate setting. This includes rapid and efficient access to diagnostic services, stroke expertise and medical and surgical
interventions, rehabilitation and support for ongoing recovery and community reintegration.
TAKING ACTION requires a committed team approach and strong coordination of care across regions and networks, with
pre-hospital, acute care, rehabilitation and community-based healthcare providers working together to ensure optimal
outcomes for patients and their families, regardless of geographic location.
TAKING ACTION also applies to patients who have experienced a stroke, their families and informal caregivers. Stroke
patients and their families need to actively participate in their recovery and openly communicate with their healthcare team.
Patients and families must participate in setting the goals they want to achieve during recovery from a stroke, and share
concerns, as well as physical, mood and cognitive issues with their team, which will lead to the care required for optimal
recovery in all aspects of health.
SECTION 2.0
PREVENTION OF STROKE OVERVIEW
TAKING ACTION FOR THE PREVENTION OF STROKE
TAKING ACTION is an imperative within primary and secondary stroke prevention and applies to systems of care, healthcare
providers, patients and the broader community. The primary underpinnings of ‘prevention’ require TAKING ACTION to prevent
first stroke or transient ischemic attack or recurrence of a cerebrovascular event. The actions required to prevent first and
recurrent stroke include rapid access to specialized stroke prevention services; promotion of healthy lifestyles to minimize
vascular risk; aggressive risk factor management, especially even the slightest elevation in blood pressure; appropriate
prescription of medications for prevention; patient compliance with medication regimes and lifestyle changes such as diet
and smoking cessation; timely access to interventions such as carotid endarterectomy; and, screening of appropriate
patients for smoking status, mood, cognition and sleep difficulties.
TAKING ACTION in stroke prevention involves healthcare providers, policy makers, patients and the public. A critical
component of stroke prevention is access to specialized stroke prevention services, ideally provided by dedicated stroke
prevention clinics. Stroke prevention clinics (or similar vascular prevention clinics) provide a comprehensive interdisciplinary
approach to prevention of first or recurrent stroke, conduct detailed assessments by a range of healthcare disciplines,
facilitate timely access to appropriate diagnostics and interventions, and provide education to patients and families. They
also promote continuity of care between acute care facilities, the patient and their primary care providers.
Recent reports on the quality of stroke services across Canada and within specific provinces have shown that there is an
insufficient number of stroke prevention clinics or similar services, even in urban areas where large volumes of stroke
patients reside, and even fewer in rural settings. Establishing stroke prevention clinics and services within all regions of care
is an imperative in TAKING ACTION for stroke prevention.
Fourth Edition FINAL
Update: September 2012
Page 2 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
HIGHLIGHTS OF THE PREVENTION OF STROKE 2012 UPDATE
The 2012 update of the Stroke Prevention Chapter of the Canadian Best Practice Recommendations for Stroke Care
reinforces the growing and changing body of research evidence available to guide stroke prevention services. Aggressive
risk factor management is emphasized throughout this chapter.
Highlights of the moderate and significant updates to stroke prevention recommendations for 2012 include:
 the potential stroke risk of oral contraceptives and hormone replacement therapy, especially in patients who also
smoke;
 continued emphasis on the important role of blood pressure for stroke, and diligent monitoring and treatment to
keep blood pressure levels well below 140 mm Hg systolic and 90 mm Hg diastolic;
 alignment of diabetes and stroke recommendations with updated guidelines by the Canadian Diabetes
Association; revisions to the lipid management section to reflect ongoing analysis and interpretation of the
SPARCL trial;
 the release of the findings from the ASA versus ASA + clopidogrel arm of the SPS3 study that reinforced
recommendations advising against the use of dual-antiplatelet therapy;
 anticoagulant therapy recommendations for patients with atrial fibrillation and stroke have been significantly
revised to reflect the release of the new classes of anticoagulants, based on the RE-LY (dabigatran), ROCKET
(rivaroxaban) and ARISTOTLE (apixaban) trials;
 carotid interventions for asymptomatic patients incorporate the 10 year follow-up findings of the ACST trial;
 new recommendations on screening, identification and management of patients with obstructive sleep apnea,
diagnosed both pre and post stroke;
 expansion of recommendations on smoking cessation assessment and management, including pharmacotherapy,
in collaboration with the CAN-ADAPTE and C-CHANGE guideline groups; and,
 development of a TAKING ACTION FOR STROKE PREVENTION quick response guide and pocket card.
PREVENTION OF STROKE 2012 UPDATE RESOURCE PACKAGE INCLUDES:
 Prevention of Stroke Best Practice Recommendations
 Prevention of Stroke Quick Reference Guide
 Prevention of Stroke Patient Order Set
 Prevention of Stroke Educational Slide Decks
1. Assessment of a Patient with TIA or Non-Disabling Stroke
2. Lifestyle and Risk Factor Management To Reduce Stroke (Non-pharmacological)
3. Pharmacotherapy for the Prevention of Stroke
 Reference Materials from:
o Canadian Hypertension Education Program (Diagnostic Algorithm, Pharmacotherapy Table)
o Canadian Cardiovascular Society (Atrial Fibrillation Guidelines Pocket Reference Booklet)
Fourth Edition FINAL
Update: September 2012
Page 3 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
STROKE PREVENTION DEFINITIONS
Primary prevention is an individually based clinical approach to disease prevention, directed toward preventing the initial
occurrence of a disorder in otherwise healthy individuals.1,2 Primary prevention is usually implemented in the primary care
setting, and the physician, advanced practice nurse, pharmacist or patient may initiate a discussion of stroke risk reduction.
Primary prevention and health promotion recommendations related to stroke (lifestyle and risk factor management,
hypertension screening, dyslipidemia screening, diabetes management, management of atrial fibrillation, and asymptomatic
carotid stenosis) emphasize the importance of screening and monitoring those patients at high risk of a first stroke event.
Primary prevention strategies are also promoted through health-oriented organizations and agencies such as the Heart and
Stroke Foundation, Canadian Cardiovascular Society, Hypertension Canada, and Health Canada.
Primary prevention and the reduction of risk factor prevalence in the general population are the not main focus of the
Canadian Best Practice Recommendations for Stroke Care; therefore, only selected recommendations related to primary
prevention are included. A comprehensive set of recommendations in this area is being developed for inclusion in future
updates. A list of existing high quality stroke prevention guidelines, including primary prevention, are provided in the
reference section at the end of this chapter for further guidance (Hyperlink).
Secondary prevention is an individually based clinical approach aimed at reducing the risk of a recurrent vascular events
in individuals who have already experienced a stroke or transient ischemic attack and in those who have one or more of the
medical conditions or risk factors that place them at high risk of stroke.1,2 Secondary prevention recommendations in this
document are directed to those risk factors most relevant to stroke, including lifestyle (diet, sodium intake, exercise, weight,
smoking, and alcohol intake), hypertension, dyslipidemia, previous stroke or transient ischemic attack, atrial fibrillation and
stroke, and carotid stenosis. Secondary prevention recommendations can be addressed in a variety of settings—acute care,
stroke prevention clinics, and community-based care settings. They pertain to patients initially seen in primary care, those
who are treated in an emergency department and then released and those who are hospitalized because of stroke or
transient ischemic attack.
Recommendations for secondary prevention of stroke should be implemented throughout the recovery phase, including
during inpatient and outpatient rehabilitation, reintegration into the community and ongoing follow-up by primary care
practitioners. Secondary prevention should be addressed at all appropriate healthcare encounters on an ongoing basis
following a stroke or transient ischemic attack.
Fourth Edition FINAL
Update: September 2012
Page 4 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
DEVELOPMENT OF THE CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
For detailed methodology on the development and dissemination of the Canadian Best Practice Recommendations for
Stroke Care please refer to the stroke best practices website at http://www.strokebestpractices.ca/index.php/methods/.
Acknowledgements
The Canadian Stroke Network gratefully acknowledges the task group leaders and members, the external reviewers, all of
who volunteered their time and expertise to this project. We thank the Canadian Stroke Information and Evaluation Working
Group for its work in updating and confirming the performance measures that accompany each recommendation. We
acknowledge Corrine Davies-Schinkel for work on the systematic reviews of the literature and meeting coordination; and, we
thank Marie-France Saint-Cyr and Jan Carbon for their work on the French translations.
Funding
The development of these Canadian stroke care guidelines is funded in its entirety by the Canadian Stroke Network, which is
in turn funded by the Networks of Centres of Excellence program. No funds for the development of these guidelines come
from commercial interests, including pharmaceutical 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.
Citing the Prevention of Stroke Update 2012
Coutts S, Kelloway L, on behalf of the Prevention of Stroke Writing Group. Chapter 2: Prevention of Stroke.
In Lindsay MP, Gubitz G, Bayley M, and Phillips S (Editors) on behalf of the Canadian Stroke Best Practices and Standards
Working Group. Canadian Best Practice Recommendations for Stroke Care: 2012; Ottawa, Ontario Canada: Canadian
Stroke Network.
Comments
We invite comments, suggestions, and inquiries on the development and application of the Canadian Best Practice
Recommendations for Stroke Care and ongoing updates.
Please forward comments to the Canadian Stroke Network’s Performance and Standards office at
[email protected]
Fourth Edition FINAL
Update: September 2012
Page 5 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
Stroke Prevention Working Group 2011-2012:
NAME
PROFESSIONAL ROLE
Coutts, Shelagh
Co-Chair, Stroke Neurologist, Calgary Stroke Program,
Hotchkiss Brain Institute, University of Calgary
Alberta
Kelloway, Linda
Co-Chair, Best Practices Leader, Ontario Stroke Network
Ontario
Boulos, Mark
Neurology Fellow, Sunnybrook Health Sciences Centre
Ontario
Professor of Medicine and Director of the Division of Respirology and of the Centre for
Sleep Medicine and Circadian Biology of the University of Toronto
and of the Centre for Sleep Health and Research, University Health Network
Emergency Physician, University Health Network; Lecturer, Faculty of Medicine,
University of Toronto
Ontario
Hudon, Mark
Assistant Professor of Radiology and Clinical Neurosciences, University of Calgary
Diagnostic and Interventional Neuroradiology
Foothills Medical Centre
Alberta
Kwiatkowski, Brenda
Nurse, Clinical Coordinator, Stroke Prevention Clinic
Loewen, Sherry
Nurse, Case Manager, Stroke Prevention Clinic
McMurtry, Michael Sean
Assistant Professor, University of Alberta; Canadian Cardiovascular Society Liaison
Bradley, Douglas
Ellis, Paul
LOCATION
Ontario
Saskatchewan
Manitoba
Alberta
Mysak, Tania
Neurologist, Director of Sunnybrook Health Sciences Centre Sleep Laboratory, University
of Toronto
Pharmacist, Clinical Practice Manager, Pharmacy Services
Nearing, Shannon
Nurse Practitioner, Acute Stroke Program, QEII Health Sciences Centre
Pettersen , Jaqueline
Cognitive/Behavioural Stroke Neurologist; Assistant Professor, University of British
Columbia
Pipe, Andrew
Chief, Division of Prevention and Rehabilitation,
University of Ottawa Heart Institute
Ontario
Reid, Robert
Associate Director, Minto Prevention and Rehabilitation Centre
University of Ottawa Heart Institute
Ontario
Rioux, Annie
Nurse Practitioner, Stroke Prevention Clinic
Ontario
Ryan, Clodagh
Respirologist, Assistant Director of the Centre for Sleep Health and Research, University
Health Network; Assistant Professor, Faculty of Medicine, University of Toronto
Ontario
Murray, Brian
Ontario
Alberta
Nova Scotia
British Columbia
Selby, Peter
Chief, Addictions Division, Ambulatory Care and Structured Treatment Program
Head, Nicotine Dependence Clinic, Centre for Addiction and Mental Health
Associate Professor, Departments of Family and Community Medicine, Psychiatry and
Dalla Lana School of Public Health Sciences, University of Toronto
Ontario
Silver, Karen
Family Physician, Department of Family Medicine, Dalhousie University, and Memorial
University
Program Director, Department of Neurology, Infant Jesus Hospital, Quebec City,
University of Laval
New Brunswick
Verreault, Steve
Fourth Edition FINAL
Update: September 2012
Quebec
Page 6 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Prevention of Stroke
Overview
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
Stroke Prevention External Reviewers 2012:
EXTERNAL REVIEWER
PROFESSIONAL ROLE
Briggs, Helen
Drug Utilization Pharmacist, Lakeridge Health
Ontario
Butcher, Ken
Associate Professor in the Department of Medicine, Division of Adult Neurology at
the University of Alberta
Alberta
Cote, Robert
Professor, Department of Neurology and Neurosurgery, McGill University
Quebec
Dean, Naeem
Dowlatshahi, Dariush
Hanley, Patrick
Hart, Robert
Johnson-Clatworthy, Linda
McNeill, Jeanne
Travers, Andrew
Clinical Professor, Faculty of Medicine, University of Alberta
Director, Stroke Program, Royal Alexandra Hospital
Stroke Neurologist; Associate Scientist, Neurosciences, OHRI
Assistant Professor of Medicine, University of Ottawa
Medical Director, Sleep Centre, Foothills Medical Centre
Professor, Department of Medicine, University of Calgary
Vascular Neurologist, Division of Neurology, Hamilton Health Sciences; Professor,
Faculty of Medicine, McMaster University
Stroke Nurse Practitioner, Markham-Stouffville Hospital
Medical Director Family Practice/Geriatrics/Palliative Care,
Horizon Health Network
Provincial Medical Director, Emergency Medical Services
LOCATION
Alberta
Ontario
Alberta
Ontario
Ontario
New Brunswick
Nova Scotia
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
Best Practices and Standards Working Group
MEMBER
Phillips, Stephan
Co-Chair
Bayley, Mark
Co-Chair
Gubitz, Gord
Lindsay, Patrice
Smith, Eric
Harris, Devin
Graham, Ian
Joiner, Ian
LeBrun, Louise- Helene
Lafferty, Katie
Millbank, Robin
Fourth Edition FINAL
PROFESSIONAL ROLE
Stroke Neurologist, Queen Elizabeth II Health Sciences Centre; Professor,
Division of Neurology, Faculty of Medicine, Dalhousie University
Physiatrist; Associate Professor, University of Toronto
Acquired Brain Injury, Physical Medicine & Rehabilitation
Toronto Rehabilitation Institute
Assistant Professor, Division of Neurology
Faculty of Medicine, Dalhousie University
Director of Performance and Standards, Canadian Stroke Network
Staff Lead, Canadian Best Practice Recommendations for Stroke Care
Stroke Neurologist & Researcher
Hotchkiss Brain Institute
Clinical Associate Professor, UBC
Staff, Department of Emergency Medicine, St. Paul's Hospital
Senior Scientist, Centre for Practice-Changing Research, The Ottawa Hospital
Research Institute; Associate Professor, School of Nursing, University of Ottawa
Director of Stroke, Heart and Stroke Foundation of Canada
Stroke Neurologist, CHUM
Executive Director, Canadian Stroke Network
Manager, Professional Development and Training, Canadian Stroke Network
Update: September 2012
LOCATION
Nova Scotia
Ontario
Nova Scotia
Canada
Alberta
British Columbia
Ontario
Canada
Quebec
Canada
Canada
Page 7 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Section 2: Prevention
2.1 Lifestyle and Risk Factor Management
Prevention of Stroke Best Practice Recommendations
Best Practice Recommendation 2.1
Lifestyle and Risk Factor Management
* Minor revisions for 2012
Persons at risk of stroke and patients who have had a stroke should be assessed for vascular disease risk factors and lifestyle
management issues (diet, sodium intake, exercise, weight, alcohol intake, and use of oral contraceptives and hormone
replacement therapy) [Evidence Level B].
i. They should receive information and counseling about possible strategies to modify their lifestyle and risk factors
[Evidence Level B].
ii. Referrals to appropriate specialists should be made where required to provide more comprehensive assessments and
structured programs to manage risk factors [Evidence Level B].
Lifestyle and risk factor information and counseling should be provided and include:
2.1.1
Healthy balanced diet: Eating a diet high in fresh fruits, vegetables, low-fat dairy products, dietary and soluble fibre,
whole grains and protein from plant sources and low in saturated fat, cholesterol (< 200 mg daily for patients at
increased vascular risk) and sodium, in accordance with Canada's Food Guide to Healthy Eating [Evidence Level B].
2.1.2
Sodium: Following the recommended daily sodium intake from all sources, known as the Adequate Intake. For
persons 9 to 50 years, the Adequate Intake is 1500 mg. Adequate Intake decreases to 1300 mg for persons 51 to 70
years and to 1200 mg for persons over 70 years. A daily upper consumption limit of 2300 mg should not be exceeded
by any age group [Evidence Level B].3
2.1.3
Exercise: Participating in moderate exercise such as walking (ideally brisk walking), jogging, cycling, swimming or
other dynamic exercise four to seven days each week in addition to routine activities of daily living [Evidence Level A].
i. Patients should be counseled to achieve an accumulation of at least 150 minutes of moderate to vigorous
activity per week, in episodes of 10 minutes or more (Refer to the CSEP Canadian Physical Activity
Guidelines 2011 for additional information) [Evidence Level B].4
ii. Most stroke patients should be encouraged to start a regular exercise program.
a. Supervision by a healthcare professional (physical therapist or cardiac rehab) at exercise initiation
should be considered in stroke patients at risk of falls or injury, or in patients with other comorbid
disease (such as cardiac disease), which may place them at higher risk of medical complications
[Evidence Level C].
2.1.4
Weight: Maintaining a body mass index (BMI) of 18.5 to 24.9 kg/m2 or a waist circumference of <80 centimetres for
women and <94 centimetres for men* [Evidence Level B]. (Note: these numbers are reflective of current research
based mostly on Caucasian patients. Refer to References 24 to 31 for waist circumference values for other ethnic
groups)
2.1.5
Alcohol consumption: Limiting consumption to two or fewer standard drinks per day for men and one drink per day
for women who are not pregnant [Evidence Level B].
2.1.6
Birth Control and Hormone Replacement Therapy: Patients who are taking estrogen-containing oral contraceptives
or hormone replacement therapy in the presence of stroke should have the risks and benefits of these treatments
discussed with them. Management alternatives should be considered in these patients [Evidence Level B].
Rationale
A healthy lifestyle reduces the risk of an initial stroke and the risk of a subsequent stroke for patients with a prior stroke.
Hypertension is the single most important modifiable risk factor for stroke. Current research reports estimate that reducing
sodium in foods would abolish high blood pressure for almost one in three Canadians. Furthermore, this evidence suggests that
lowering sodium consumption to adequate intake levels could reduce the incidence of stroke and heart disease by as much as
30 percent, and has a significant impact on lowering blood pressure.18 There is a growing concern for obesity in the Canadian
population, especially in younger adults and this must be addressed with all patients with stroke or at risk. Regular exercise also
Fourth Edition FINAL
Update: September 2012
Page 8 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Section 2: Prevention
2.1 Lifestyle and Risk Factor Management
reduces the risk of stroke and other vascular diseases.5 Research has demonstrated an increased risk of thrombosis with
estrogen-based hormone therapy (both oral contraceptives and hormone-replacement therapy).
Although causes of stroke are generally different for children, lifestyle management issues as described above are equally as
important for the paediatric population, particularly as the long-term risk of recurrence for children is much higher.
System Implications
•
Health promotion efforts that contribute to the prevention of stroke in all communities (integrated with existing chronic
disease prevention initiatives) must be established.
•
Coordinated and comprehensive stroke prevention should be offered by primary care providers, and a mechanism in
place to ensure that stroke risk is addressed during encounters with healthcare professionals throughout the continuum
of care.
•
A public focus on arterial health for paediatric cases–such as diet, exercise, non-smoking, avoidance of drugs that
increase stroke risk.
•
National and international efforts to reduce sodium intake and increase public knowledge about the risks of sodium,
directly targeting the food industry.
•
Access to risk factor management programs (such as hypertension and smoking cessation programs) in all communities,
primary healthcare settings and workplaces.
•
Government actions to restrict smoking in public areas and discourage smoking through legislation and taxation
initiatives.
•
Coordinated efforts among stakeholders such as Heart and Stroke Foundations (national and provincial), the Canadian
Stroke Network, public health agencies, ministries of health and care providers across the continuum to produce patient,
family and caregiver education materials with consistent information and messages on risk factor management.
•
Coordinated process for ensuring access to and awareness of educational materials, programs, activities and other
media related to risk factor management by healthcare professionals, patients and caregivers, including advertising the
availability of educational material, effective dissemination mechanisms and follow-up.
•
Educational resources, that are culturally and ethnically appropriate, are available in multiple languages and that address
the needs of patients with aphasia.
•
Access to healthy living programs, educational materials and healthcare professionals for persons living in rural and
remote locations.
Performance Measures
1. Proportion of the population with major risk factors for stroke, including hypertension, obesity, history of
smoking, low physical activity, hyperlipidemia, diabetes, and atrial fibrillation (core).
2. Annual occurrence rates for stroke in each province and territory by stroke type (core).
3. Stroke mortality rates across provinces and territories, including in-hospital or 30-day rate and one-year rate
(core).
4. Percentage of the population who can identify the major risk factors for stroke, including hypertension, sodium intake,
diet, weight, exercise, smoking and alcohol intake.
5. Percentage of people who are aware of the healthy targets for each stroke risk factor.
6. The annual readmission rate for a recurrent stroke event in patients with previous stroke or transient ischemic attack.
Measurement notes
•
For performance measures 1, 2 and 3: self-reported data can be extracted from provincial and national health surveys.
•
Performance measures 4 and 5: administrative data are available at the local, provincial and national levels.
•
Mortality rates should be risk adjusted for age, sex, stroke severity and comorbidities.
Implementation Resources and Knowledge Transfer Tools
o Canadian Stroke Network Stroke Prevention Teaching Slide Decks: Assessment, Lifestyle Management, Medical
Management www.strokebestpractices.ca
Fourth Edition FINAL
Update: September 2012
Page 9 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
o
o
o
o
o
o
o
Section 2: Prevention
2.1 Lifestyle and Risk Factor Management
Canada’s Food Guide to Healthy Eating- http://www.hc-sc.gc.ca/fn-an/food-guide-aliment/index-eng.php
Instructions for proper waist circumference
http://www.heartandstroke.on.ca/site/c.pvI3IeNWJwE/b.4018281/k.8698/Healthy_Waists.htm
PHAC Eat Well and Be Active Education Toolkit: http://www.hc.sc.gc.ca/fn-an/food-guide-ailment/edu-comm/indexeng.php
Sodium 101: www.sodium101.ca
Sodium Daily Intake Tables3
Canadian Diabetes Association - http://www.diabetes.ca/
Mediterranean diet http://www.mayoclinic.com/health/mediterranean-diet/CL00011
Summary of the Evidence
LINK to Evidence Tables for Lifestyle and Risk Factor Management
Diet
Gillman and associates reported that, based on data collected as part of the Framingham Study, age-adjusted risk for stroke
decreased as consumption of fruits and vegetables increased such that relative risk (RR) = 0.78 for each increase of three
servings per day.6 This effect was independent of BMI, smoking, glucose intolerance, physical activity, blood pressure, serum
cholesterol and intake of energy, ethanol and fat. A meta-analysis of fruit and vegetable consumption and stroke, which included
eight studies and 257,551 individuals over a 13-year follow- up period, showed that consumption of five or more servings of fruits
and vegetables per day is associated with a lower risk of stroke.7 Compared with individuals who had fewer than three servings
of fruit and vegetables per day, the pooled relative risk of stroke was 0.89 (95% confidence interval [CI] 0.83–0.97) for those with
three to five servings per day, and 0.74 (95% CI 0.69–0.79) for those with more than five servings per day.7
Several studies have been published in the past four years comparing different diet types in relation to stroke. Liu (2011), found
that a dietary pattern characterized by high intakes of rice and vegetables and moderate intakes in animal foods was related to
the lowest prevalence of stroke compare to a dietary pattern characterized by high intakes of refined cereal products, potatoes,
and salted vegetables [OR = 1.96 (95% CI = 1.48–2.60); P<0.0001].8 Adjustment for conventional cardiovascular risk factors did
not appreciably change the association [multivariate adjusted OR = 1.59 (95% CI = 1.16–2.17); P = 0.004]. Similarly, Mahe and
colleagues (2010) found that, compared to controls, ischemic stroke patients under the age of 65 had higher saturated fatty acid
scores and lower monounsaturated, omega-6, omega-3 fatty acid scores, lower fruits and vegetables, and a lower overall dietary
score.9 Agnoli and colleagues (2-11) compared four diet regimes and their impact on stroke.10 These included adherence to the
Healthy Eating Index 2005 (HEI-2005), Dietary Approaches to Stop Hypertension (DASH), Greek Mediterranean Index, or the
Italian Mediterranean Index. This study found that all 4 diets had an inverse relationship to stroke occurrence, and that the Italian
Mediterranean Index showed the best overall results [HR = 0.37 (95% CI = 0.19–0.70) and was the only one of the four diets to
show an association with hemorrhagic stroke as well [HR = 0.51(95%CI = 0.22–1.20); P = 0.07)].10
Analyses of data from the Nurses’ Health Study, the Health Professionals Follow-up Study and the Women’s Health Study
supported the association between consumption of fruit and vegetables and reduction of stroke risk in men and women.11,12 In
an analysis of combined data from the Nurses’ Health Study and the Health Professionals Follow-up Study, Joshipura and
associates found that an increase of 1 serving per day of fruits or vegetables was associated with a reduction of risk of six
percent and that cruciferous vegetables, leafy green vegetables and citrus fruit (including juice) contributed most to this effect.11
Liu and colleagues reported a significant inverse relationship between consumption of fruits and vegetables and risk for
cardiovascular disease including stroke.12 When individuals consuming the most fruits and vegetables were compared with those
consuming the least, a relative risk reduction of 0.68 was demonstrated in favour of those with higher consumption levels.12
Sodium
It is well documented that a chronically high dietary sodium intake is associated with elevated blood pressure.13-23 A high intake
of sodium also has direct negative effects independent of blood pressure, such as fibrosis of the heart, kidneys and arteries,
including cerebral arteries.17,20,22,23 The Institute of Medicine of the National Academies established in 2004 a daily Adequate
Intake for sodium of 1500 mg and a daily Tolerable Upper Intake Level of 2300 mg for healthy adults.3 Canadian and American
governments have adopted these values for setting public health policy. The Canadian Heart Health Survey found that the
average Canadian consumes about 3500 mg of sodium a day, and this high sodium intake is estimated to be responsible for a
additional 1 million cases of hypertension.14,15
The literature relating to sodium consumption and health published since the release of the dietary reference intakes in 2004 is
Fourth Edition FINAL
Update: September 2012
Page 10 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Section 2: Prevention
2.1 Lifestyle and Risk Factor Management
currently undergoing scientific review. The reviews will be released later this year, but it is not anticipated that these will result in
a revision of the current sodium intake guidelines.
Weight
Observational studies have examined the relationship between body mass index (BMI) and stroke risk. Saito (2011) compared
high BMIs of 27.0 to 29.9 kg/m2 and BMI of >= 30.0 kg/m2 to a ‘healthy’ BMI between 23.0 and 24.9 kg/m2.24 They reported
hazards ratios for increased stroke risk as 1.09 and 1.25 for men, and 1.29 and 2.16 for women. In addition, in women a weight
increase of greater than 10% over the previous five years was also associated with increased stroke risk. Bazzano (2010)
reported similar findings in a study of Chinese men and women, where the hazard ratios for increased stroke risk were 1.43 for
persons considered overweight (BMI 25.0 to 29.9 kg/m2) and 1.72 for those who were obese with a BMI of 30 kg/m2 or greater. 25
Yatsuya (2010) found similar results (men – HR of 1.81; women – HR of 1.65), and further reported that when the analysis
adjusted for systolic blood pressure, much of the BMI risk affect was attenuated.26
Some researchers have suggested that waist circumference is a preferred measurement of obesity than BMI. 27-29 Dalton et al
(2003) compared BMI, waist circumference (WC) and waist to hip ratio (WHR) for cardiovascular disease risk.30 Overall WHR
showed the strongest correlations in unadjusted results, and these differences diminished when the results were adjusted for
age. Women showed relationships between elevated blood pressure and both WHR and BMI. Jannsen (2004) calculated
several regression models to examine the predictability of BMI and WC for hypertension, dyslipidemia and metabolic syndromes.
31 They reported that waist circumference was a better overall predictor of obesity-related CVD risk. Yau (2011) measured waist
circumference in a case-controlled observational study of independent risk factors for stroke, and reported an odds ration of 4.0
for persons with increased waist to hip ratios. 32 Zhu et al (2004) identified formulas for calculating cardiovascular risk using a
combination of BMI and WC.33 They determined that in while males a formula of 0.68 x BMI + 0.32 x WC was most predictive;
yet in females the WC alone was a strong predictor of cardiovascular risk. Clark and colleagues (2012) caution that the current
parameters for waist circumference may not be applicable to African-Americans and that research should be conducted to
establish appropriate measurement standards for this population.34
Physical activity
Physical activity is an important modifiable lifestyle factor that can influence both the primary and secondary prevention of stroke.
Reimers and colleagues (2009) published a meta-analysis that showed physical activity reduced the risk of all stroke types
(RR=0.32) for men and women combined.35 The results were derived from 33 prospective cohort studies and 10 case-control
studies that addressed the potential effect of physical activity on stroke.
Lee and collaborators published a meta-analysis of 23 studies published between 1983 and 2002 examining the association
between physical activity and stroke incidence or mortality.36 Eighteen cohort studies and 5 case–control studies were included
for analysis. When both types of study were examined together, highly active individuals were reported as having a 27 percent
lower risk of stroke than individuals who were designated as “low active.” Individuals who were designated as moderately active
also had a significantly reduced risk of stroke when compared with low active individuals (RR = 0.80, p < 0.001). The benefits of
high and moderate levels of activity were reported for both ischemic and hemorrhagic strokes. The meta-analysis showed
increasing benefit with increasing activity, a dose–response relationship was also established. However, as Lee and
collaborators pointed out, given the range of definitions of “level of physical activity” in the studies included for assessment, their
analysis suffered from the lack of a single, cohesive definition of what constitutes low, moderate and high levels of activity.36 The
question of what type or quantity of activity is required to reach a moderate level and so to benefit from a 20 percent reduction in
the risk of stroke is one that needs to be investigated by means of a randomized controlled trial.
Patient adherence is important for physical activity to be effective. Jurkiewicz and colleagues (2011) found that patients that
attended a rehabilitation center regularly had higher adherence to an exercise program compared to participants that had
graduated and were required to do solely home-based exercise.37 Common factors preventing exercise that were reported by
patients included: lack of motivation, musculoskeletal issues, and fatigue.
Alcohol
A meta-analysis of 35 observational studies examining the effects of alcohol consumption on stroke risk revealed a significant (p
= 0.004) J-shaped relationship between the amounts of alcohol consumed per day and the risk of ischemic stroke.38 In that
Fourth Edition FINAL
Update: September 2012
Page 11 of 69
Canadian Best Practice Recommendations for Stroke Care
Update 2012 - 2013
Section 2: Prevention
2.1 Lifestyle and Risk Factor Management
analysis, individuals who consumed 1 to 2 drinks per day had the least risk for ischemic stroke (RR = 0.72), while those having
more than 5 drinks per day had the most risk (RR = 1.69) when compared with a group of abstainers.39 The analysis also
confirmed that alcohol consumption has a linear, dose-dependent effect on risk of hemorrhagic stroke. Heavy drinking (more
than 5 drinks per day) was associated with a relative risk of hemorrhagic stroke of 2.18. Irregular and binge drinking (more than 5
drinks at one sitting) have also been associated with an increase in risk for hemorrhagic stroke.39
Data from the Copenhagen City Heart Study were used to examine whether the type of alcohol consumed was related to the
apparent decreased risk of ischemic stroke with moderate alcohol consumption.40 The overall beneficial effect of moderate
alcohol consumption was confirmed; however, the benefit was seen mostly among those individuals who consumed wine. Wine
drinking on a daily, weekly or monthly basis was associated with reduced risk of ischemic stroke (RR = 0.68, 0.66 and 0.88,
respectively, after adjustments for age, sex, smoking, BMI, physical activity, systolic blood pressure, cholesterol,
antihypertensive treatment, triglycerides, education, and diabetes). No similar effect was demonstrated among drinkers of beer
or spirits. Both Kiechl and associates and Sacco reported the greatest risk reduction (RR = 0.41 and 0.40, respectively) among
wine drinkers; however, this was not significantly lower than among drinkers of beer, liquor or a combination of types of
alcohol.41,42
Oral Contraceptives, Hormone Replacement Therapy and Stroke risk in Women
Women taking oral contraceptive or hormone replacement therapy may be at an increased risk of stroke.43-49 Bath and Gray
(2005) conducted a meta-analysis to assess the association between hormone replacement therapy and subsequent stroke.46
They identified 28 trials that included almost 40,000 patients. Their analysis found that hormone replacement therapy was
associated with significant increases in total stroke (OR =1.29, 95% CI 1.13 to 1.47), non-fatal stroke (OR=1.23, 1.06 to 1.44),
stroke leading to death or disability (OR=1.56, 1.11 to 2.20), ischaemic stroke (OR=1.29, 1.06 to 1.56), and a trend to more fatal
stroke (OR=1.28, 0.87 to 1.88). They also found that hormone replacement therapy was not associated with haemorrhagic stroke
(OR=1.07, 0.65 to 1.75) or transient ischaemic attack (OR=1.02, 0.78 to 1.34). Similarly, Renoux and colleagues (2010) found
that, compared to non-users, women using both low and high dose oral hormone replacement therapy had a higher rate of stroke
(Rate Ration= 1.28, 1.15-1.42); however, the use of low-dose transdermal hormone replacement therapy did not increase the
risk of stroke (Rate Ratio=0.95, 0.75-1.20).
Cole and colleagues (2007) identified women using transdermal contraceptive therapy and norgestimate-containing oral
contraceptives.48 There was an increase in the rate of venous thromboembolism among transdermal contraceptive system users
compared with norgestimate-containing oral contraceptives users (incidence rate ratio 2.2, 95% confidence interval [CI] 1.3-3.8).
Stroke rate differences could not be calculated. In a large cohort study of 49, 259 Swedish women aged 30-49, found that the
risk of hemorrhagic stroke was not statistically significantly raised in women who started using oral contraceptive over the age of
30 (Hazard ratio=2.3, 0.8-6.8).49
Fourth Edition FINAL
Update: September 2012
Page 12 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
Best Practice Recommendation 2.2 Blood Pressure Management
* Minor revisions for 2012
Hypertension is the single most important modifiable risk factor for stroke. Blood pressure should be monitored and managed in
all persons at risk for stroke [Evidence Level A].
2.2.1
Blood pressure assessment
All persons at risk of stroke should have their blood pressure measured routinely, ideally at each healthcare encounter, but no
less than once annually [Evidence Level C].
i.
Proper standardized techniques should be followed for initial and subsequent blood pressure measurement including
office, home, and community testing [Evidence Level B] as outlined by the Canadian Hypertension Education
Program.50
ii.
Patients found to have elevated blood pressure (systolic greater than 130 mmHg and/or diastolic greater than 85
mmHg) should undergo thorough assessment for the diagnosis of hypertension [Evidence Level C].
a. A specific follow-up visit should be scheduled and completed for the assessment and diagnosis of hypertension
following an initial elevated blood pressure measurement [Evidence Level C].
b. The specific visit for assessment of hypertension should include three measurements and be conducted in
accordance with the current guidelines of the Canadian Hypertension Education Program [Evidence Level
C].50 See Figure 2.2.1
iii.
Patients with refractory hypertension should have comprehensive investigations for secondary causes of
hypertension [Evidence Level B].
iv.
Patients with hypertension or at risk for hypertension should receive aggressive risk factor modification counseling
and interventions [Evidence Level B]. Refer to recommendation 2.1 for additional information.
2.2.2
Blood pressure management
Blood pressure should be managed in all patients to reach optimal control as follows:
i.
For the prevention of first stroke in the general population the systolic blood pressure treatment goal is a pressure
level of consistently lower than 140 mm Hg [Evidence Level B]. The diastolic blood pressure treatment goal is a
pressure consistently lower than 90 mm Hg [Evidence Level A].
ii.
For patients who have had a stroke or transient ischemic attack, blood pressure lowering treatment is
recommended to achieve a target of consistently lower than 140/90 mm Hg [Evidence Level B].
iii.
In patients with diabetes, blood pressure lowering treatment is recommended for the prevention of first or recurrent
stroke to attain systolic blood pressure targets consistently lower than 130 mm Hg [Evidence Level B] and diastolic
blood pressure targets consistently lower than 80 mm Hg [Evidence Level A].
iv. In patients with nondiabetic chronic kidney disease, blood pressure lowering treatment is recommended for the
prevention of first or recurrent stroke to attain a blood pressure consistently lower than 140/90mm Hg [Evidence Level
C].
v.
For recommendations on specific agents and sequence of agents for the secondary prevention of stroke, refer to the
Canadian Hypertension Education Program treatment guidelines [Evidence Level C].50 Refer to Table 2.2.
vi. Randomized controlled trials have not defined the optimal time to initiate blood pressure lowering therapy after stroke
or transient ischemic attack. Blood pressure lowering treatment should be initiated or modified before discharge from
hospital [Evidence Level B]. Refer to recommendation 3.3 for blood pressure management during the acute phase of
stroke (0 – 72 hours).
Rationale
Elevated blood pressure is the single most important risk factor for stroke. One in five adult Canadians has blood pressure in the
range of 130–139/85–89 mm Hg (labeled by some investigators as “pre-hypertension”), and up to 60 percent of them will
develop hypertension within four years. Among persons aged 55 and older with normal blood pressure, 90 percent will develop
Fourth Edition FINAL
Update: September 2012
Page 13 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
hypertension if they live to an average age. All adults require ongoing assessment of blood pressure throughout their lives.50, 51
Each 1 mm Hg increase in blood pressure increases the risk of poor late-life cognitive function by approximately one percent.52,53
Epidemiologic studies have shown a graded increase in the risk of stroke as blood pressure increases.
Numerous population-based studies have found that elevated blood pressure is a significant risk factor for first and recurrent
stroke; hypertension is estimated to account for about 60 percent of the population-attributable risk for cerebrovascular disease.
The Interstroke study reported an odds ratio of 2.64 for patients with hypertension experiencing a stroke.54 A number of trials
have shown a 28 percent risk reduction in recurrent stroke in patients treated with blood pressure lowering medication.
The optimal target for blood pressure in people who have had a stroke and people at risk of stroke has not been formally defined
through randomized controlled trials. The current treatment recommendation is to attain a blood pressure of consistently lower
than 140/90 mm Hg for people who have had a cerebrovascular event. Epidemiologic data have shown that those with a
response to treatment attaining blood pressure levels well below 140 systolic and 90 diastolic have better outcomes yet these
treatment trials have not yet clearly defined how far blood pressure should be lowered.
System Implications
•
Coordinated hypertension awareness programs at the provincial and community levels that involve community groups,
primary care providers (physicians, nurse practitioners and pharmacists) and other relevant partners.
•
Stroke prevention, including routine blood pressure monitoring, offered by primary care providers in the community as
part of comprehensive patient management.
•
Increased availability and access to education programs for healthcare providers across the continuum of care on
hypertension diagnosis and management for adults and children
•
Increased programs for patients and families on home monitoring of blood pressure and blood pressure selfmanagement programs
Performance Measures
1. Proportion of persons at risk for stroke who had their blood pressure measured at their last healthcare
encounter.
2. Proportion of the population who have diagnosed elevated blood pressure (hypertension).
3. Proportion of the population who are aware of hypertension and the risks of high blood pressure.
4. Proportion of the population who report having hypertension.
5. Percentage of the population with known hypertension who are on blood pressure lowering therapy.
6. Proportion of the population with hypertension who are being treated and have achieved control of their blood pressure
within defined targets (as per Canadian Hypertension Education Program guidelines).
7. Proportion of stroke and transient ischemic attack patients who have received a prescription for blood pressure lowering
agents on discharge from acute care.
8. Proportion of stroke and transient ischemic attack patients who have received a prescription for blood pressure lowering
agents after assessment in a secondary prevention clinic.
Measurement Notes
•
Performance measures 1 through 4: data may be available through the Canadian Hypertension Education Program
database, the Canadian Community Health Survey, and other provincial and local health surveys and patient self-reports.
•
Performance measures 5 and 6: data may be available through audit of primary care physician charts. Prescription
information may also be available through provincial drug plan databases, although these may have limitations with
respect to the age of those covered by the plans, and there is variation across provinces and territories.
•
Performance measures 7 and 8: prescriptions for blood pressure lowering agents may be given during the inpatient stay
or during a secondary prevention assessment and follow- up. When tracking these performance rates, it is important to
record the setting where this therapy is initiated. Data sources may include physician order sheets, physicians’ or nurses’
notes, discharge summaries or copies of prescriptions given to patients.
•
Prescriptions given to a patient do not imply compliance.
•
Algorithms to identify incidence and prevalence of hypertension from administrative databases have been validated in
Canada and should be used for consistency in measurement when possible.104
Fourth Edition FINAL
Update: September 2012
Page 14 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
Implementation Resources and Knowledge Transfer Tools
o Canadian Hypertension Education Program: http://www.hypertension.ca/chep-recommendations
o Home monitoring and other Canadian Hypertension Education Program resources: http://hypertension.ca/measuringblood-pressure
o Canadian Task Force on Preventive Health Care for primary prevention screening guidelines for hypertension
http://www.canadiantaskforce.ca/recommendations__current_eng.html
o Aboriginal Hypertension Management Resources:
http://www.heartandstroke.on.ca/site/c.pvI3IeNWJwE/b.5339657/k.9CEB/HCP__Aboriginal_Hypertension_Manageme
nt_Program_Resources.htm
Summary of the Evidence
LINK to Evidence Tables for Blood Pressure Management
Hypertension is a major problem in nearly all countries around the world, including Canada, and it is the most important modifiable risk factor
for stroke. The INTERSTROKE study is an ongoing standardized case-controlled study of the contribution of specific risk factors to the burden
of stroke across 22 countries.54 The first report from this study, based on 3,000 stroke cases and 3,000 controls, identified five risk factors
which account for more than 80 percent of the risk for stroke, including hypertension, current smoking, abdominal obesity, diet, and physical
activity. Among these risk factors, hypertension was the most significant risk factor for stroke and contributed to 34.6 percent of the populationattributed risk (PAR), and this rose to 52 percent when measured blood pressures of greater than 160/90 mm Hg were added to the model.
When these results were compared to a similar study conducted for heart disease, hypertension was found to have a more significant impact
on stroke than on heart disease. The study further noted that “blood pressure is the most amenable to change in low-income settings because
screening programs need modest equipment and little specialized expertise. Additionally, blood pressure is readily reduced by inexpensive
generic drugs and non-pharmacological approaches (e.g., salt reduction)” (page 9).
Du and associates reported that some 20–30 percent of adult populations are affected with high blood pressure, as are over 60 percent of
people 65 years and older and about 70 percent of stroke patients.55 Hypertension is quantitatively the largest single risk factor for premature
death and disability, because of the large number of people afflicted and the consequences of uncontrolled hypertension.55Hypertension is
closely associated with the risk of total mortality and the risk of all types of stroke, coronary artery disease, diabetes and renal disease. No
other modifiable factor has been identified that contributes more to the development of stroke than hypertension. The authors further
emphasized that hypertension should not be regarded so much as a disease but more as one of the treatable or reversible risk factors for
premature death due to arterial disease.55 At least three-quarters of strokes in hypertensive patients are preventable by treatment of elevated
blood pressure. However, strokes are caused not by a single risk factor such as hypertension but by the interaction of multiple risk factors,
some having a stronger independent relationship with risk of stroke than others. The probability of stroke in an individual depends on the
presence and level of other risk factors.
The National Heart, Lung, and Blood Institute Joint National Committee on Prevention, Detection, Evaluation, and Treatment of High Blood
Pressure has defined normal blood pressure as less than 120/80 mm Hg.56 A continuous and linear relationship between blood pressure and
risk of stroke has been reported, which holds even in individuals with normal blood pressure. Lewington found that an increase of 20 mm Hg in
systolic and 10 mm Hg in diastolic blood pressure can lead to a two-fold increase in stroke mortality in persons aged 40 – 69.57 Weber reported
that the high sensitivity of the relationship between blood pressure and stroke risk is now more fully realized.58 Single studies do not always
have the power to identify the impact that blood pressure changes of only a few millimetres of mercury have on risk. However, a meta-analysis
of 61 studies with a total of more than 1 million participants, an average 12-year follow-up and 120 000 recorded deaths showed that each 2
mm Hg reduction in systolic blood pressure was associated with a 7 percent reduction in mortality from ischemic heart disease and a 10
percent reduction in mortality from stroke. A subsequent prospective study by Bestehorn et al found an overall 10-year stroke rate of 26
percent in patients with hypertension and for 16.7 percent f the cohort the risk was greater than 50 percent if they had additional comorbidities.59 In a meta-analysis of risk stratification for the prevention of cardiovascular complications of hypertension, Girerd and Giral
conclude that each reduction of 2 mm Hg in systolic blood pressure is associated with a 25 percent reduction in stroke events.60
A collaborative meta-analysis was conducted to assess the age-specific relevance of blood pressure to cause-specific mortality.57 Combining
61 prospective observational studies of blood pressure and vascular mortality, each difference of 20 mm Hg in systolic blood pressure (or,
approximately equivalently, 10 mm Hg in usual diastolic blood pressure) was associated with more than a two-fold difference in the stroke
death rate, without any evidence of a threshold down to at least 115/75 mm Hg for all vascular deaths. Age-specific associations were found to
be similar for men and women and for cerebral hemorrhage and cerebral ischemia.
Lee (2011) reported that patients with a systolic BP of 120 to 129 mmHg have a relative risk of stroke of 1.22, and this increases to a relative
risk of 1.79 when systolic blood pressure is between 130 and 139 mmHg.61 These increased risk patterns have been measured for recurrent
stroke and appear similar. Obviagele (2011) reported a linear relationship between systolic blood pressure and risk of recurrent stroke, with the
high SBP group (SBP 140-<150 mm Hg) (AHR, 1.23; 95% CI, 1.07-1.41), and in the very high SBP group (SBP > 150 mmHg) (AHR, 2.08;
95% CI, 1.83-2.37). 62
Fourth Edition FINAL
Update: September 2012
Page 15 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
A Cochrane meta-analysis by Musini and associates also examined the effects of age and blood pressure lowering on overall mortality,
cardiovascular mortality and morbidity and withdrawal due to adverse effects in people 60 years and older with mild to moderate systolic or
diastolic hypertension.63 Fifteen trials (24,055 subjects ≥ 60 years) with moderate to severe hypertension were included which mostly
evaluated first-line thiazide diuretic therapy for a mean duration of treatment of 4.5 years. Treatment reduced total mortality, RR 0.90 (0.84,
0.97); event rates per 1000 participants reduced from 116 to 104. Treatment also reduced total cardiovascular morbidity and mortality, RR 0.72
(0.68, 0.77); event rates per 1000 participants reduced from 149 to 106. In the three trials restricted to persons with isolated systolic
hypertension the benefit was similar. In very elderly patients ≥ 80 years the reduction in total cardiovascular mortality and morbidity was similar
RR 0.75 [0.65, 0.87] however, there was no reduction in total mortality, RR 1.01 [0.90, 1.13]. Withdrawals due to adverse effects were
increased with treatment, RR 1.71 [1.45, 2.00]. The authors concluded that treating healthy persons (60 years or older) with moderate to
severe systolic and/or diastolic hypertension reduces all cause mortality and cardiovascular morbidity and mortality. The decrease in all cause
mortality was limited to persons 60 to 80 years of age.
The relationship between blood pressure and cardiovascular risk is “continuous, consistent, and independent of other risk factors. The
American Heart Association guidelines for the primary prevention of ischemic stroke report that the higher the blood pressure, the greater the
stroke risk.64 The working group acknowledged the benefit of treatment of hypertension for the primary prevention of stroke and concluded that
the reduction of blood pressure is generally more important than the agent used to aid in this goal.
Hypertensive patients with a history of cerebral vascular disease are at particularly high risk of stroke recurrence. Gueyffier and associates
performed a meta-analysis using all available randomized controlled clinical trials assessing the effect of blood pressure lowering drugs on
clinical outcomes (recurrence of stroke, coronary events, cause-specific and overall mortality) in patients with prior stroke or transient ischemic
attack.65 Nine trials that included a total of 6752 patients were identified, and it was found that the recurrence of stroke, fatal and nonfatal, was
significantly reduced in treatment groups compared with control groups consistently across the different sources of data (RR = 0.72, 95% CI
0.61–0.85). There was no evidence that this intervention induced serious adverse effects.
For several reasons, categorizing patients as “hypertensive” or “normotensive” based on an arbitrary blood pressure threshold may not be
helpful with respect to secondary stroke prevention. First, the relationship between blood pressure and stroke is continuous and graded, with
no evidence of a lower blood pressure threshold for stroke risk. Second, several controlled trials have demonstrated that blood pressure
reduction benefits patients who would not normally be designated as hypertensive (Heart Outcomes Prevention Evaluation
[HOPE],67PROGRESS68). Blood pressure lowering therapy reduces the risk of vascular events across a wide spectrum of initial blood
pressures.66-68
Angiotensin receptor blockers have demonstrated efficacy for the prevention of stroke in both the primary and secondary prevention settings.
Three recently completed trials of angiotensin receptor blockers were the Losartan Intervention For Endpoint Reduction Study (LIFE),69 the
Acute Candesartan Cilexetil Therapy in Stroke Survivors Study (ACCESS pilot study),70 and the Study on Cognition and Prognosis in the
Elderly (SCOPE).71 All 3 trials demonstrated consistent relative risk reductions for stroke in the range of 24 percent to 34 percent, despite the
enrolment of different patient populations, the use of varying angiotensin receptor blockers and differing interventions in the control group
(placebo-based or conventional therapy).
The Ongoing Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial (ONTARGET) compared the ACE inhibitor ramipril, the
angiotensin-receptor blocker telmisartan and the combination of the 2 drugs in patients with vascular disease or high-risk diabetes.72 Patients
underwent double-blind randomization, with 8576 assigned to receive 10 mg of ramipril per day, 8542 assigned to receive 80 mg of telmisartan
per day and 8502 assigned to receive both drugs (combination therapy). The primary composite outcome was death from cardiovascular
causes, myocardial infarction, stroke or hospitalization for heart failure. The researchers found that telmisartan was equivalent to ramipril in
patients with vascular disease or high-risk diabetes and was associated with less angioedema. The combination of the 2 drugs was associated
with more adverse events without an increase in benefit.
Studies of hypertension management in the very elderly have emerged in the past few years.73,74 The HYVET study involving 3845 patients
examined the benefit of antihypertensive treatment for hypertensive patients who were 80 years or older.73 Patients were randomly assigned
to receive either antihypertensive therapy or matching placebo. The results showed that lowering mean blood pressure by 15.0/6.1 mm Hg was
associated with a 30 percent reduction in the rate of fatal or nonfatal stroke (95% CI –1% to 51%, p = 0.06), a 39 percent reduction in the rate
of death from stroke (95% CI 1% to 62%, p = 0.05), a 21 percent reduction in the rate of death from all causes (95% CI 4% to 35%, p = 0.02)
and a 23 percent reduction in the rate of death from cardiovascular causes (95% CI –1% to 40%, p = 0.06). Fewer serious adverse events were
reported in the active treatment group (358 v. 448 in the placebo group, p = 0.001). The authors concluded that antihypertensive treatment in
patients 80 years of age or older was beneficial.
An open-label active treatment extension of the HYVET study included 1682 patients both from the original treatment and the placebo group
(Beckett et al 2012).74 All patients followed the medication regime as the original trial (indapamide SR 1.5 mg (plus perindopril 2-4 mg if
required), had the same target blood pressure levels of less than 150/80 mm Hg, and measured the same end point of stroke, all cause
mortality, cardiovascular mortality, and other cardiovascular events. No differences were found between the groups, demonstrating that both
early and long-term antihypertensive therapies are beneficial to elderly patients.
Fourth Edition FINAL
Update: September 2012
Page 16 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
Launer and coworkers assessed the long-term relationship of midlife blood pressure levels to late-life cognitive function in the surviving cohort
members of the prospective Honolulu Heart Program.76 The subjects were 3735 Japanese American men living in Hawaii either in the
community or in institutions, with an average age of 78 years at the fourth examination. Cognitive function, measured by the 100-point
Cognitive Abilities Screening Instrument, was categorized as good (reference category, with score of 92 to 100), intermediate (score < 92 to 82)
and poor (score < 82). Midlife systolic blood pressure and diastolic blood pressure values were measured in 1965, 1968 and 1971. A
respondent was classified into one of the following categories if 2 of 3 measurements fell into the following groups: for systolic blood pressure,
< 110, 110 to 139, 140 to 159 and ≥ 160 mm Hg; and for diastolic blood pressure, < 80, 80 to 89, 90 to 94 and 95 mm Hg. The risk for
intermediate and poor cognitive function increased progressively with increasing level of midlife systolic blood pressure category (p for trend <
0.03 and < 0.001, respectively) when controlled for age and education. For every 10 mm Hg increase in systolic blood pressure there was an
increase in risk for intermediate cognitive function of 7 percent (95% CI 3%–11%) and for poor cognitive function of 9 percent (95% CI 3%–
16%). The level of cognitive function was not associated with midlife diastolic blood pressure. The authors concluded that early control of
systolic blood pressure levels may reduce the risk for cognitive impairment in old age.
Fourth Edition FINAL
Update: September 2012
Page 17 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
Figure 2.2.1 Canadian Hypertension Education Program (2012)
Permission received for reproduction of this Figure, Canadian Hypertension Education Program, September 2012.
Fourth Edition FINAL
Update: September 2012
Page 18 of 69
CBPRSC 2012
Section 2: Prevention
2.2 Blood Pressure Management
Table 2.2.1 CHEP Hypertension Pharmacomanagement Table (2012)
Permission received for reproduction of this Table, Canadian Hypertension Education Program, September 2012.
Fourth Edition FINAL
Update: September 2012
Page 19 of 69
CBPRSC 2012
Section 2: Prevention
2.3 Lipid Management
Best Practice Recommendation 2.3 Lipid Management
* Moderate revisions for 2012
Patients who have had an ischemic stroke or transient ischemic attack should have their serum lipid levels assessed and
aggressively managed [Evidence level A].
2.3.1
i.
ii.
Lipid assessment
Fasting lipid levels (total cholesterol, total triglycerides, low-density lipoprotein [LDL] cholesterol, high-density lipoprotein
[HDL] cholesterol) should be measured on all patients presenting with stroke or TIA [Evidence Level C].
For management of dyslipidemia in the primary prevention of cardiovascular events, including stroke, refer to the current
Canadian Cardiovascular Society Dyslipidemia clinical practice guidelines.78
2.3.2
i.
Lipid management
A statin drug should be prescribed for primary prevention of cardiovascular events, including stroke, to most patients with
high global cardiovascular risk [Evidence Level A].
ii. A statin drug should be considered for primary prevention of cardiovascular events, including stroke, for those patients at
intermediate cardiovascular risk [Evidence Level B]. Refer to Canadian Cardiovascular Guidelines for Dyslipidemia 2012
for additional information.
iii. A statin drug should be prescribed as secondary prevention to most patients who have had an ischemic stroke or
transient ischemic attack in order to achieve an LDL cholesterol of less than 2.0 mmol/L, or a 50% reduction in LDL
cholesterol from baseline [Evidence Level B]
iv. Patients with ischemic stroke or transient ischemic attack should be managed with aggressive therapeutic lifestyle
changes, including dietary modification, as part of a comprehensive approach to lower risk of first or recurrent stroke
[Evidence Level A].49
v. Statin therapy is not indicated for prevention of intracerebral hemorrhage. For intracerebral hemorrhage patients who
have a clear concomitant indication for cholesterol lowering treatment, statin therapy should be individualized and should
take into account the patient’s overall thrombotic risk as well as the possibility of increased risk of intracerebral
hemorrhage on statin therapy [Evidence Level B].
vi. Statin therapy has not been well studied in certain sub-populations of stroke patients (for example, patients over 80 years
of age, patients with cardioembolic stroke, arterial dissection). Decisions for prescribing should be based on current
health status, comorbidities and other indicators of systemic vascular disease (such as coronary artery disease,
peripheral vascular disease, and renal vascular disease)[Evidence Level C].
Rationale
High cholesterol and lipids in the blood are associated with a higher risk of vascular events including stroke and myocardial
infarction. People who have already had an ischemic stroke or transient ischemic attack will benefit from cholesterol-lowering
medications with a statin class of drug. Aggressive reduction of low-density lipoprotein cholesterol is likely to yield greater benefit
than more modest reductions. A 20 to 30 percent relative risk reduction has been reported in recurrent vascular events for
patients with a history of stroke without coronary artery disease who are treated with statin agents.
The Cholesterol Treatment Trialists meta-analysis of 14 statin trials showed a dose-dependent relative reduction in
cardiovascular disease with low-density lipoprotein cholesterol lowering. Every 1.0 mmol/L reduction in low-density lipoprotein
cholesterol is associated with a corresponding 20 to 25 percent reduction in cardiovascular disease mortality and nonfatal
myocardial infarction.77
With the childhood obesity epidemic, dyslipidemia is becoming a growing issue in paediatric stroke cases; therefore, fasting lipid
panels should be part of the assessment of paediatric stroke cases.
Note: The current clinical trial evidence does not include enough stroke patients with atrial fibrillation or other cardioembolic
sources to make specific recommendations for this patient population. The decision to use statins in this setting should be based
on the patient's global cardiovascular risk. It is unclear whether statins are of benefit in patients with a combination of atrial
fibrillation and stroke.
Fourth Edition FINAL
Update: September 2012
Page 20 of 69
CBPRSC 2012
Section 2: Prevention
2.3 Lipid Management
System Implications
•
Coordinated dyslipidemia awareness programs at the provincial and community levels that involve community groups,
primary care providers (including physicians, nurse practitioners and pharmacists), and other relevant partners.
•
Stroke prevention, including lipid level monitoring offered by primary care providers in the community as part of
comprehensive patient management.
•
Increased availability and access to education programs for healthcare providers across the continuum of care on
dyslipidemia diagnosis and management
•
Continued alignment with recommendations and guidelines developed by the Canadian Cardiovascular Society
Dyslipidemia group.
Performance Measures
1. Proportion of stroke patients prescribed lipid-lowering agents for secondary prevention of stroke, either at
discharge from acute care, through a secondary prevention clinic or by primary care physician.
2. Proportion of the population who report that they have elevated lipid levels, especially low-density lipoprotein.
3. Proportion of stroke patients with a low-density lipoprotein cholesterol between 1.8 and 2.5 mmol/L at three months
following the stroke event.
4. Proportion of stroke patients who have lipid levels completed as part of initial comprehensive assessment.
5. Proportion of stroke patients who have lipid levels completed as part of initial comprehensive assessment with elevated
lipid levels that require and receive treatment.
Measurement Notes
•
Performance measures 1 and 2: Data may be available through the Canadian Community Health Survey.
•
Performance measure 2: Data sources may include physician order sheets, physicians’ and nurses’ notes, discharge
summaries, or copies of prescriptions given to patients.
•
Performance measure 3: Blood values should be taken from official laboratory reports where possible.
•
Prescriptions for lipid-lowering agents may be given during the inpatient stay or during a secondary prevention
assessment and follow-up, either in a stroke prevention clinic or in a primary care setting. When tracking these
performance rates, it is important to record the setting where this therapy was initiated.
•
Prescriptions given to a patient do not imply compliance.
Implementation Resources and Knowledge Transfer Tools
o Canadian Cardiovascular Society Dyslipidemia Recommendations:
(http://www.ccs.ca/download/consensus_conference/consensus_conference_archives/2009_DyslipidemiaGuidelines.pdf)
o Heart and Stroke Foundation of Canada:
http://www.heartandstroke.com/site/c.ikIQLcMWJtE/b.3484027/k.8419/Heart_disease__High_blood_cholesterol.htm
o Dieticians of Canada: http://www.dietitians.ca/Nutrition-Resources-A-Z/Factsheets/Heart-Health/Heart-Healthy-Eating-Cholesterol.aspx
o Framingham Cardiovascular Risk Calculator: http://www.framinghamheartstudy.org/risk/gencardio.html#
o Cholesterol Levels Calculator:
http://bodyandhealth.canada.com/health_tools.asp?t=12&text_id=2751&channel_id=10&relation_id=10864
o National Heart, Lung and Blood Institute Patient Educational Materials:
http://www.nhlbi.nih.gov/health/public/heart/index.htm#chol
o Live Strong: http://www.livestrong.com/article/288004-how-to-teach-about-high-cholesterol/
Summary of the Evidence
LINK to Evidence Tables for Lipid Management
The causal relationship between dyslipidemia and atherosclerosis is well documented. Screening and appropriate management of dyslipidemia
by healthcare providers is imperative in both primary and secondary prevention of coronary artery disease, peripheral vascular disease and
stroke.77 The 2009 update of the Canadian dyslipidemia guidelines provides a detailed description of the current recommended treatment levels
and management modalities for dyslipidemia.78 They emphasize a need to balance lifestyle and risk factor modifications through behaviors
change with pharmacological intervention to maximize treatment and improve outcomes for cardiovascular disease and stroke.
Fourth Edition FINAL
Update: September 2012
Page 21 of 69
CBPRSC 2012
Section 2: Prevention
2.3 Lipid Management
Several systematic reviews of lipid-lowering therapies have affirmed the following points: (1) the relative reduction in stroke risk is on the order
of 25–30 percent, (2) ischemic stroke is reduced, with little effect on hemorrhagic stroke and (3) the relative reduction in stroke events is
constant irrespective of the baseline risk of stroke. The latter indicates that a greater absolute benefit may accrue from treating patients with a
history of stroke or transient ischemic attack, who have a markedly higher baseline risk of recurrent cerebrovascular events. 79-84
O’Regan and collaborators conducted a comprehensive review of randomized trials evaluating statin therapy for stroke prevention.80 Data were
pooled using a random-effects model, and meta-regression techniques were employed. Following a thorough search, 42 trials assessing statin
therapy for all-stroke prevention (n = 121 285) were included, resulting in a pooled RR of 0.84 (95% CI 0.79–0.91). The pooled relative risk of
statin therapy for all-cause mortality (n = 116 080) was 0.88 (95% CI 0.83–0.93). Each unit increase in LDL resulted in a 0.3 percent increased
RR of death (p = 0.02). Seventeen trials evaluated the effect of statins on cardiovascular death (n = 57 599, RR 0.81, 95% CI 0.74–0.90), and
11 evaluated non-hemorrhagic cerebrovascular events (n = 58 604, RR 0.81, 95% CI 0.69–0.94). Eleven trials reported hemorrhagic stroke
incidence (total n = 54 334, RR 0.94, 95% CI 0.68–1.30), and 21 trials reported on fatal strokes (total n = 82 278, RR 0.99, 95% CI 0.80–
1.21).80 Only one trial reported on statin therapy for secondary prevention. Statin therapy provides high levels of protection for all-cause
mortality and non-hemorrhagic strokes, reinforcing the need to consider prolonged statin treatment for patients at high risk of major vascular
events, but a need for caution remains for patients at risk of bleeds. A large meta-analysis of various lipid-lowering therapies (including statins,
fibrates, niacin, bile acid sequestrants and diet) found that only statins reduced the risk of stroke, with a risk reduction of 26 percent (95% CI
14%–36%) for secondary prevention.81 Non-statin drug therapy (with 32 550 subjects studied, of whom 73% were randomized in trials
employing fibrates) was associated with a nonsignificant risk reduction of seven percent (RR 0.93, 95% CI 0.79–1.08).
The Heart Protection Study contributed a substantial amount of information about the role of statin therapy in persons at high risk of serious
vascular events.82 This study randomized 20,536 patients with a total serum cholesterol of > 3.4 mmol/L to simvastatin or placebo for a mean
duration of five years. The inclusion criteria were any of the following: coronary artery disease, cerebrovascular disease, peripheral vascular
disease, diabetes or patients over 65 years with hypertension. The study showed that simvastatin 40 mg once daily rapidly produced a definite
and substantial reduction in ischemic stroke (relative risk reduction 25 percent; 95% CI 15%– 44%), irrespective of the patient’s age, sex or
blood lipid concentrations when treatment was initiated.82 It also demonstrated that statin therapy reduced the risk of major vascular events
among people who have previously had a stroke or other cerebrovascular event, even if they did not already have manifest coronary disease.
In addition, there was a highly significant reduction in the simvastatin arm in the frequency of carotid endarterectomy and angioplasty. These
benefits were evident in every subgroup tested: patients who had or did not have coronary artery disease; those with cerebrovascular disease,
peripheral vascular disease or diabetes; men or women; those over or under 75 years at entry; and those whose LDL cholesterol was over or
under 2.6 mmol/L. Treatment benefits were independent of the baseline cholesterol level. The results of the Heart Protection Study imply that
the initiation of statin therapy should be based more on the assessment of a patient’s absolute risk of cardiovascular disease, rather than just
the baseline LDL cholesterol concentration.
The Stroke Prevention by Aggressive Reduction in Cholesterol Levels trial (SPARCL) randomly assigned 4731 patients who had had a stroke
or transient ischemic attack within one to six months before study entry, had LDL levels of 2.6 to 4.9 mmol/L and had no known coronary artery
disease to double-blind treatment with atorvastatin 80 mg once daily or placebo.83 The mean LDL level during the trial was 1.9 mmol/L among
patients receiving atorvastatin and 3.3 mmol/L in the placebo group. The 5-year absolute reduction in risk of any stroke was 2.2 percent;
relative risk reduction of 16%, adjusted hazard ratio (HR) 0.84 (95% CI 0.71–0.99; p = 0.03). The reduction in ischemic stroke (HR 0.78, 95%
CI 0.66–0.94) should be weighed against the statistically significant increase in hemorrhagic stroke (HR 1.66, 95% CI 1.08 – 2.55). The fiveyear absolute reduction in risk of major cardiovascular events was 3.5 percent (HR 0.80, 95% CI 0.69–0.92; p = 0.002). The statistically
significant increase in hemorrhagic stroke, not seen in other statin trials, remains unexplained.83
In the second Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL2) study, atorvastatin 80 mg/day reduced the risk of
stroke in patients with recent stroke or transient ischemic attack.84 This overall benefit included an increase in the numbers of treated patients
having hemorrhagic stroke (n = 55 for active treatment v. n = 33 for placebo), prompting investigators to further explore the relationships
between hemorrhage risk and treatment, baseline patient characteristics, most recent blood pressure and most recent LDL cholesterol levels
before the hemorrhage. Of 4731 patients, two percent had hemorrhagic strokes as entry events.85 In addition to atorvastatin treatment (HR
1.68, 95% CI 1.09–2.59; p = 0.02), Cox multivariable regression showed that hemorrhagic stroke risk was higher in those having a hemorrhagic
stroke as the entry event (HR 5.65, 95% CI 2.82– 11.30; p < 0.001), in men (HR 1.79, 95% CI 1.13–2.84; p = 0.01) and with age (10-yr
increments, HR 1.42, 95% CI 1.16– 1.74; p < 0.001). There were no statistical interactions between these factors and treatment. Multivariable
analyses also found that having stage 2 (JNC-7) hypertension at the last study visit before a hemorrhagic stroke increased risk (HR 6.19, 95%
CI 1.47–26.11; p = 0.01), but there was no effect of most recent LDL cholesterol level in those treated with atorvastatin.
Fourth Edition FINAL
Update: September 2012
Page 22 of 69
CBPRSC 2012
Section 2: Prevention
2.4 Diabetes Management
Best Practice Recommendation 2.4 Diabetes Management
* Minor revisions for 2012
Patients with diabetes who have had an ischemic stroke or transient ischemic attack should have their diabetes assessed and
optimally managed [Evidence level A].
2.4.1 Diabetes assessment
i. For patients with diabetes and either ischemic stroke or transient ischemic attack, glycated hemoglobin (HbA1C )
should be measured as part of a comprehensive stroke assessment [Evidence Level B].
ii. In all patients with stroke or TIA, fasting lipid levels (total cholesterol, high-density lipoprotein cholesterol, total
glycerides and calculated low-density lipoprotein cholesterol) should be measured at the time of diagnosis and at
appropriate intervals if therapy initiated [Evidence Level C].
iii. Blood pressure should be measured at every diabetes visit in patients with stroke or at risk of stroke [Evidence Level
C].
2.4.2 Diabetes management
i. Glycemic targets must be individualized; however, therapy in most patients with type 1 or type 2 diabetes and stroke
or TIA should be treated to achieve a glycated hemoglobin (HbA1C) level ≤7.0 percent to reduce the risk of
microvascular complications [Evidence Level A] and, in individuals with type 1 diabetes, macrovascular complications
[Evidence Level C].
ii. To achieve an HbA1C ≤7.0%, patients with type 1 or type 2 diabetes should aim for a fasting plasma glucose or
preprandial plasma glucose target of 4.0 to 7.0 mmol/L [Evidence Level B].
iii. The 2-hour postprandial plasma glucose target is 5.0 to 10.0 mmol/L [Evidence Level B]. If HbA1C targets cannot be
achieved with a postprandial target of 5.0 to 10.0 mmol/L, further postprandial blood glucose lowering, to 5.0 to 8.0
mmol/L, can be considered [Evidence Level C].
iv. Adults with diabetes and ischemic stroke are at high risk of further vascular events and should also be treated with a
statin to achieve a low-density lipoprotein cholesterol ≤2.0 mmol/L [Evidence Level A].
v. Unless contraindicated, low dose acetylsalicylic acid therapy (81 to 325 mg/day) is recommended in all patients with
diabetes with evidence of cardiovascular disease such as stroke [Evidence Level A].
Rationale
Diabetes is a major risk factor for cardiovascular disease and is recognized as an independent risk factor for ischemic stroke.86
Most adults with type 1 or type 2 diabetes should be considered at high risk for vascular disease. The exceptions are younger
adults with type 1 and type 2 diabetes with shorter duration of disease and without complications of diabetes (including
established cardiovascular disease) and without other cardiovascular disease risk factors. Diabetes increases the risk of stroke
and is a particularly potent risk factor in younger individuals, with studies suggesting an increase in stroke risk of as much as 10fold in some younger subgroups. Overall, diabetes is considered a major risk factor for many conditions and is considered here
as part of a comprehensive package supporting prevention and lifestyle management.
System Implications
•
Coordinated diabetes awareness programs at the provincial and community levels that involve community groups,
primary care providers (including physicians, nurse practitioners and pharmacists), and other relevant partners.
•
Coordinated education and support programs for persons with diabetes to increase compliance and reduce ongoing risks
for cardiovascular complications.
•
Increased availability and access to education programs for healthcare providers across the continuum of care on
management of patients with diabetes and stroke
•
Continued alignment with recommendations and guidelines developed by the Canadian Diabetes Association.
Performance Measures
1. Proportion of persons with diabetes presenting to hospital with a new stroke event.
2. Proportion of the population with a confirmed diagnosis of diabetes (type 1 and type 2).
3. Proportion of patients presenting to hospital with a stroke who receive a subsequent diagnosis of diabetes while in
Fourth Edition FINAL
Update: September 2012
Page 23 of 69
CBPRSC 2012
Section 2: Prevention
2.4 Diabetes Management
hospital for stroke care.
Measurement Notes
•
Data sources may include physician order sheets, physicians’ or nurses’ notes, discharge summaries, or copies of
prescriptions given to patients.
•
Blood values should be taken from official laboratory reports where possible.
•
Trends and benchmarks may be monitored and tracked through the National Diabetes Surveillance System data.
•
Performance measure 2: Rates may be obtained for Canada from the Public Health Agency of Canada Diabetes
Surveillance database.
Implementation Resources and Knowledge Transfer Tools
o Canadian Diabetes Association - http://www.diabetes.ca/
o Canadian Diabetes Association Clinical Practice Guidelines http://www.diabetes.ca/for-professionals/resources/2008cpg/
Summary of the Evidence
LINK to Evidence Tables for Diabetes Management in Stroke and TIA Patients
Diabetes is an important modifiable risk factor for a first ischemic stroke. In a review of stroke and diabetes, Idris and colleagues stated that the
combination of diabetes and stroke is a major cause of morbidity and mortality worldwide.87 Evidence from large clinical trials performed in
patients with diabetes supports the need for aggressive and early intervention to target patients’ cardiovascular risks to prevent the onset,
recurrence and progression of acute stroke. They describe the epidemiology of diabetes and stroke, and report an estimate that the risk of
stroke is increased 1.5- to three-fold for patients with diabetes. Diabetes also doubles the risk of stroke recurrence, and stroke outcomes are
significantly worse among patients with diabetes, with increased hospital and long-term stroke mortality, more residual neurologic and
functional disability and longer hospital stays. From a clinical perspective, diabetes increases the risk of ischemic stroke more than
hemorrhagic stroke, resulting in a greater ischemic to hemorrhagic stroke ratio in people with diabetes compared with the general population.
Idris and colleagues further reported that although strokes in patients with diabetes are associated with a worse outcome, there is no evidence
to suggest that diabetes induces a larger area of cerebral infarction.
The high stroke risk in diabetes may be due to the complex interplay between the various hemodynamic and metabolic components of the
diabetes syndrome. Other than the many recognized risk factors associated with acute stroke (e.g., hypertension, dyslipidemia and atrial
fibrillation), specific risk factors attributable to diabetes have also been reported. Components of the metabolic syndrome such as insulin
resistance, central obesity, impaired glucose tolerance and hyperinsulinemia, both individually and collectively, are associated with an excess
risk of stroke disease.87
Many diabetes patients exhibit metabolic syndrome and these additional risk factors, such as raised hypertension and cholesterol, multiply the
overall risk. Reducing these risk factors to target levels is essential and requires a multifactor approach. Lifestyle changes, tight glycemic
control, antiplatelet drugs (ASA) and control of lipid levels, (e.g., using statins), can all have significant beneficial effects. Blood pressure control
is another vital aspect in reducing risk, and a number of recent studies have provided evidence supporting the use of ACE inhibitors as first-line
treatment in patients with diabetes.
Karapanayiotides and collaborators reported that the Framingham Study found a 2.5-fold incidence of ischemic stroke in diabetic men and a
3.6-fold incidence in diabetic women.88 In the largest case–control study with adjustment for multiple known risk factors, the risk of ischemic
stroke for diabetic individuals was increased 2.3-fold. Two other large studies reported similar findings with odds ratios (ORs) of 2.12 and 2.47.
However, it is difficult to determine the level of association between diabetes and ischemic stroke, as diabetes is also associated with a two-fold
higher incidence of hypertension and cardiac disease and with an increased incidence of asymptomatic carotid artery disease and
hyperlipidemia. Karapanayiotides and collaborators concluded that other risk factors for stroke such as hypertension, hypercholesterolemia,
cardiac ischemic disease and vascular claudication are significantly more frequent in diabetic individuals, confirming that diabetic patients have
high cerebral and cardiovascular risk.
Lehto and coworkers conducted a seven-year follow-up study on diabetic patients and nondiabetic controls to assess risk for stroke.89 They
found diabetic men had a two- to three-fold higher risk, and diabetic women a five-fold higher risk for stroke than corresponding nondiabetic
subjects (men: OR 2.4, 95% CI 1.2–4.9 in East Finland; OR 3.3, 95% CI 1.6–6.9 in West Finland; women: OR 5.5, 95% CI 2.4–12.9 in East
Finland; OR 5.4, 95% CI 2.3–12.6 in West Finland).133 Ischemic stroke was the most common cause of stroke in nondiabetic subjects and type
2 diabetes patients in both areas. High fasting plasma glucose was a risk factor for stroke even after adjustment for other variables. In addition
to fasting plasma glucose, glycemic control was also assessed by HbA1c, which reflects hyperglycemia during the preceding two months. There
was a dose–response relationship between HbA1c and risk of stroke. The duration of diabetes was also an important risk factor for stroke
events in type 2 subjects In addition, low levels of HDL cholesterol (less than 0.90 mmol/L), high levels of total triglyceride (more than 2.30
Fourth Edition FINAL
Update: September 2012
Page 24 of 69
CBPRSC 2012
Section 2: Prevention
2.4 Diabetes Management
mmol/L) and the presence of hypertension were associated with a 2-fold increase in the risk of stroke mortality or morbidity.
The Treating to New Targets study showed that intensive lipid-lowering therapy with atorvastatin 80 mg/day provides significant clinical benefit
beyond that afforded by atorvastatin 10 mg/day in patients with stable coronary artery disease.90 A total of 1501 patients with diabetes and
coronary artery disease, with LDL cholesterol levels of < 3.36 mmol/L, were randomized to double-blind therapy with either atorvastatin 10 (n =
753) or 80 (n = 748) mg/day. Patients were followed for a median of 4.9 years. The primary end point was the time to first major cardiovascular
event, defined as death from coronary heart disease, nonfatal non–procedure related myocardial infarction, resuscitated cardiac arrest, or fatal
or nonfatal stroke. The results found end-of-treatment mean LDL cholesterol levels were 2.55 mmol/L with atorvastatin 10 mg and 1.99 mmol/L
with atorvastatin 80 mg. A primary event occurred in 135 patients (17.9%) receiving atorvastatin 10 mg, compared with 103 patients (13.8%)
receiving atorvastatin 80 mg (HR 0.75, 95% CI 0.58–0.97; p = 0.026). Significant differences between the groups in favour of atorvastatin 80
mg were also observed for time to cerebrovascular event (HR 0.69, 95% CI 0.48–0.98; p = 0.037) and any cardiovascular event (HR 0.85, 95%
CI 0.73–1.00; p = 0.044). There were no significant differences between the treatment groups in the rates of treatment-related adverse events
and persistent elevations in liver enzymes. The researchers concluded that among patients with clinically evident coronary artery disease and
diabetes, intensive therapy with atorvastatin 80 mg significantly reduced the rate of major cardiovascular events by 25 percent compared with
atorvastatin 10 mg.
The Action to Control Cardiovascular Risk in Diabetes Study (ACCORD) investigators assessed whether intensive therapy to target normal
HbA1c levels would reduce cardiovascular events in patients with type 2 diabetes who had either established cardiovascular disease or
additional cardiovascular risk factors.91 Patients (n = 10 251) with a median HbA1c level of 8.1 percent were randomly assigned to receive
intensive therapy (targeting an HbA1c level below 6.0 percent) or standard therapy (targeting a level from 7.0 percent to 7.9 percent). The
finding of higher mortality in the intensive- therapy group led to a discontinuation of intensive therapy after a mean of 3.5 years of follow-up.
During follow- up, the primary outcome occurred in 352 patients in the intensive-therapy group, as compared with 371 in the standard- therapy
group (HR 0.90, 95% CI 0.78–1.04; p = 0.16). At the same time, 257 patients in the intensive-therapy group died, as compared with 203
patients in the standard therapy group (HR 1.22, 95% CI 1.01–1.46; p = 0.04).135 These findings identify a previously unrecognized harm of
intensive glucose lowering in high-risk patients with type 2 diabetes.
The Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation (ADVANCE) trial randomly
assigned patients (n = 11 140) with type 2 diabetes to undergo either standard glucose control or intensive glucose control, defined as the use
of gliclazide (modified release) plus other drugs as required to achieve an HbA1c value of 6.5% or less.92 After a median of 5 years of follow- up,
the mean glycated hemoglobin level was lower in the intensive-control group (6.5%) than in the standard-control group (7.3%). Intensive control
reduced the incidence of combined major macrovascular and microvascular events (18.1% v. 20.0% with standard control; HR 0.90, 95% CI
0.82–0.98; p = 0.01), as well as that of major microvascular events (9.4% v. 10.9%; HR 0.86, 95% CI 0.77–0.97; p = 0.01), primarily because of
a reduction in the incidence of nephropathy (4.1% v. 5.2%; HR 0.79, 95% CI 0.66–0.93; p = 0.006), with no significant effect on retinopathy (p =
0.50).
Fourth Edition FINAL
Update: September 2012
Page 25 of 69
CBPRSC 2012
Section 2: Prevention
2.5 Antiplatelet Therapy
Best Practice Recommendation 2.5
Antiplatelet Therapy
* Minor revisions for 2012
All patients with ischemic stroke or transient ischemic attack should be prescribed antiplatelet therapy for secondary prevention
of recurrent stroke unless there is an indication for anticoagulation [Evidence Level A].
i.
Acetylsalicylic acid (81 mg to 325 mg), combined acetylsalicylic acid (25 mg) and extended-release dipyridamole
(200 mg), or clopidogrel (75 mg) are all appropriate options and selection should depend on the clinical
circumstances [Evidence Level A].
For adult patients on acetylsalicylic acid, most patients should be on a maintenance dose of 81 mg/day unless
other indications are present which may suggest a higher dose is required [Evidence Level A].
ii.
In children with stroke the usual maintenance dosage of acetylsalicylic acid is 1 to 5 mg/kg per day for the
prevention of recurrent stroke [Evidence Level B]. The usual maximum dose is 81 mg/day.
iii.
The evidence for clopidogrel use in children is sparse at this time. Clopidogrel may be considered an alternative for
adolescents at a dose of 1 mg/kg/day up to a maximum of 75 mg/day. Younger children may have higher antiplatelet effects of clopidogrel, and the suggested doses should be considered within the range of 0.2 - 0.5
mg/kg/day [Evidence Level C].93
iv.
Short-term concurrent use of acetylsalicylic acid and clopidogrel (up to 90 days) has not shown an increased risk of
bleeding [Evidence Level B]; however, longer-term use is not recommended for secondary stroke prevention,
unless there is an alternate indication (e.g., drug-eluting carotid artery stent requiring dual antiplatelet therapy), due
to an increased risk of bruising and bleeding [Evidence Level A]. 104a 104b match sps3
v.
At the present time, there is not enough evidence to guide management if a patient has a stroke while on a specific
antiplatelet agent. Some clinicians may choose to switch to an alternate antiplatelet agent. In all cases other
vascular risk factors should be aggressively managed [Evidence Level C].
Refer to recommendation 2.6 for additional information on combination therapy for patients with stroke and atrial
fibrillation.
Rationale
Antiplatelet agents are considered a fundamental component of secondary stroke prevention.95 Several clinical trials have
shown that antiplatelet medications (such as acetylsalicylic acid) reduce the risk of further vascular events after transient
ischemic attack or ischemic stroke (25 percent relative risk reduction).138 This effect is modest and is clinically useful because
antiplatelet therapy is tolerated by the majority of patients who have had a transient ischemic attack or ischemic stroke. Trials
comparing different antiplatelet therapy regimes show quite small absolute differences in efficacy, rendering the options
equivocal.
System Implications
•
Stroke prevention clinics to improve secondary stroke prevention (effective, consistent prevention with early recognition
of risk factors and timely, targeted interventions).
•
Optimization of strategies at the local, regional and provincial levels to prevent the recurrence of stroke.
•
Stroke prevention awareness and education about secondary prevention for primary care practitioners and specialists
who manage stroke patients during the acute phase and after discharge from acute care.
Implementation Resources and Knowledge Transfer Tools
o Canadian Cardiovascular Society
o CHEST Guidelines
Performance Measures
1. Proportion of acute ischemic stroke and TIA patients who receive acute antiplatelet therapy within the first 48
hours of hospital arrival (core).
2. Proportion of patients with ischemic stroke or transient ischemic attack prescribed antiplatelet therapy on
discharge from acute care (core).
3. Proportion of patients with ischemic stroke or transient ischemic attack prescribed antiplatelet therapy on
discharge from secondary prevention clinic care (core).
Fourth Edition FINAL
Update: September 2012
Page 26 of 69
CBPRSC 2012
Section 2: Prevention
2.5 Antiplatelet Therapy
Measurement Notes
•
Data sources include patient chart, nurses’ notes, physicians’ orders and discharge summary note. Documentation
quality may affect ability to accurately monitor this performance measure.
•
It may be a challenge to measure compliance and prescribing patterns in primary care.
•
Some patients may be on anticoagulants and would therefore be considered exclusions to these measures. See
Canadian Stroke Strategy Performance Measurement Manual for additional measures on all antithrombotic prescribing
(www.canadianstrokestrategy.ca).
•
Reasons potentially eligible patients are not prescribed antiplatelet agents should be included in data collection. This
information may contribute to the interpretation of the findings of the performance measures and guide quality
improvement initiatives.
Summary of the Evidence
LINK to Evidence Tables for Antiplatelet Therapy in Stroke and TIA Patients
Substantial evidence from randomized trials and meta-analyses supports the use of antithrombotic agents in patients who have experienced an
ischemic stroke. The most commonly recommended antiplatelet agents for secondary stroke prevention in North America and Europe are
acetylsalicylic acid (ASA) , clopidogrel and the combination of ASA and extended-release dipyridamole.95-97 Although some controversy
regarding ASA dosage still exists, most guidelines recommend medium dose ASA (75 to 325 mg/day) as the first choice in secondary
prevention of stroke. Other antiplatelet agents are acceptable alternatives. For patients with a stroke due to a cardioembolic source (e.g., atrial
fibrillation, mechanical heart valve), warfarin is generally recommended (see recommendation 2.6, “Antithrombotic therapy in atrial fibrillation”)
unless contraindicated. Warfarin is not recommended for secondary stroke prevention in patients presumed to have a non-cardioembolic stroke
or transient ischemic attack.
Systematic reviews
In a critical review by O’Donnell and colleagues, immediate and long-term ASA therapy was found to reduce the risk of recurrent stroke,
myocardial infarction and vascular-related death for patients with ischemic stroke or transient ischemic attack.98 Oral anticoagulation was not
more effective than ASA. In comparison to ASA, long-term clopidogrel reduces the relative risk of stroke, myocardial infarction or vascular
death by approximately nine percent. Any benefit of combination antiplatelet therapy with clopidogrel and ASA appears to be offset by an
increased incidence of major bleeding complications compared with either agent alone. The combination of ASA and extended- release
dipyridamole appeared to reduce the relative odds of stroke, myocardial infarction or vascular death by about 18 percent (OR 0.82, 95% CI
0.74–0.91) compared with ASA alone, without causing more bleeding.140
Verro and associates recently published a review of randomized controlled trials comparing ASA plus dipyridamole with ASA alone in patients
with stroke and transient ischemic attack to determine the efficacy of these agents in preventing recurrent vascular events.99 Separate analyses
of the incidence of stroke alone and the composite outcome of stroke, myocardial infarction or vascular death were performed, as well as two a
priori subset analyses examining effect size based on trials using (1) exclusively immediate-release and (2) predominantly extended-release
dipyridamole. Results indicated a significant reduction in the overall risk ratio in favour of ASA plus dipyridamole for stroke (RR 0.77, 95% CI
0.67–0.89) and for the composite end point (RR 0.85, 95% CI 0.76–0.94). Studies using immediate-release dipyridamole showed a nonstatistically significant trend in favour of the combination for stroke (RR 0.83, 95% CI 0.59–1.15) and for the composite outcome (RR 0.95, 95%
CI 0.75–1.19). Studies using predominantly extended-release dipyridamole showed a statistically significant difference in favour of the
combination for stroke (RR 0.76, 95% CI 0.65–0.89) and for the composite outcome (RR 0.82, 95% CI 0.73–0.92). These findings indicate that
ASA in combination with dipyridamole was more effective than ASA alone in preventing recurrent stroke in patients with minor stroke or
transient ischemic attack.99 The risk reduction was greater and statistically significant for studies using primarily extended-release
dipyridamole, which may be a reflection of a true pharmacologic effect or lack of statistical power in studies using immediate-release
dipyridamole.
A 2007 review surveyed the clinical trials and guidelines concerning the use of antiplatelet therapy in the prevention of recurrent stroke after
transient ischemic attack or ischemic stroke of arterial origin.100 Meta-analyses of the results from the randomized controlled trials
demonstrated that, compared with control, the relative risk reduction for recurrent stroke and other serious vascular events was 13 percent with
ASA, 13 percent with dipyridamole (95% CI 4% to 21%; p = 0.046) and 34 percent with combination ASA and dipyridamole. Compared with
ASA, the relative risk of recurrent stroke and other serious vascular events was reduced by 7.3 percent with clopidogrel (95% CI –5.7% to
18.7%) and 18 percent with combination ASA and dipyridamole (9% to 26%, p = 0.0003). Long-term treatment with the combination of ASA
and clopidogrel was not significantly more effective in preventing serious vascular events than clopidogrel alone, mainly due to an increased
frequency of bleeding complications among patients receiving both agents.
An updated Cochrane systematic review assessed the efficacy and safety of dipyridamole relative to control in the secondary prevention of
vascular events in patients with vascular disease.101 The review included randomized long-term secondary prevention trials with concealed
Fourth Edition FINAL
Update: September 2012
Page 27 of 69
CBPRSC 2012
Section 2: Prevention
2.5 Antiplatelet Therapy
treatment allocation, treatment for more than one month, starting within six months after presentation of an arterial vascular disease. Treatment
consisted of dipyridamole with or without other antiplatelet drugs compared with no drug or an antiplatelet drug other than dipyridamole.
Twenty-seven trials were included, with 20 242 patients, among whom 1399 vascular deaths and 3090 fatal and nonfatal vascular events
occurred during follow-up. Compared with control, dipyridamole had no clear effect on vascular death (RR 1.02, 95% CI 0.90–1.17). This result
was not influenced by the dose of dipyridamole or type of presenting vascular disease. In the presence of ASA, dipyridamole appeared to
reduce the risk of vascular events compared with control (RR 0.90, 95% CI 0.82–0.97), due to a single large trial (Second European Stroke
Prevention Study [ESPS2]) in patients presenting with cerebral ischemia.144 The authors concluded that for patients who presented with arterial
vascular disease, there was no evidence that dipyridamole, in the presence or absence of another antiplatelet drug, reduced the risk of
vascular death, though it may reduce the risk of further vascular events. However, this benefit was found in only one large trial and only in
patients presenting after cerebral ischemia. There was no evidence that dipyridamole alone was more efficacious than ASA.
The Antithrombotic Trialists’ Collaboration produced a meta-analysis of randomized controlled trials for antiplatelet therapy in high risk
patients.96 The findings indicated that ASA and other forms of antiplatelet drugs reduced the incidence of nonfatal stroke by one-quarter.
Absolute reduction in the rates of having a serious vascular event were 36 (standard deviation [SD] 6) per 1000 patients treated for two years
among those patients with previous stroke or transient ischemic attack. The authors concluded that the benefits of ASA and other antiplatelet
drugs substantially outweigh the absolute risks of major extracranial bleeding.
In a meta-analysis, Hankey and coworkers assessed the effectiveness and safety of thienopyridine derivatives (ticlopidine and clopidogrel)
compared with ASA for the prevention of serious vascular events in high-risk patients.103 Four high-quality and comparable trials, including 22
656 patients at high risk for adverse vascular events, were identified (three compared ASA to ticlopidine and one compared ASA to
clopidogrel). The use of a thienopyridine was associated with a marginally significant reduction in the odds of serious vascular event (12.0%
v.13%; OR 0.91, 95% CI 0.84–0.98; p = 0.01). There was also a reduction in stroke events in favour of thienopyridines compared with ASA
(5.7% v. 6.4%; OR 0.88, 95% CI 0.79–0.98) corresponding to an avoidance of 7 (95% CI 1–13) stroke events per 1000 patients treated for two
years. In a subgroup analysis of patients with ischemic stroke or transient ischemic attack, the results were similar to those of all patients
combined; however, thienopyridine allocation was associated with a larger absolute reduction in stroke (10.4% v. 12.0%; OR 0.86, 95% CI
0.75–0.97) with an avoidance of 16 (95% CI 3–28) stroke events per 1000 patients treated for two years.103
A review by Halkes and colleagues studied five trials of dipyridamole plus ASA versus ASA alone for secondary prevention of stroke or TIA,
and included the ESPRIT results.104 A total of 7612 patients A total of 7612 patients were included in the analyses (five trials). The trialadjusted hazard ration (HR) for composite event of vascular death, non-fatal myocardial infarction and non-fatal stroke was 0.82 (95%
confidence interval, 0.72 to 0.92). Hazard ratios did not differ in subgroup analyses based on age, sex, qualifying event, hypertension, diabetes,
previous stroke, ischemic heart disease, ASA dose, type of vessel disease and dipyridamole formulation, nor across baseline risk strata as
assessed with two different risk scores. Dipyridamole plus ASA was more effective than ASA alone in preventing recurrent stroke; HR 0.78
(95% CI 0.68 to 0.90). The dose of ASA was fixed in four trials: 25 mg twice daily, 300 mg three times daily, 325 mg three times daily or 330mg
three times daily. IN ESPRIT, the dose of ASA was left to the discretion of the treating physician, provided it was between 30 and 325 mg daily.
The investigators concluded the combination of ASA and dipyridamole is more effective that ASA alone in patients with TIA or ischemic stroke
of presumed arterial origin in the secondary prevention of stroke and other vascular events. The superiority was found in all subgroups and was
independent of baseline risk.
Clinical trials
The most recent trial, SPS3, examined the effects of antiplatelet therapy on lacunar strokes that occur as a result of cerebral small vessel
disease.110 This double blind randomized trial compared daily clopidogrel to placebo, with both groups also receiving 325 mg of aspirin daily.
The results demonstrated that the risk of recurrent stroke was not significantly reduced with aspirin and clopidogrel (dual antiplatelet therapy)
(125 strokes; rate, 2.5% per year) as compared with aspirin alone (138 strokes, 2.7% per year) (hazard ratio, 0.92; 95% confidence interval
[CI], 0.72 to 1.16), nor was the risk of recurrent ischemic stroke (hazard ratio, 0.82; 95% CI, 0.63 to 1.09) or disabling or fatal stroke (hazard
ratio, 1.06; 95% CI, 0.69 to 1.64). However, there was a significant difference in the number of major hemorrhagic events, where the rate was
almost doubled with dual antiplatelet therapy (105 hemorrhages, 2.1% per year) as compared with aspirin alone (56, 1.1% per year) (hazard
ratio, 1.97; 95% CI, 1.41 to 2.71; P<0.001).
The Prevention Regimen For Effectively avoiding Second Stroke (PRoFESS) trial, randomized, double-blind study, investigated the effects of
ASA plus extended-release dipyridamole versus clopidogrel on the prevention of vascular events in patients who had a transient ischemic
attack or ischemic stroke within the preceding 120 days.105 Patients participating in the trial (n = 20 332 across 35 countries) were followed for a
period of four years. Stroke recurrence rates were similar in both arms of the trial (9.0 percent among patients assigned to receive ASA plus
extended-release dipyridamole and 8.8 percent among patients assigned to receive clopidogrel; HR 1.01, 95% CI 0.92–1.11). Nor was there a
significant difference in the composite outcome of stroke, myocardial infarction or vascular death. The trial did not meet its primary end point of
noninferiority for ASA plus extended-release dipyridamole versus clopidogrel.
The European/Australian Stroke Prevention Reversible Ischemia Trial (ESPRIT) group conducted a randomized controlled trial in which
patients were assigned to ASA (30–325 mg daily) with (n = 1363) or without (n = 1376) dipyridamole (200 mg twice daily) within six months of a
transient ischemic attack or minor stroke of presumed arterial origin.106 The primary outcome was the composite of death from all vascular
Fourth Edition FINAL
Update: September 2012
Page 28 of 69
CBPRSC 2012
Section 2: Prevention
2.5 Antiplatelet Therapy
causes, nonfatal stroke, nonfatal myocardial infarction or major bleeding complication, whichever happened first. Treatment was open, but
auditing of outcome events was blinded. Primary analysis was by intention to treat. Mean follow-up was 3.5 years (SD 2.0). Median ASA dose
was 75 mg in both treatment groups (range 30–325); extended-release dipyridamole was used by 83 percent (n = 1131) of the patients on the
combination regimen. Primary outcome events arose in 173 (13%) patients on ASA and dipyridamole and in 216 (16%) on ASA alone (HR
0.80, 95% CI 0.66–0.98; absolute risk reduction 1.0% per year, 95% CI 0.1%–1.8%). Addition of the ESPRIT data to the meta-analysis of
previous trials resulted in an overall risk ratio for the composite of vascular death, stroke or myocardial infarction of 0.82 (95% CI 0.74–0.91).101
Patients on ASA and dipyridamole discontinued trial medication more often than those on ASA alone (470 v. 184), mainly because of
headache. Expressed differently, ESPRIT showed that 104 patients would need to be treated with the combination regimen for 1 year to
prevent 1 additional vascular death, nonfatal stroke or nonfatal myocardial infarction.
The Clopidogrel for High Atherothrombotic Risk and Ischemic Stabilization, Management, and Avoidance (CHARISMA) trial randomly assigned
15 603 patients with clinically evident cardiovascular disease or multiple risk factors to receive clopidogrel (75 mg/day) plus low-dose ASA (75
to 162 mg/day) or placebo plus low-dose ASA and followed them for a median of 28 months.107 The primary efficacy end point, a composite of
nonfatal stroke, nonfatal myocardial infarction or vascular death, was reached by 6.8 percent of patients assigned to receive clopidogrel plus
ASA and 7.3 percent of those assigned to receive placebo plus ASA (RR 0.93, 95% CI 0.83–1.05; p = 0.22). The respective rate of the principal
secondary efficacy end point, which included hospitalizations for ischemic events, was 16.7 percent and 17.9 percent (RR 0.92. 95% CI 0.86–
0.995; p = 0.04). Severe bleeding occurred in 1.7 percent of patients assigned to receive clopidogrel plus ASA and 1.3% percent of those
assigned to receive placebo plus ASA (RR 1.25, 95% CI 0.97–1.61; p = 0.09).107 Among patients with multiple risk factors, the primary end
point was reached by 6.6% of the clopidogrel plus ASA group and 5.5 percent of the placebo plus ASA group (RR 1.2, 95% CI 0.91–1.59; p =
0.20). Death from cardiovascular causes occurred in 3.9% of patients assigned to receive clopidogrel plus ASA and 2.2 percent of those
assigned to receive placebo plus ASA (p = 0.01). In the subgroup with clinically evident atherothrombosis, a marginally significant reduction in
the primary end point of 6.9 percent with clopidogrel and 7.9 percent with placebo was indicated (RR 0.88, 95% CI 0.77–0.998; p = 0.046). The
investigators concluded that there was a suggestion of benefit with clopidogrel treatment in patients with symptomatic atherothrombosis and a
suggestion of harm in patients with multiple risk factors; however, overall, clopidogrel plus ASA was not significantly more effective than ASA
alone in reducing the rate of myocardial infarction, stroke or vascular death.
The Management of Atherothrombosis with Clopidogrel in High-risk patients with recent TIA or ischemic stroke (MATCH) trial was a
randomized, double-blind, placebo-controlled comparison of ASA (75 mg/day) with placebo in 7599 high-risk patients with recent ischemic
stroke or transient ischemic attack and at least 1 additional vascular risk factor who were already receiving clopidogrel 75 mg/day.108 Duration
of treatment and follow-up was 18 months. The primary end point was a composite of ischemic stroke, myocardial infarction, vascular death or
rehospitalization for acute ischemia (including rehospitalization for transient ischemic attack, angina pectoris or worsening of peripheral arterial
disease). The primary end point was reached by 596 (15.7%) of the patients assigned to receive ASA and clopidogrel, and 636 (16.7%) of the
patients assigned to receive placebo plus clopidogrel (relative risk reduction 6.4%, 95% CI –4.6% to 16.3%; absolute risk reduction 1%, 95% CI
–0.6% to 2.7%). Life-threatening bleeding was higher in the group assigned to receive ASA and clopidogrel (2.6%) than in the group assigned
to receive placebo plus clopidogrel (1.3%) (absolute risk increase 1.3%, 95% CI 0.6% to 1.9%). Major bleeding was also increased in the group
assigned to receive ASA and clopidogrel. There was no difference in mortality between the 2 groups. The investigators concluded that adding
ASA to clopidogrel in high-risk patients with recent ischemic stroke or transient ischemic attack was associated with a nonsignificant reduction
in major vascular events and an increase in the risk of life-threatening or major bleeding after 18 months of follow-up.
The Clopidogrel versus ASA in Patients at Risk of Ischemic Events (CAPRIE) trial randomized 19 185 symptomatic patients (one-third had
experienced a previous stroke, one-third had a previous myocardial infarction, and one-third had peripheral vascular disease) to clopidogrel (75
mg) or ASA (325 mg).151 An 8.7 percent (95% CI 0.3%–16.5%; p = 0.043) reduction in the primary end point of ischemic stroke, myocardial
infarction or vascular death in favour of clopidogrel was reported. Among the patients whose qualifying event was a stroke, the number needed
to treat with clopidogrel instead of ASA to prevent a recurrent ischemic event was about 180 per year.109
Pediatrics
ASA is frequently used in children for the secondary prevention of recurrent stroke following a transient ischemic attack or stroke event.111 In
adults, it has been demonstrated that treatment with ASA can reduce the risk of recurrent stroke. Data on the efficacy and optimal dosage of
ASA for paediatric stroke patients are not yet available, but it is clear that no treatment is associated with increased risk of recurrent stroke.112
ASA use has been recommended as a reasonable option for secondary prevention of arterial ischemic stroke for children not at high risk of
recurrent embolism or a hypercoaguable disorder.113
Clopidogrel has been studied as an alternative to ASA in children when ASA is not tolerated or failed in children with arterial ischemic stroke.88,
114,115 Seventeen children were included in the study and started on clopidogrel at 1 mg/kg per day up to a maximum of 75 mg/day.88, Of these,
eight were on clopidogrel alone and nine in combination with ASA. Two patients developed significant intracranial hemorrhage while on the
combination of clopidogrel and ASA – one has recent surgery and the other had hypertension prior to the start of therapy, as well as marked
cerebral atrophy. This small initial investigation concluded that clopidogrel appears to be a reasonable option in children who cannot tolerate
ASA, and the combination of clopidogrel and ASA should be used with caution.
Studies have examined the use of Clopidogrel in children with heart disease for platelet inhibition and reported it safe and efficacious at similar
doses reported by Soman et al for stroke patients. In one study of Clopidogrel in 46 children with heart disease, the mean age of first
Fourth Edition FINAL
Update: September 2012
Page 29 of 69
CBPRSC 2012
Section 2: Prevention
2.5 Antiplatelet Therapy
clopidogrel dose was 4.9 +/- 4.1 years.115 The study dosage ranged from 0.1 to 0.7 mg/kg/day clopidogrel. Almost all patients received
concomitant ASA therapy. The primary outcome was thrombolytic events, and after treatment no thrombotic events developed. Hematological
abnormalities occurred in one child after one year of therapy and these reversed after cessation of therapy. Similarly, Li and colleagues (2008)
found that clopidogrel 0.20 mg · kg−1 · d−1 in children 0 to 24 months of age achieved a platelet inhibition level similar to that in adults taking 75
mg/d. 116 These studies both concluded Clopidogrel appeared safe and efficacious in children, but stressed the need for randomized controlled
research trials to establish dosing and safety in children.
Fourth Edition FINAL
Update: September 2012
Page 30 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Best Practice Recommendation 2.6
Antithrombotic Therapy in Patients
with Atrial Fibrillation
* Significant revisions for 2012
Atrial fibrillation is a significant risk factor for stroke and should be aggressively managed to reduce the risk of cerebrovascular
events.
2.6.1
Primary prevention of stroke in patients with non-valvular atrial fibrillation:
i.
Patients with atrial fibrillation or atrial flutter (paroxysmal, persistent or permanent) should be stratified using a predictive
index for stroke risk (e.g. CHADS2) and for the risk of bleeding, and most patients should receive either an oral
anticoagulant or ASA [Evidence Level A].117
a.
As part of the risk stratification, patients should be assessed for additional risk factors for stroke, including age
65-74yr, female sex, and presence of vascular disease. 117
ii.
Patients with atrial fibrillation at low risk of stroke (CHADS2 = 0) should receive ASA (81-325 mg/day) [Evidence Level
A].118
iii.
Patients with atrial fibrillation at intermediate risk of stroke (CHADS2 =1] should receive oral anticoagulation therapy.
The choice of OAC should be based on patient factors including age, renal function, additional health factors, likelihood
of compliance, patient preferences, and costs.
a.
Most patients should receive dabigatran, rivaroxaban or apixaban^ (^pending approval for use in Canada)
[Evidence Level B] or warfarin [Evidence Level A]. Refer to Anticoagulant Medication Profile table 2.6 to guide
choice of therapy.
b.
Acetylsalicylic acid is a reasonable alternative for some lower risk patients, depending on their individual
risk/benefit profile (age <65 and absence of vascular disease) [Evidence Level A]. (link to CCS table with
CHADS scores and treatment recommendations)
iv. Patients with atrial fibrillation at high risk of stroke (CHADS2 ≥ 2) should receive oral anticoagulation therapy. The
choice of OAC should be based on patient factors including age, renal function, additional health factors, likelihood of
compliance, and patient preferences.
a.
v.
Most patients should receive dabigatran, rivaroxaban or apixaban^ (^pending approval for use in Canada)
[Evidence Level A] or warfarin [Evidence Level A]. Refer to Medication Profile table 2.6 to guide choice of
therapy.
Patients with atrial fibrillation who are already well-controlled on warfarin with a stable INR (with a documented
therapeutic INR greater than 70% of the time) may continue on warfarin and may not need to switch to dabigatran, or
rivaroxiban, or apixaban [Evidence Level C].
Note, in ROCKET, warfarin patients were in therapeutic range an average of 55% of the time, IQR 43 to 71%;
RELY therapeutic range was 64% of the time; and in ARISTOTLE 66% of the time.
vi. Patients with non-valvular atrial fibrillation who are treated with warfarin should have a target INR of 2.5 (maintained in
the range of 2.0 to 3.0); for patients with atrial fibrillation and mechanical heart valves, the target INR is 3.0 (range 2.5 to
3.5) [Evidence Level A].
vii. The combination of ASA and clopidogrel is not routinely recommended for the treatment of atrial fibrillation and should
be reserved for patients in whom treatment with warfarin or the newer oral anticoagulants is not feasible [Evidence Level
A].
viii. There is presently no evidence to recommend dabigatran, rivaroxaban, apixaban^ (^pending approval for use in
Canada)outside the setting of atrial fibrillation for stroke prevention [Evidence Level C].
2.6.2
i.
ii.
Prevention of recurrent stroke in patients with non-valvular atrial fibrillation
Patients with transient ischemic attack and atrial fibrillation should begin oral anticoagulation* with dabigatran, or
rivaroxiban, or apixaban (pending approval for use in Canada)* , or warfarin immediately after brain imaging has
excluded intracranial hemorrhage or large infarct [Evidence Level B].
Most patients with acute ischemic stroke and atrial fibrillation should receive oral anticoagulant therapy with dabigatran,
or rivaroxiban, or apixiban (pending approval for use in Canada) [Evidence Level A], or warfarin [Evidence Level A].
Fourth Edition FINAL
Update: September 2012
Page 31 of 69
CBPRSC 2012
iii.
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Therapy should be started as soon as it is thought to be safe for the patient (see “Clinical Considerations” box for
common treatment protocols and expert suggestions)
For stroke prevention, the new oral anticoagulant agents (dabigatran, rivaroxaban, and apixaban) have been shown to
have advantages in efficacy or safety over warfarin; however, patient specific criteria should be considered when
prescribing medication (refer to Table 2.6 for drug efficacy and toxicity descriptions). (Evidence level A).
iv. For patients with acute ischemic stroke and atrial fibrillation, routine use of bridging with heparin or heparinoid
anticoagulation is not recommended [Evidence Level B]. Most physicians would use ASA until the patient is
anticoagulated [Evidence Level C].
2.6.3
Enhancing anticoagulation therapy and minimizing bleeding complications
i.
Patients prescribed any oral anticoagulation* for atrial fibrillation should be educated about the diagnosis of atrial
fibrillation, the risk of stroke with atrial fibrillation, the importance of medication adherence, and compliance with
international normalized ratio monitoring, if required [Evidence Level B].
ii.
For patients with atrial fibrillation that are taking warfarin, careful dosing and consistent international normalized ratio
monitoring is recommended to minimize adverse events; warfarin efficacy is dependent on maintaining therapeutic
international normalized ratio control, and declines significantly when the international normalized ratio falls below 2.0
[Evidence Level A].
iii. Concomitant antiplatelet therapy with oral anticoagulation* is not recommended in patients with atrial fibrillation unless
there is a specific medical indication such as a coronary stent [Evidence Level B].
iv. With the exception of patients with mechanical heart valves, the addition of acetylsalicylic acid to warfarin in patients with
atrial fibrillation has not been shown to be of benefit in stroke prevention [Evidence Level B]
v. For patients prescribed dabigatran, rivaroxaban, or apixaban, renal function should be routinely monitored, and measured
at least once annually [Evidence Level C].
Clinical Considerations
o Most patients presenting with stroke and TIA will have a CHADS2 score indicating the need for anticoagulation.
o
The third-party funding policies for the new oral anticoagulants (OACs) may not completely align with the Canadian
Best Practice Recommendations for Stroke Care . Individual clinical factors and prescribing rationale should be
clearly documented and included when physicians are advocating for coverage of these medications for their
patients.
o
Based on clinical trial data, warfarin failure was defined as an INR out of therapeutic range more than 30% of time
(i.e., stability is defined as INR in range more than 70% of time), based on findings in RE-LY, ROCKET and
ARISTOTLE.
o
The decision to start anticoagulant therapy is optimally made during the acute phase of hospitalization.
o
The optimal timing of oral anticoagulation following acute stroke for patients in atrial fibrillation is unclear; it is
common practice to wait two to fourteen days and repeat brain imaging (CT or MRI) to rule out asymptomatic
intracranial hemorrhage before starting warfarin [Evidence Level C]. Physicians will often use the size of the infarct
and other clinical circumstances to help judge timing to initiate anticoagulation. For example, in patients with very
small strokes on imaging, anticoagulation may be initiated immediately; in patients with large strokes, initiation of
anticoagulation may be deferred for several weeks.
o
The RE-LY trial of dabigatran did not enroll patients within the first 14 days after stroke, or patients with severe
stroke within the previous six months.156 (7 days- Aristotle).
o
For some patients with acute ischemic stroke and atrial fibrillation, the individual's preferences, level of disability,
prognosis, and overall clinical status, including the size of the infarct on neuroimaging, may contraindicate oral
anticoagulant therapy [Evidence Level C].
o
Patient compliance plays a significant role in the effectiveness of traditional medications (e.g., warfarin) for atrial
fibrillation. Given the shorter half-lives of the new classes of antithrombotics, compliance with these therapies are
also very important to monitor.
o
The CHA2DS2-VASc scoring system is an emerging addition that can be considered for use particularly in patients
Fourth Edition FINAL
Update: September 2012
Page 32 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
with intermediate risk for stroke. The CHA2DS2-VASc has some advantages in that it includes additional clinical data
considered to be more relevant to stroke. In the CHA2DS2-VASc, age ≥ 75 years counts as 2 points instead of one
as in CHADS2, and an additional age category of 65 to 74 years with a score of 1 point has been included. The
presence of vascular disease (peripheral artery disease, MI, aortic plaque) and gender (i.e., female) are also
included in the expanded CHA2DS2-VASc. These additional factors may further help to define the need for
anticoagulation and better prediction of risk of first hositalization.119
Rationale
Atrial fibrillation is a significant risk factor for stroke, with one in six patients with ischemic stroke found to have atrial fibrillation.
Stroke caused by atrial fibrillation is highly preventable if patients are treated with anticoagulants.
System Implications
•
Stroke prevention clinics to improve secondary stroke prevention including management of atrial fibrillation in patients with
stroke and transient ischemic attack (effective, consistent prevention with early recognition of risk factors and timely,
targeted interventions).
•
A process for appropriate outpatient monitoring of patients’ international normalized ratio and follow-up communication with
patients taking anticoagulants.
•
Optimization of strategies at the local, regional and provincial levels to prevent the recurrence of stroke.
•
Stroke prevention awareness and education about secondary prevention for primary care practitioners and specialists who
manage stroke patients during the acute phase and after discharge from acute care.
•
For patients taking warfarin, access to a dedicated anticoagulant management clinic is associated with better patient
outcomes compared to routine medical care.
Performance Measures
1. Proportion of acute ischemic stroke patients with atrial fibrillation who are treated with anti-coagulant therapy
unless contraindicated (core).
2. Proportion of eligible stroke and transient ischemic attack patients with atrial fibrillation prescribed anticoagulant
therapy on discharge from acute care (core).
3. Proportion of eligible stroke and transient ischemic attack patients with atrial fibrillation prescribed anticoagulant
therapy after a visit to a secondary prevention clinic (core).
4. Proportion of atrial fibrillation patients taking anticoagulant therapy at the time of hospital admission for acute ischemic
stroke or transient ischemic attack.
5. Proportion of atrial fibrillation patients with stroke or transient ischemic attack on antiplatelet therapy and not prescribed
anticoagulant therapy.
6. Proportion of atrial fibrillation patients with stroke or transient ischemic attack continuing on anticoagulant therapy at 3
months, 6 months, and 1 year following initiation of therapy.
7. For atrial fibrillation patients on warfarin, the proportion with an international normalized ratio in the therapeutic range at
three months.
Measurement Notes
•
Performance measure 3: reasons why patients with atrial fibrillation and stroke are not on anticoagulants should be
collected and reported. These may include contraindications, compliance issues and physician prescribing patterns, among
others. This additional information will help to inform the direction for quality improvement initiatives.
•
If there is documentation of atrial fibrillation, the chart should be reviewed for medications prescribed to the patient at the
time of discharge, specifically including warfarin, dabigatran, rivaroxiban, apixiban or heparin. Performance measures
should be stratified to include proportions prescribed each of these medications.
•
Data sources may include discharge summary, history and physical examination, physician’s orders, nurses’ notes from
inpatient chart, stroke prevention clinic documents, and primary care charts.
•
To measure whether the patient’s International Normalized Ratio was in the therapeutic range, laboratory reports or other
reliable documentation are required to verify the International Normalized Ratio levels, and these should be reviewed over
a period of time rather than as one single measure.
•
Providing a prescription does not ensure patient adherence with medication administration. Adherence can be determined
Fourth Edition FINAL
Update: September 2012
Page 33 of 69
CBPRSC 2012
•
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
through patient self-report and through International Normalized Ratio measurements over time.
At this time, adherence to the new oral anticoagulants cannot be objectified in the same way as having INR in therapeutic
range
Implementation Resources and Knowledge Transfer Tools
 Thrombosis Interest Group of Canada (available at www.tigc.org)
 Canadian Cardiovascular Society http://www.ccs.ca/guidelines/cc_library_e.aspx
 Canadian Cardiovascular Society Atrial Fibrillation Pocket Guide 2012
 http://www.ccsguidelineprograms.ca/pocket_card/2012_Afib_PG/AtFib_PG_Flipbook/index.html,
Summary of the Evidence
LINK to Evidence Tables for Management of Atrial Fibrillation and Stroke
Atrial fibrillation is a common arrhythmia. The prevalence of atrial fibrillation in the general population increases with age; about 12 percent of
people aged over 75 years have atrial fibrillation.120
Atrial fibrillation is a major risk factor for stroke. The presence of atrial fibrillation increases the risk of stroke approximately five-fold; the relative
risk is higher still if there is associated valvular heart disease.121 Stroke risk also varies according to the presence of other risk factors and can be
estimated using a score that takes into account the presence of congestive heart failure, hypertension, age, stroke and systemic embolism.118
Atrial fibrillation most often causes stroke via embolism of thrombus from the left atrium. Several clinical trials involving many thousands of
patients with AF have examined the efficacy of various antithrombotic drug regimens. The majority of the trial evidence concerns prevention of
first stroke; the evidence-base for prevention of stroke recurrence in patients with AF is much smaller. Strokes due to atrial fibrillation are
generally more severe than those occurring in patients in sinus rhythm. Consequently, atrial fibrillation strokes are associated with higher casefatality, longer hospitalization, and increased disability.122
In spite of convincing evidence-linking AF to stroke, and the benefits of warfarin therapy in reducing this risk, AF patients are often not optimally
managed. An analysis by Gladstone and colleagues using data from the Registry of the Canadian Stroke Network (RCSN) examined this issue in
a cohort of 597 patients presenting to hospital with an ischemic stroke and previously known atrial fibrillation.123 They found that on admission for
stroke, only 40 percent of patients were on warfarin, 30 percent on antiplatelet therapy, and 29% were not on any antithrombotics. Of those taking
warfarin, three fourths had a subtherapeutic INR (<2.0) at the time of stroke admission. Overall, only 10 percent of patients with acute stroke and
previously known atrial fibrillation were therapeutically anticoagulated (INR >2.0) at admission. Among the high-risk subgroup of stroke patients
with a history of atrial fibrillation and a previous TIA or ischemic stroke (n=323), only 18 percent were taking warfarin with a therapeutic INR at the
time of admission for their recurrent stroke; 39 percent were taking warfarin with a subtherapeutic INR, and 15 percent were on no antithrombotic
therapy. These findings are particularly troublesome given that all subjects selected for inclusion in this study were considered high risk for stroke
according to published criteria (low and moderate risk patients were not included), were living independently, and were considered ideal
candidates for warfarin therapy (patients with known contraindications to warfarin were excluded from the study).
The management of atrial fibrillation has changed significantly since 2010 with the approval of dabigatran (RE-LY trial) and rivaroxiban (ROCKET
AF), and the trials on apixaban (ARISTOTLE trial) in Canada.124,125,126 These represent a new class of anticoagulants that do not require the
same close monitoring and dose titration that is involved in warfarin management. Selection of appropriate anticoagulation depends on many
factors including comorbidities, age, and compliance (refer to Medication Reference Guide provided in Table 2.6.1 within these guidelines).
Dabigatran:
Dabigatran is a thrombin inhibitor with a serum half-life is 12 to 17 hours, and 80% of the given dose is excreted by the kidneys. The RE-LY
(Randomized Evaluation of Long-term anticoagulant therapy) study tested two doses of dabigatran against warfarin in a non-inferiority trial with a
prospective, randomized, open-label (warfarin only), blinded-end-point (PROBE) design.124 Collaborators in 44 countries enrolled 18,1130
patients (mean age 71 years, 64 percent men) who had AF and at least one of: past stroke or TIA (20 percent of participants), left ventricular
ejection fraction less than 40 percent, heart failure symptoms within six months (New York Heart Association class II or higher), age 75 years or
higher, or 65-74 years of age plus diabetes mellitus, hypertension, or coronary heart disease. Exclusion criteria included severe valvular heart
disease (including prosthetic valves), stroke within 14 days or severe stroke within six months, conditions that increase risk for hemorrhage,
creatinine clearance less than 30 mL/min, active liver disease, and pregnancy. Participants were randomly allocated to receive dabigatran, 110
mg (n=6015) or 150 mg (n=6076) twice daily, or warfarin adjusted to an INR of 2.0-3.0 (n=6022), and followed for a median of two years (followup was 99.9 percent complete). The primary outcome was a composite of stroke or systemic embolism. Other outcomes included major
hemorrhage, stroke, and death.
Dabigatran 150 mg twice daily reduced the risk of stroke or systemic embolism more than warfarin or dabigatran 110 mg twice daily. Rates of
major hemorrhage were similar for warfarin and dabigatran 150 mg twice daily; dabigatran 110 mg twice daily was associated with lower rates of
major hemorrhage than warfarin. Specifically, the rates of the primary outcome were 1.69 percent per year in the warfarin group, as compared
with 1.53 percent per year in the group that received 110 mg bid of dabigatran (relative risk with dabigatran, 0.91; 95% confidence interval [CI],
Fourth Edition FINAL
Update: September 2012
Page 34 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
0.74 to 1.11; P<0.001 for noninferiority) and 1.11 percent per year in the group that received 150 mg bid of dabigatran (relative risk, 0.66; 95% CI,
0.53 to 0.82; P<0.001 for superiority). The rate of major bleeding was 3.36 percent per year in the warfarin group, as compared with 2.71 percent
per year in the group receiving 110 mg bid of dabigatran (P = 0.003) and 3.11 percent per year in the group receiving 150 mg bid of dabigatran (P
= 0.31). The rate of hemorrhagic stroke was 0.38 percent per year in the warfarin group, as compared with 0.12 percent per year with 110 mg bid
of dabigatran (P<0.001) and 0.10 percent per year with 150 mg bid of dabigatran (P<0.001). The mortality rate was 4.13 percent per year in the
warfarin group, as compared with 3.75 percent per year with 110 mg bid of dabigatran (P = 0.13) and 3.64 percent per year with 150 mg bid of
dabigatran (P = 0.051). A two-fold increase in major bleeding was seen in both dabigatran treatment arms (as well as in the warfarin arm) when
ASA was administered concomitantly.
The only adverse effect that was significantly more common with dabigatran than with warfarin was dyspepsia. Dyspepsia occurred in 348
patients (5.8 percent) in the warfarin group and in 707 patients (11.8 percent) and 688 patients (11.3 percent) in the 110-mg and 150-mg
dabigatran groups, respectively (p<0.001 for both comparisons). Elevations in the serum aspartate aminotransferase or alanine aminotransferase
of more than three times the upper limit of the normal range did not occur more frequently with dabigatran, at either dose, than with warfarin. The
rate of myocardial infarction was 0.53% per year with warfarin and was higher with dabigatran: 0.72 percent per year in the 110-mg group
(relative risk 1.35; 95% CI 0.98-1.97; p=0.07) and 0.74 percent in the 150-mg group (relative risk 1.38, 95% CI 1.00-1.91; p=0.048). An as yet
unpublished trial is testing dabigatran in acute coronary syndromes. Patients taking dabigatran had more gastrointestinal (GI) bleeding, twice the
likelihood of dyspepsia, and dis- continued therapy almost 50% more often in the first year of therapy (CCS 2012).
Warfarin patients in the RE-LY trial had a therapeutic INR only about 64 percent of the time.124 This is consistent with other clinical trials and
underscores the problems with warfarin therapy.127 To have a stroke rate similar to that of he dabigatran 150 mg twice daily group, patients
assigned to warfarin in RE-LY needed to have a therapeutic INR 80 percent of the time.124 This degree of control is unlikely to be achieved in
clinical trials or clinical practice. 129 Ongoing questions remain about dabigatran.128,130 These include: a) the potential safety and utility of the
lower, 110 mg twice daily, dose in older patients with renal impairment; b) the safety and efficacy of dabigatran in the longer term, beyond the
mean follow-up of two years, which is being evaluated in an ongoing follow-up study of RE-LY patients (NCT00808067); c) the implications, if
any, of there being no antidote to dabigatran; and d) the cost of dabigatran.
Rivaroxiban:
Rivaroxaban is a direct factor Xa inhibitor that has been shown to prevent venous thromboembolism in patients undergoing hip surgery more
effectively than enoxaparin (ROCKET Trial 2011).125 The ROCKET AF trial was a multi- center, randomized, double-blind, double-dummy, eventdriven trial that was conducted at 1178 participating sites in 45 countries. Enrolment included 14,264 patients who were randomly assigned to
receive fixed- dose rivaroxiban (20 mg daily or 15 mg daily in patients with a creatinine clearance of 30 to 49 ml per minute) or adjusted-dose
warfarin (target international normalized ratio [INR], 2.0 to 3.0. The primary end point was the composite of stroke (ischemic or hemorrhagic) and
systemic embolism. Stroke or systemic embolism occurred in 188 patients in the rivaroxaban group (1.7% per year) and in 241 patients in the
warfarin group (2.2% per year) (hazard ratio in the rivaroxaban group, 0.79; 95% confidence interval [CI], 0.66 to 0.96; P<0.001 for noninferiority).
Major and clinically relevant nonmajor bleeding occurred in 1475 patients in the rivaroxaban group and in 1449 patients in the warfarin group
(14.9% and 14.5% per year, respectively; hazard ratio in the rivaroxaban group, 1.03; 95% CI, 0.96 to 1.11; P = 0.44). Rates of intracranial
hemorrhage were significantly lower in the rivaroxaban group than in the warfarin group (0.5% vs. 0.7% per year; hazard ratio, 0.67; 95% CI, 0.47
to 0.93; P = 0.02). Major bleeding from a gastrointestinal site was more common in the rivaroxaban group, with 224 bleeding events (3.2%), as
compared with 154 events in the warfarin group (2.2%, P<0.001). The results of the ROCKET AF trial demonstrated that rivaroxaban was
noninferior to warfarin. In the primary safety analysis, there was no significant difference between rivaroxaban and warfarin with respect to rates
of major or non major clinically relevant bleeding. Adverse events occurred in 81.4% of rivaroxaban subjects vs. 83.1% taking warfarin, with only
epistaxis and hematuria significantly more common with rivaroxiban.125
Apixaban: Apixaban is also a direct Factor Xa inhibitor with a half-life of 12 hours.126 The ARISTOTLE trial consisted of a double-blind, doubledummy design, where 18,201 patients were randomly assigned to treatment with apixaban or dose-adjusted warfarin.126 The primary objective
was to determine whether apixaban was noninferior to warfarin in reducing the rate of stroke (ischemic or hemorrhagic) or systemic embolism
among patients with atrial fibrillation and at least one other risk factor for stroke. The primary safety outcome was major bleeding, according to the
criteria of the International Society on Thrombosis and Haemostasis (ISTH). The primary outcome of stroke or systemic embolism occurred in
212 patients in the apixaban group (1.27% per year) as compared with 265 patients in the warfarin group (1.60% per year) (hazard ratio in the
apixaban group, 0.79; 95% confidence interval [CI], 0.66 to 0.95; P<0.001 for noninferiority and P=0.01 for superiority). The rate of hemorrhagic
stroke was 49% lower in the apixaban group than in the warfarin group, and the rate of ischemic or uncertain type of stroke was 8% lower in the
apixaban group than in the warfarin group. Fatal or disabling stroke occurred in 84 patients in the apixaban group (0.50% per year) as compared
with 117 patients in the warfarin group (0.71% per year) (hazard ratio, 0.71; 95% CI, 0.54 to 0.94). Major bleeding, as defined according to ISTH
criteria, occurred in 327 patients in the apixaban group (2.13% per year), as compared with 462 patients in the warfarin group (3.09% per year)
(hazard ratio, 0.69; 95% CI, 0.60 to 0.80; P<0.001). Overall, ARISTOTLE trial found that in patients with atrial fibrillation, apixaban was superior
to warfarin in preventing stroke or systemic embolism, caused less bleeding, and resulted in lower mortality.
Apixiban was also compared directly to ASA in atrial fibrillation patients in the reduction of stroke.131 The Apixaban Versus Acetylsalicylic Acid
(ASA) to Prevent Strokes in Atrial Fibrillation Patients Who Have Failed or Are Unsuitable for Vitamin K Antagonist Treatment (AVERROES) trial
enrolled 5599 patients to receive apixaban 5 mg twice daily or ASA at a dose of 81 to 324 mg daily. The primary efficacy outcome was the
occurrence of stroke (ischemic or hemorrhagic) or systemic embolism. This trial was terminated after the first planned interim analysis of efficacy,
Fourth Edition FINAL
Update: September 2012
Page 35 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
which showed that 104 events had occurred. The effective treatment benefit was more than 4 standard deviations in favour of apixaban.
Principal outcome rates (stroke or STE) were 3.7% per year with ASA/clopidogrel vs. 1.6% per year with apixaban (RR vs. ASA/clopidogrel 0.45;
0.32-0.62; P< 0.001). The rates of major bleeding were 1.2% per year with ASA/clopidogrel vs. 1.4% with apixaban (RR 1.13; P < 0.57)
Warfarin:
Warfarin has been well established as an effective medication in reducing the risk of stroke in patients with atrial fibrillation and atrial flutter. To
characterize the efficacy and safety of antithrombotic agents for stroke prevention in patients who have atrial fibrillation a meta-analysis was
conducted of all randomized trials published between 1966 and March 2007, identified by using the Cochrane Stroke Group search strategy.132
The analysis included 29 trials involving 28 044 patients who had non-valvular atrial fibrillation (mean age 71 years; mean follow-up 1.5 years).
Most trials studied warfarin or ASA in varying dosages and intensities, but other anticoagulants (low-molecular-weight heparin, ximelagatran, and
dabigatran) and other antiplatelet agents (clopidogrel, dipyridamole, indobufen, and trifusal) were also tested. There were 3003 participants
assigned to placebo or control groups in 12 trials. The average stroke rate for these untreated participants was 13 percent per year in trials that
were restricted to those who had previous stroke or TIA (secondary prevention trials) and 4.1 percent per year for those in primary prevention
trials.
Compared with control, adjusted-dose warfarin (6 trials, 2,900 participants 20 percent of whom had previous stroke or TIA) and antiplatelet
agents (8 trials, 4,876 participants 29 percent of whom had previous stroke or TIA) reduced stroke by 64 percent (95% CI 49-74%) and 22
percent (95% CI 6-35%), respectively.132 Adjusted-dose warfarin was substantially more efficacious than antiplatelet therapy (relative risk
reduction, 39 percent [95% CI 22-52%]) (12 trials, 12,963 participants 23 percent of whom had previous stroke or TIA). Adjusted dose warfarin
doubled the risk of intracranial and major extracranial hemorrhage; however, absolute rates of these adverse events were only 0.2 percent per
year.
The largest trial comparing adjusted-dose warfarin with antiplatelet therapy was ACTIVE-W (Atrial fibrillation Clopidogrel Trial with Irbesartan for
prevention of Vascular Events), which included 6706 participants.133 Anticoagulant therapy was superior to the combination of clopidogrel plus
ASA (relative risk reduction, 40 percent [95% CI 18-56%]). The ACTIVE-A trial, published after the meta-analysis by Hart et al, included 7,554
participants in whom warfarin was unsuitable, randomly allocated them to receive clopidogrel plus ASA or ASA alone, and followed them for a
median of 3.6 years.162 The results showed that treating 1000 AF patients for one year with clopidogrel plus ASA prevented eight major vascular
events (including two fatal and three disabling strokes) and caused seven major hemorrhages (one fatal).
Published after the meta-analysis by Hart et al was the BAFTA trial (the Birmingham Atrial Fibrillation Treatment of the Aged study) which
recruited 973 patients (12.5 percent of whom had previous stroke or TIA) aged 75 years or more (mean 81.5 years) from primary care, randomly
assigned them to adjusted-dose warfarin (INR 2.0 - 3.0) or ASA 75 mg once daily, and followed them for a mean of 2.7 years.135 The primary
endpoint was fatal or disabling stroke (ischemic or hemorrhagic), other intracranial hemorrhage, or clinically significant systemic embolism. There
were 24 primary events (21 strokes, two other intracranial hemorrhages, and one systemic embolus) among participants assigned warfarin and
48 primary events (44 strokes, one other intracranial hemorrhage, and three systemic emboli) among those assigned ASA (annual risk 1.8% vs.
3.8%, relative risk reduction 52 percent [95% CI 20-72%], p=0.003; treat 50 patients for one year to prevent one event). The annual risk of
extracranial hemorrhage was 1.4 percent for patients assigned warfarin and 1.6 percent for those assigned ASA.
Two trials in the meta-analysis by Hart et al included only patients who recently had a stroke or TIA. The European Atrial Fibrillation Trial (EAFT)
involved 455 patients who were within three months of TIA or minor stroke, randomly assigned them to warfarin (INR 2.5 to 4.0) or ASA (300
mg/day), and followed them for a mean of 2.3 years.136 In the Studio Italiano Fibrillazione Atriale (SIFA) trial,916 patients within 15 days of TIA or
minor stroke were randomized to open-label warfarin (INR 2.0 to 3.5) or indobufen (a reversible platelet cyclooxygenase inhibitor, 100 or 200 mg
twice a day), and followed for one year.137 The combined results showed that anticoagulants were significantly more effective than antiplatelet
therapy for the prevention of all ischemic vascular events (odds ratio 0.67, 95% CI 0.50–0.91) and for the prevention of stroke recurrence (odds
ratio 0.49, 95% CI 0.33–0.72). Warfarin did not significantly increase the frequency of intracranial bleeding.138 Although major extracranial
bleeding complications occurred more often in patients on warfarin (odds ratio 5.16, 95% CI 2.08–12.83), the absolute difference was small (2.8
percent per year versus 0.9 percent per year in EAFT and 0.9 percent per year versus zero percent in SIFA). Heparin anticoagulation confers no
net benefit over antiplatelet therapy in patients with AF and recent (within 14 days) acute ischemic stroke.139
A Danish cohort study investigated the risks of bleeding on monotherapy compared to combined therapy in patients with atrial fibrillation.140
Records from nation-wide registries were used to identify patients with a first-time hospitalization for AF between 1997 and 2006. A total of 82
854 of 118 606 patients (69.9 percent) surviving AF hospitalization had at least 1 prescription filled for warfarin, ASA, or clopidogrel after
discharge. During mean (SD) follow-up of 3.3 (2.6) years, 13 573 patients (11.4 percent) experienced a nonfatal or fatal bleeding. The crude
incidence rate for bleeding was highest for dual clopidogrel and warfarin therapy (13.9 percent per patient-year) and triple therapy (15.7 percent
per patient- year). Using warfarin monotherapy as a reference, the hazard ratio (95% confidence interval) for the combined end point was 0.93
(0.88-0.98) for ASA, 1.06 (0.87-1.29) for clopidogrel, 1.66 (1.34-2.04) for ASA-clopidogrel, 1.83 (1.72-1.96) for warfarin-ASA, 3.08 (2.32-3.91) for
warfarin-clopidogrel, and 3.70 (2.89- 4.76) for warfarin-ASA-clopidogrel. Triple therapy posed a bleeding risk, which was three times higher than
the risk of bleeding for warfarin alone. The problems associated with warfarin therapy have stimulated the development of oral agents that
produce more predictable anticoagulation and do not require frequent monitoring.
The Canadian Cardiovascular Society guidelines for the management of atrial fibrillation conclude that compared with warfarin, both dabigatran
and apixaban are more efficacious than warfarin for the prevention of stroke and STE, while rivaroxaban is noninferior to warfarin.117 Apixaban
Fourth Edition FINAL
Update: September 2012
Page 36 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
causes less major bleeding than warfarin, while in comparison with warfarin there is no more major bleeding with either dabigatran 150 mg or
rivaroxaban. There is significantly less intracranial bleeding with each of the new agents than with warfarin” .117 Clinical trials that directly
compare dabigatran, rivaroxiban and apixaban have not been published, and the long term effects of these newer oral anticoagulants has not
been studied due to their recent release. These trials will be required to provide specific guidance on choice of oral anticoagulant. At this time,
physicians should consider patient factors, medical status and risks as well as likelihood of patient compliance in selecting a treatment regime in
the presence of atrial fibrillation.
Figure 2.6: Canadian Cardiovascular Society Algorithm For Stroke Risk and Management Decisions in Patients with
Atrial Fibrillation (Canadian Journal of Cardiology, 2012; 28:125-136).
** (awaiting official permission from published to reproduce this diagram)
Figure 1
Source: Canadian Journal of Cardiology 2012; 28:125-136 (DOI:10.1016/j.cjca.2012.01.021 )
Copyright © 2012 Canadian Cardiovascular Society Terms and Conditions
Fourth Edition FINAL
Update: September 2012
Page 37 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Table 2.6: Oral Anticoagulants for the Prevention of Stroke in Atrial Fibrillation Patients
This table provides a summary of the pharmacotherapeutic properties, side effects, drug interactions and other important information on the four
anticoagulant medications currently in use or under review for use in Canada. This table should be used as a reference guide by health care
professionals when selecting an appropriate agent for individual patients. Patient compliance, side effects, drug interactions and bleeding risk status
should all be taken into consideration during decision-making, in addition to other information provided in this table and available elsewhere
regarding these medications. (Notes: * Apixaban currently not available in Canada. Application has been submitted to Health Canada and is under
review.)
Dabigatran
Mechanism of Action
Rivaroxaban
Apixaban*
Side Effects
Fourth Edition FINAL
Vitamin K antagonism of factors II,
VII, IX, X
Direct thrombin inhibitor
Direct Xa inhibitor
Direct Xa inhibitor
-Stroke prophylaxis in nonvalvular atrial fibrillation
-Prevention of VTE in TKA and
THR
-Stroke prophylaxis in nonvalvular atrial fibrillation
-Prevention of VTE in TKA and
THR
-Prevention of VTE in TKA and
THR
-Prophylaxis and/or treatment of
VTE, atrial fibrillation with
embolization, and as an adjunct in
the prophylaxis of systemic
embolism after myocardial
infarction, including stroke and
reinfarction
Active bleeding or significant risk
factors for bleeding
CrCl <30 ml/min (Cockcroft
Gault Equation NOT eGFR)*
Active bleeding or significant risk
factors for bleeding
CrCl <30 ml/min (Cockcroft
Gault Equation NOT eGFR)*
Active bleeding or significant risk
factors for bleeding
CrCl <15m/min (Cockcroft Gault
Equation NOT eGFR)*
Active bleeding or significant risk
factors for bleeding
Pregnancy
Gastritis-like symptoms;
dyspepsia, Bleeding:
150 mg BID – similar bleeding
risk to warfarin
110 mg BID – lower bleeding risk
than warfarin. Lower risk of ICH
than warfarin; Higher risk of GI
bleed vs. warfarin with 150 mg
BID dose
Bleeding: Similar risk to warfarin
overall. Lower risk of ICH vs.
warfarin.
Higher risk of transfusion vs.
warfarin. Higher risk of GI bleed
vs. warfarin
Bleeding: Lower risk of bleeding
vs. warfarin
Bleeding: Purple toe syndrome
(rare)
Indications
Contraindications*
Warfarin
Update: September 2012
Page 38 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Dabigatran
Landmark Trials
Inclusion
Criteria
Exclusion
Criteria
Rivaroxaban
Apixaban*
Warfarin
Multiple RCTs and Meta-Analyses
in both valvular and non-valvular
Atrial Fibrillation
RE-LY
NEJM 2009;361:1139-51
ROCKET-AF
NEJM 2011;365:883-91
ARISTOTLE
NEJM 2011;365:981-92
Documented non-valvular AFib
within 6 mos and at least 1 of:
Previous stroke/TIA
Heart failure
Age > 75
Age >65 + DM
HTN
CAD
Documented non-valvular AFib,
with history of stroke, TIA, or
systemic embolism or at least 2 of
the following:
Heart failure
HTN
Age >75
DM
Documented AFib or AFlutter plus
at least one of:
Previous stroke, TIA, systemic
embolism
Age >75
Heart failure
DM
HTN requiring treatment
Severe heart-valve disorder
Stroke within 14 days
Severe stroke within 6 months
Condition that increased risk of
hemorrhage
CrCl <30ml/min
Active liver disease
Pregnancy
ASA >100 mg/d
Severe heart valve disease
TIA caused by reversible disorder
Active IE
Condition that increased risk of
hemorrhage
Uncontrolled HTN
Stroke within 14 days or severe
stroke within 3 months
Significant liver disease
Use of strong CYP 3A4 inhibitors
Chronic NSAIDs
Pregnancy
HIV
CrCl <30ml/min
ASA >100mg/d
Afib due to reversible cause
Moderate or severe valvular
disease
Stroke in past 7 days
CrCl <25 ml/min
Need for ASA+Plavix or ASA >165
mg/d
LANDMARK TRIAL BLEEDING RISKS
Overall Bleeding
Fourth Edition FINAL
110 mg: 14.6%/yr
150 mg: 16.4%/yr
14.9 per 100 pt-yr
Update: September 2012
18.1%/yr
RE-LY: 18.2%/yr
ROCKET-AF: 14.5 per 100 pt-yr
ARISTOTLE: 25.8%yr
Page 39 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Dabigatran
Rivaroxaban
Apixaban*
Warfarin
Major Bleeding
110 mg: 2.7%/yr
150 mg: 3.1%/yr
5.6 per 100 pt-yr
2.13%/yr
RE-LY: 3.4%/yr
ROCKET-AF: 5.4 per 100 pt-yr
ARISTOTLE: 3.09%/yr
ICH
110 mg: 0.23%/yr
150 mg: 0.30%/yr
0.5 per 100 pt-yr
0.33%/yr
RE-LY: 0.74%/yr
ROCKET-AF: 0.7 per 100 pt-yr
ARISTOTLE: 0.8%/yr
GI Bleed
110 mg: 1.1%/yr
150 mg: 1.5%/yr
3.2%
0.76%/yr
RE-LY: 1%/yr
ROCKET-AF: 2.2%
ARISTOTLE: 0.86%/yr
CYP3A4 and P-glycoprotein (e.g.
anticonvulsants, rifampin,
dexamethasone, trazodone,
amiodarone, cyclosporine,
diltiazem, verapamil, azole
antifungals, macrolides,
efavirenz), other agents that
effect bleeding
CYP3A4 and P-glycoprotein
(e.g. anticonvulsants, rifampin,
dexamethasone, trazodone,
amiodarone, cyclosporine,
diltiazem, verapamil, azole
antifungals, macrolides,
efavirenz), other agents that
effect bleeding
CYP2C9 and CYP3A4 (e.g.
anticonvulsants, rifampin, amiodarone,
azole antifungals, macrolides, efavirenz),
vitamin K containing foods, other agents
that effect bleeding
1-3 hours
3-4 hours
1-3 hours
3-5 days
14-17 hours
7-11 hours
8-15 hours
20-60 hours
6%
>80%
66%
Rapid and extensive
80% renal
66% renal
25% renal
None
Drug Interactions
* Note: This is NOT a
complete list, rather
examples of some of
the more frequent or
serious drug
interactions with
these OACs.
Comments
P-glycoprotein (e.g.
carbamazepine, rifampin,
dexamethasone, trazodone,
amiodarone, cyclosporine,
diltiazem, verapamil,
ketoconazole), other agents
that effect bleeding
Prodrug – dabigatran exetilate
(needs acidic environment for
optimal absorption)
Time to Peak Effect
Half-life
Bioavailability
Excretion
Effect of Food
Delayed absorption
Increases absorption of 20 mg
dose but not 10 mg dose
No effect
Usual Dosing in Atrial
Fibrillation
150mg BID
110mg BID
20mg OD
15mg OD (if CrCl 30-49)
5mg BID
2.5mg BID if ≥2 of: age≥80,
wt≤60kg, SrCr≥133
Fourth Edition FINAL
Update: September 2012
Slows rate but not extent; Vitamin K
content should be kept consistent
Initial: 2.5-10 mg daily
Maintenance based on INR (target 2.5,
range 2-3)
Page 40 of 69
CBPRSC 2012
Section 2: Prevention
2.6 Anticoagulant Therapy for Atrial Fibrillation
Dabigatran
Rivaroxaban
Anticoagulation
Monitoring
Not Required
Not Required
Additional Monitoring
SCr at baseline and at least
annually
SCr at baseline and at least
annually
No
Antidote
Hold for Invasive
Surgery
$ per month/coverage
in Canada
Post-Marketing
Notes/Comments
Apixaban*
Warfarin
Not Required
INR
Prothrombin Complex
Concentrate (preliminary
evidence in healthy volunteers):
Circulation 2011;124:1573–1579
No
Vitamin K and Prothrombin Complex
Concentrate: Chest 2012;141;e152Se184S
At least 24 hours
At least 24 hours
5 days
Not yet covered for AFib in
public drug plans
$3.20/day
Not yet covered for AFib in public
drug plans
~$3/day
Does not yet have AFib NOC
Full benefit
$0.06/day, $1.16/day including monitoring
costs
-Safety reviews in multiple
countries
-Meta-analysis supports
increased risk of MI seen in RELY trial
-Minimal post-marketing
experience in atrial fibrillation
-No post-marketing experience
in atrial fibrillation
1-2 days (if CrCl >50)
3-5 days (if CrCl <50)
Hold for 24 hours prior to
ablation for atrial fibrillation
-Extensive post-marketing experience
Note: * The FDA standard for drug dosing recommendations is the Cockroft-Gault equation
Fourth Edition FINAL
Update: September 2012
Page 41 of 69
CBPRSC 2012
Section 2: Prevention
2.7 Carotid Disease and Intracranial Atherosclerosis
Best Practice Recommendation 2.7
Management of Extracranial Carotid
Disease and Intracranial Atherosclerosis
* Minor revisions for 2012
2.7.1 Symptomatic carotid stenosis
Patients with transient ischemic attack or non-disabling stroke and ipsilateral 50 to 99 percent internal carotid artery stenosis
(measured by two concordant non-invasive imaging modalities such as dopplers, CTA, or MRA) should be evaluated by an
individual with stroke expertise (neurosurgeon/vascular surgeon) and selected patients should be offered carotid endarterectomy
as soon as possible, with the goal of operating within fourteen days of the incident event once the patient is clinically stable
[Evidence Level A].
i.
Carotid endarterectomy should be performed by a surgeon with a known perioperative morbidity and mortality of less
than 6 percent [Evidence Level A].
ii.
Carotid stenting may be considered for patients who are not operative candidates for technical, anatomic or medical
reasons [Evidence Level A]. Interventionalists should have expertise in carotid procedures and an expected risk of
peri-procedural morbidity and mortality rate of less than 5 percent.
iii.
Carotid endarterectomy is more appropriate than carotid stenting for patients over age 70 years who are otherwise fit
for surgery because stenting carries a higher short-term risk of stroke and death [Evidence Level A].
2.7.2 Asymptomatic and Remotely Symptomatic Carotid Stenosis
Carotid endarterectomy may be considered for selected patients with 60 to 99 percent carotid stenosis who are asymptomatic or
were remotely symptomatic (i.e., greater than three months) [Evidence Level A].
i.
Patients with asymptomatic carotid disease should be evaluated by a physician with expertise in stroke
management [Evidence Level A].
ii.
Patients should be less than 75 years old with a life expectancy of more than 5 years, and an acceptable risk of
surgical complications [Evidence Level A].
iii.
Carotid endarterectomy should be performed by a surgeon with a less than 3 percent risk of peri-operative
morbidity and mortality [Evidence Level A].
iv.
The benefits of carotid endarterectomy in women have shown mixed findings. Although the initial (5-year) results of
the ACST showed no benefit in women, ten-year follow-up in the ACST trial suggest long-term benefit for both men
and women [Evidence Level B]. Female sex in isolation is not an exclusion criterion for surgery, but should be
considered as part of the overall risk benefit assessment with specific attention to co-morbid disease and general
health status. [Evidence Level C]. 141,142
v.
Carotid stenting may be considered in patients who are not operative candidates for technical, anatomic or medical
reasons provided there is a less than 3 percent risk of peri-procedural morbidity and mortality [Evidence Level A].
2.7.3 Intracranial Stenosis
i.
Intracranial stenting is not recommended for the treatment of recently symptomatic intracranial 70% to 99% stenosis
[Evidence Level B].
ii.
In the SAMMPRIS trial the medical management arm included dual antiplatelet therapy with ASA 325 mg and
Clopidogrel 75 mg for up to 90 days, as well as aggressive management of all vascular risk factors including blood
pressure, lipids, diabetes mellitus, and other at-risk lifestyle patterns [Evidence Level A].143 It is not clear if all these
components are necessary for medical management. Management decisions should be based upon individual
vascular risk profiles.
iii.
In patients who have been managed with maximal medical therapy in the presence of intracranial stenosis and
experience a recurrent stroke (as per SAMMPRIS), there is lack of clear evidence to guide further management
decisions. Management decisions should be based upon individual vascular risk profiles [Evidence Level C].
Rationale
Carotid endarterectomy is a surgical procedure that removes atherosclerotic plaque from the proximal internal carotid artery.
Successful carotid endarterectomy substantially reduces the risk of recurrent stroke in patients who present with a hemispheric
Fourth Edition FINAL
Update: September 2012
Page 42 of 69
CBPRSC 2012
Section 2: Prevention
2.7 Carotid Disease and Intracranial Atherosclerosis
transient ischemic attack or minor stroke and an ipsilateral high-grade carotid stenosis. One death or severe stroke is prevented
for every nine patients with symptomatic severe (70 to 99 percent) carotid stenosis treated with carotid endarterectomy). For
selected patients with asymptomatic carotid stenosis, carotid endarterectomy reduces the risk of stroke from about two percent
per year to about one percent per year. Aggressive medical management was superior to intracranial stenting for patients with
70 to 99% stenosis of a major intracranial artery.
System Implications
•
Protocols to ensure timely access to diagnostic services for evaluating carotid arteries.
•
Development of agreements and processes for rapid access to surgical consults, including a mechanism for expedited
referrals as required for carotid interventions.
Performance Measures
1. Proportion of stroke patients with moderate to severe (50 percent to 99 percent) carotid artery stenosis who
undergo a carotid intervention procedure following an index stroke event.
2. Median time from stroke symptom onset to carotid endarterectomy surgery (core).
3. Proportion of stroke patients requiring carotid intervention who undergo the procedure within two weeks of the
index stroke event.
4. Proportion of stroke patients with moderate carotid stenosis (50 percent to 69 percent) who undergo carotid intervention
procedure following the incident stroke event.
5. Proportion of stroke patients with mild carotid stenosis (less than 50 percent) who undergo carotid intervention procedure
following the incident stroke event.
6. Proportion of carotid endarterectomy patients who experience perioperative in-hospital stroke, acute myocardial
infarction or death.
7. The 30-day in-hospital mortality rate after carotid endarterectomy and stroke rate by carotid occlusion severity.
8. Proportion of patients who undergo carotid endarterectomy within two weeks, between two and four weeks, between four
weeks and three months, and between three and six months of stroke onset.
9. Proportion of patients who wait more than three months for carotid endarterectomy or whose surgery is cancelled
because of long wait times.
10. Proportion of patients who experience a subsequent stroke event or death while waiting for carotid endarterectomy.
Measurement Notes
•
Time interval measurements should be taken from the time the patient or family reports as the time of stroke symptom
onset to the actual date of surgery.
•
The stroke onset time will depend on patient report or that of a reliable observer at the time of the event.
•
Analysis should be stratified between those patients undergoing carotid stenting and those patients undergoing carotid
endarterectomy, by severity of stenosis and by whether the patient had symptomatic or asymptomatic carotid artery
disease.
•
Data source for surgical date should be surgical note, nurses’ notes and discharge summary.
•
In some cases, it may be more appropriate or relevant to record the time interval from the first time the patient has
contact with medical care until the time of carotid surgery. This has occurred in cases where the patient was out of the
country at the time of the stroke event and chose to return to Canada before seeking definitive medical intervention. It is
important to note the nature of the start time when calculating turnaround times or intervention times.
Implementation Resources and Knowledge Transfer Tools
 NINDS: http://www.ninds.nih.gov/disorders/stroke/carotid_endarterectomy_backgrounder.htm
 National Heart, Lung and Blood Institute: http://www.nhlbi.nih.gov/health/health-topics
/topics/carend/printall-index.html
Summary of the Evidence
LINK to Evidence Tables for Management of Extracranial Carotid Disease and Intracranial Atherosclerosis
It has been well established that carotid endarterectomy is beneficial for stroke prevention in appropriate patients. There are three large trials of
endarterectomy for symptomatic stenosis: the North American Symptomatic Carotid Endarterectomy Trial (NASCET),144 the European Carotid
Surgery Trial (ECST)145 and the Veterans Affairs 309 Trial.146 According to a pooled analysis of these trials, endarterectomy is highly beneficial
in symptomatic patients with severe (70–99 percent) angiographic stenosis (NNT = six to prevent one stroke over five years), moderately
beneficial for symptomatic patients with moderate (50–69%) stenosis (NNT = 22 to prevent one stroke over five years) and not beneficial for
mild (< 50%) stenosis.147 Guidelines on carotid endarterectomy from the American Heart Association and the Canadian Neurosurgical Society
Fourth Edition FINAL
Update: September 2012
Page 43 of 69
CBPRSC 2012
Section 2: Prevention
2.7 Carotid Disease and Intracranial Atherosclerosis
recommend surgery for symptomatic high-grade stenosis (70–99%), but have not been updated to include the most recent evidence regarding
symptomatic patients with moderate stenosis or patients with asymptomatic stenosis. 148,149
The risks of carotid endarterectomy in relation to the timing of surgery was investigated in a systematic review of the literature on the
complications of carotid endarterectomy.150 The operative risk of stroke and death was not increased in neurologically stable patients when
surgery was performed early (< 3 to 6 weeks) rather than late (> 3 to 6 weeks). However, in unstable patients who underwent “urgent”
endarterectomy for “stroke-in-evolution” or “crescendo transient ischemic attacks,” there was an increased perioperative risk (20%) that was
significantly higher than the risk in stable patients.
A recent study by Gladstone, using data from the Registry of the Canadian Stroke Network (RCSN), examined factors associated with the
timing of carotid endarterectomy surgery.151 A cohort of 1011 patients were found to have symptomatic carotid stenosis, and among those, 105
patients with severe (80 percent of cohort) or moderate (29 percent of cohort) stenosis underwent carotid endarterectomy within six months
and were included in the analysis. The median time from index event to surgery was 30 days (interquartile range, 10 to 81). Overall,
approximately one third (38 of 105) underwent surgery within two weeks, half (53 of 105) received surgery within 1 month, and one fourth (26 of
105) had surgery >3 months after the presenting event. In the multivariable analysis, early surgery (within two weeks) was significantly more
likely to occur if the index event was a TIA rather than a completed stroke (OR, 2.6; 95% CI, 1.1 to 6.1). Age, sex, and degree of stenosis were
not found to be significant predictors of early surgery. Over the study timeframe, there was an improvement in the median time to
endarterectomy, decreasing from 74 days in 2003 to 21 days in 2006 (P_0.022 for median regression analysis). The proportion of patients
undergoing early carotid endarterectomy (within 2 weeks) improved significantly over time: 18.2 percent in 2003, 25.0 percent in 2004, 45.5
percent in 2005, and 44.8 percent in 2006 (P_0.036; Cochran-Armitage trend test). Patients who did not undergo surgery were significantly
older with more severe strokes and more comorbidities. The six-month mortality rate was 3.4 percent in the surgical group and 12.9 percent in
the non-surgical group (p=0.0003).
Endarterectomy for symptomatic patients should be performed with a maximum combined perioperative stroke and death rate of six percent,
according to the American Academy of Neurology guidelines152 and the Canadian Neurosurgical Society guidelines;149 the American Heart
Association guidelines148 recommend a five percent rate for patients with transient ischemic attack and seven percent for patients with stroke.
Women appear to have a higher perioperative risk and do not appear to benefit from carotid endarterectomy for symptomatic moderate (50–
69%) stenosis, 153 or when performed after greater than 2 weeks for symptomatic, high-grade (70–99%) stenosis.185All of these guidelines
recommend that endarterectomy for asymptomatic patients be performed with a maximum combined perioperative stroke and death rate of
less than three percent.
For the Carotid Endarterectomy Trialists’ Collaboration, Rothwell and associates analyzed pooled data (5893 patients with 33 000 patient-years
of follow-up) from the European Carotid Surgery Trial and North American Symptomatic Carotid Endarterectomy Trial.154 The findings indicated
that the benefit from endarterectomy depends not only on the degree of carotid stenosis but also on several other clinical characteristics,
including the timing of surgery after the presenting event. In patients with severe stenosis (70–99 percent), surgery was most effective when
performed within two weeks of the index transient ischemic attack or stroke (NNT = three to prevent one stroke in five years), and this benefit
declined quickly over time (NNT = 125 for patients who undergo surgery more than 12 weeks after the symptomatic event). This timedependent decline in benefit was even more pronounced in patients with moderate stenosis (50%–69%); endarterectomy performed within the
first two weeks of the ischemic event was beneficial, but the benefit was lost (and there was net harm) when surgery was delayed more than
three months. Therefore, the Carotid Endarterectomy Trialists’ Collaboration recommended that carotid endarterectomy should be done within
two weeks of the patient’s last symptoms.
Carotid endarterectomy for asymptomatic carotid artery disease has been controversial. The Asymptomatic Carotid Atherosclerosis Study
(ACAS) Group randomized 1662 asymptomatic patients with carotid artery stenosis of 60 percent or greater reduction in diameter to receive
carotid endarterectomy, with daily ASA administration and medical risk factor management for all patients.155 After a median follow-up of 2.7
years, the absolute risk reduction for ipsilateral stroke was 3.0 percent for surgical patients compared with patients treated medically. The MRC
[Medical Research Council] Asymptomatic Carotid Surgery Trial (ACST) Collaborative Group randomized 3120 asymptomatic patients with
substantial carotid narrowing equally between earlier carotid endarterectomy (half received carotid endarterectomy by one month, 88 percent
by one year) and indefinite deferral of any carotid endarterectomy (only four percent per year received carotid endarterectomy) over a 10-year
period.141 Patients were followed for up to five years (mean 3.4 years). The absolute risk reduction for ipsilateral stroke was 3.1 percent.
Subgroup analyses found no significant heterogeneity in the perioperative hazards or (apart from the importance of cholesterol) in the longterm postoperative benefits. These benefits were separately significant for males and females, for those with about 70 , 80 and 90 percent
carotid artery narrowing on ultrasound and for those younger than 65 and 65–74 years of age (though not for older patients, half of whom died
within five years from unrelated causes).
Asymptomatic carotid artery stenosis (unlike symptomatic carotid artery stenosis) is a relatively low-risk condition, and these studies confirm its
natural history, although there is evidence that patients with higher degrees of asymptomatic stenosis are at a higher risk over time.156 Overall,
the absolute risk reduction with carotid endarterectomy is small (3.0 percent), translating into a number needed to treat of about 33. Gladstone
and Sahlas recommended that carotid endarterectomy should be considered only for carefully selected patients with carotid artery stenosis of
at least 60 percent who are less than 75 years old, have a good life expectancy and are at low surgical risk.157 A similar recommendation has
been issued by the American Academy of Neurology.152 They recommended in asymptomatic patients that “it is reasonable to consider carotid
endarterectomy for patients between the ages of 40 and 75 years and with asymptomatic stenosis of 60 to 99 percent if the patient has an
Fourth Edition FINAL
Update: September 2012
Page 44 of 69
CBPRSC 2012
Section 2: Prevention
2.7 Carotid Disease and Intracranial Atherosclerosis
expected 5-year life expectancy and if the surgical stroke or death frequency can be reliably documented to be < 3 percent (Level A).” The
American Stroke Association included a recommendation that “patients with asymptomatic carotid artery stenosis be screened for other
treatable causes of stroke and that intensive therapy of all identified stroke risk factors be pursued (Level of Evidence C).158
Practice gaps in carotid disease management have been identified. According to a Canadian study, the appropriate patients who are most
likely to benefit from endarterectomy are not always being referred, and many procedures are performed inappropriately on patients at low risk
of stroke.159 In an Oxfordshire, United Kingdom, population-based study of transient ischemic attack and stroke patients referred for
endarterectomy for > 50 percent stenosis, only six percent had surgery within two weeks of their ischemic event and only 43 percent within
three months; 32 percent of patients had a recurrent stroke while awaiting endarterectomy.160 Stroke prevention clinics have been found to
have an important role in promoting adherence to guidelines and ensuring appropriate patient selection and timely referral for this procedure.
Delays from presenting event to initial assessment, carotid imaging and endarterectomy are new key indicators that should be monitored as
part of stroke quality assurance programs.
Studies that compared carotid endarterectomy to carotid stenting have recently emerged in the research literature. A Cochrane systematic
review of 10 trials which included 3178 patients with carotid stenosis comparing endovascular procedures to carotid endarterectomy surgery.161
The primary outcome comparison of any stroke or death within 30 days of treatment favoured surgery(fixed-effects OR 1.35), but the difference
was not statistically significant using the random effects model. Endovascular treatment was significantly better than surgery in avoiding cranial
neuropathy (OR 0.15) and myocardial infarction (OR 0.34). There was no significant difference between endovascular treatment and surgery in
the following comparisons: 30-day stroke, MI, or death (OR1.12); 30-day disabling stroke or death (OR 1.19); 30-day death OR 0.99); 24-month
death or stroke (OR 1.26); and 30-day death or stroke in endovascular patients treated with or without protection devices (OR 0.75). The
authors concluded that results do not support a change in clinical practice away from recommending carotid endarterectomy as the treatment
of choice for suitable carotid artery stenosis but support continued recruitment in the large ongoing trials.
A more recent meta-analysis in the British Medical Journal in 2010, which included the results from the International Carotid Stenting Study
(ICSS), evaluated the relative short term safety and intermediate term efficacy of carotid endarterectomy versus carotid artery stenting.162 The
analysis included randomized controlled trials which compared carotid endarterectomy with carotid artery stenting in patients with carotid artery
stenosis with or without symptoms. The primary end point was a composite of mortality or stroke. Secondary end points were death, stroke,
myocardial infarction, or facial neuropathy (as individual end points), and mortality or disabling stroke (as a composite end point). Eleven trials
were included (4796 patients)in the analysis, with 10 that reported on short-term outcomes (n=4709) and nine on intermediate term outcomes
(1-4 years). The peri-procedural risk of mortality or stroke was lower for carotid endarterectomy (odds ratio 0.67, 95% confidence interval 0.47
to 0.95; P=0.025) than for carotid stenting, mainly because of a decreased risk of stroke (0.65, 0.43 to 1.00; P=0.049), whereas the risk of
death (1.14, 0.56 to 2.31; P=0.727) and the composite end point mortality or disabling stroke (0.74, 0.53 to 1.05; P=0.088) did not differ
significantly. The odds of periprocedural myocardial infarction (2.69, 1.06 to 6.79; P=0.036) or cranial nerve injury (10.2, 4.0 to 26.1; P<0.001)
was higher in the carotid endarterectomy group than in the carotid stenting group. In the intermediate term, the two treatments did not differ
significantly for stroke or death (hazard ratio 0.90, 95% confidence interval 0.74 to 1.1; P=0.314). The authors concluded that carotid
endarterectomy was found to be superior to carotid artery stenting for short-term outcomes but the difference was not significant for
intermediate term outcomes; this difference was mainly driven by nondisabling stroke. Significantly fewer cranial nerve injuries and myocardial
infarctions occurred with carotid artery stenting.
Treatment: Carotid Endarterectomy and Stenting
Two randomized trials that directly compared the safety of carotid stenting to carotid endarterectomy in symptomatic patients have been
release this past year: the International Carotid Stenting Study (ICSS)163 and the Carotid Revascularization Endarterectomy versus Stenting
Trial (CREST).164 In the ICSS study, patients with recently symptomatic carotid artery stenosis were randomly assigned in a 1:1 ratio to receive
carotid artery stenting or carotid endarterectomy.163 The primary outcome measure of the trial was the three-year rate of fatal or disabling
stroke in any territory, which has not been analyzed yet. The main outcome measure for the interim safety analysis was the 120-day rate of
stroke, death, or procedural myocardial infarction.
The trial enrolled 1713 patients (stenting group, n=855; endarterectomy group, n=858). Between randomization and 120 days, there were 34
(Kaplan-Meier estimate 4·0%) events of disabling stroke or death in the stenting group compared with 27 (3·2%) events in the endarterectomy
group (hazard ratio [HR] 1·28, 95% CI 0·77—2·11). The incidence of stroke, death, or procedural myocardial infarction was 8·5 percent in the
stenting group compared with 5·2 percent in the endarterectomy group (72 vs. 44 events; HR 1·69, 1·16—2·45, p=0·006). Risks of any stroke
(65 vs. 35 events; HR 1·92, 1·27—2·89) and all-cause death (19 vs. seven events; HR 2·76, 1·16—6·56) were higher in the stenting group
than in the endarterectomy group. Three procedural myocardial infarctions were recorded in the stenting group, all of which were fatal,
compared with four, all non-fatal, in the endarterectomy group. There was one event of cranial nerve palsy in the stenting group compared with
45 in the endarterectomy group. There were also fewer haematomas of any severity in the stenting group than in the endarterectomy group (31
vs. 50 events; p=0·0197). The investigators concluded that at present, carotid endarterectomy should remain the treatment of choice for
patients suitable for surgery. The primary outcome analysis of the efficacy of carotid artery stenting compared with endarterectomy is not yet
available.194
The CREST trial is the largest trial examining the relative effectiveness of carotid artery stenting (CAS) versus carotid endarterectomy (CEA) in
preventing stroke, myocardial infarction, and death.164 The trial included 2502 patients with a median follow-up period of 2.5 years. The primary
end point was the composite of any stroke, myocardial infarction, or death during the peri-procedural period or ipsilateral stroke within four
Fourth Edition FINAL
Update: September 2012
Page 45 of 69
CBPRSC 2012
Section 2: Prevention
2.7 Carotid Disease and Intracranial Atherosclerosis
years after randomization. The study results showed no significant difference in the estimated four-year rates of the primary end point between
the stenting group and the endarterectomy group (7.2 percent and 6.8 percent, respectively; hazard ratio with stenting, 1.11; 95% confidence
interval, 0.81 to 1.51; P=0.51). There was no differential treatment effect with regard to the primary end point according to symptomatic status
(P = 0.84) or sex (P = 0.34). The four-year rate of stroke or death was 6.4 percent with stenting and 4.7 percent with endarterectomy (hazard
ratio, 1.50; P=0.03); the rates among symptomatic patients were 8.0 percent and 6.4 percent (hazard ratio, 1.37; P=0.14), and the rates among
asymptomatic patients were 4.5 percent and 2.7 percent (hazard ratio, 1.86; P = 0.07), respectively. Peri-procedural rates of individual
outcomes differed between the stenting group and the endarterectomy group: for death (0.7% vs. 0.3%, P=0.18), for stroke (4.1% vs. 2.3%, P =
0.01), and for myocardial infarction (1.1% vs. 2.3%, P = 0.03). After this period, the incidences of ipsilateral stroke with stenting and with
endarterectomy were similarly low (2.0% and 2.4%, respectively; P=0.85). The investigators concluded that among patients with symptomatic
or asymptomatic carotid stenosis, the risk of the composite primary outcome of stroke, myocardial infarction, or death did not differ significantly
in the group undergoing carotid-artery stenting and the group undergoing carotid endarterectomy. During the peri-procedural period, there was
a higher risk of stroke with stenting and a higher risk of myocardial infarction with endarterectomy.
The clinical trials and subgroup analysis reports for stenting versus endarterectomy have indicated that patient age greater than 70 years has a
significant impact on primary outcomes.165 A preplanned meta-analysis was recently reported by the Carotid Stenting Trialists Collaboration
that included patient-level data for 3433 patients with symptomatic carotid stenosis who were randomly assigned and analyzed in the
Endarterectomy versus Angioplasty in Patients with Symptomatic Severe Carotid Stenosis (EVA-3S) trial, the Stent-Protected Angioplasty
versus Carotid Endarterectomy (SPACE) trial, and the International Carotid Stenting Study (ICSS). The data was pooled and analyzed with
fixed-effect binomial regression models adjusted for source trial. The primary outcome event was any stroke or death. In the first 120 days after
randomization (ITT analysis), any stroke or death occurred significantly more often in the carotid stenting group (153 [8·9%] of 1725) than in the
carotid endarterectomy group (99 [5·8%] of 1708, risk ratio [RR] 1·53, [95% CI 1·20—1·95], p=0·0006; absolute risk difference 3·2 [1·4—4·9]).
Of all subgroup variables assessed, only age significantly modified the treatment effect: in patients younger than 70 years (median age), the
estimated 120-day risk of stroke or death was 50 (5·8%) of 869 patients in the carotid stenting group and 48 (5·7%) of 843 in the carotid
endarterectomy group (RR 1·00 [0·68—1·47]); in patients 70 years or older, the estimated risk with carotid stenting was twice that with carotid
endarterectomy (103 [12·0%] of 856 vs. 51 [5·9%] of 865, 2·04 [1·48—2·82], interaction p=0·0053, p=0·0014 for trend). In the PP analysis, risk
estimates of stroke or death within 30 days of treatment among patients younger than 70 years were 43 (5·1%) of 851 patients in the stenting
group and 37 (4·5%) of 821 in the endarterectomy group (1·11 [0·73—1·71]); in patients 70 years or older, the estimates were 87 (10·5%) of
828 patients and 36 (4·4%) of 824, respectively (2·41 [1·65—3·51]; categorical interaction p=0·0078, trend interaction p=0·0013]. The
conclusions stated by the Trialist Collaboration were that stenting for symptomatic carotid stenosis should be avoided in older patients (age
≥70 years), but might be as safe as endarterectomy in younger patients. A sub-group analysis on the influence of sex on outcomes in the
CREST trial found no significant differences in the primary endpoints of stroke, myocardial infarct or death during the peri-procedural period
(Howard 2011). End-point events occurred in 35 (4·3%) of 807 men assigned to carotid artery stenting compared with 40 (4·9%) of 823
assigned to carotid endarterectomy (HR 0·90, 95% CI 0·57–1·41) and 31 (6·8%) of 455 women assigned to carotid artery stenting compared
with 16 (3·8%) of 417 assigned to carotid endarterectomy (1·84, 1·01–3·37; interaction p=0·064).
Intracranial Stenting:
The SAMMPRIS trial, released in 2011, is the first large open-label clinical trial that randomly assigned patients who had a recent transient
ischemic attack or stroke attributed to stenosis of 70 to 99% of the diameter of a major intracranial artery to aggressive medical management
alone or aggressive medical management plus percutaneous transluminal angioplasty with stenting (PTAS), using the Wingspan stent
system.143 The primary end point was stroke or death within 30 days after enrollment or after a revascularization procedure for the qualifying
lesion during the follow-up period or stroke in the territory of the qualifying artery beyond 30 days. Enrollment was stopped after 451 patients
underwent randomization, because the 30-day rate of stroke or death was 14.7% in the PTAS group (nonfatal stroke, 12.5%; fatal stroke,
2.2%) and 5.8% in the medical-management group (nonfatal stroke, 5.3%; non–stroke-related death, 0.4%) (P=0.002). Beyond 30 days, stroke
in the same territory occurred in 13 patients in each group. Currently, the mean duration of follow- up, which is ongoing, is 11.9 months. The
probability of the occurrence of a primary end-point event over time differed significantly between the two treatment groups (P=0.009), with 1year rates of the primary end point of 20.0% in the PTAS group and 12.2% in the medical-management group.
Fourth Edition FINAL
Update: September 2012
Page 46 of 69
CBPRSC 2012
Section 2: Prevention
2.8 Assessment and Management of Sleep Apnea
Best Practice Recommendation 2.8
Assessment and Management of Obstructive Sleep Apnea
* NEW Topic for 2012
Introductory Notes About Stroke and Sleep Apnea:
Sleep apnea (OSA) is an emerging risk factor for stroke and a new section on sleep apnea has been added for the 2012
update of the Stroke prevention section of the Canadian Best Practice Recommendations for Stroke Care to recognize its
important role. Sleep apnea is usually considered an ongoing condition that disrupts a person’s sleep with persistent pauses
in breathing or breathing becomes shallow. Patients will move out of deep sleep and into light sleep several times a night.
These episodes wake the sleeper as he or she gasps for air. It prevents restful sleep and is associated with high blood
pressure, arrhythmia, stroke and heart failure. (www.medicinenet.com/sleep_apnea; www.nhlbi.nih.gov/health/healthtopics/topics/sleepapnea)
There are three types of sleep apnea, which include obstructive sleep apnea, central sleep apnea and mixed sleep apnea.
Obstructive sleep apnea is seen most often with stroke patients and results from a relaxation of the muscles around the back
of the tongue and soft palate, causing a complete or partial block to the airway and resulting in decreased oxygen levels.
Central sleep apnea occurs when there are interruptions to the signals from the brain to the muscles that control breathing.
Mixed sleep apnea is diagnosed when both central and obstructive sleep apnea are present.
The recommendations included in this section focus on obstructive sleep apnea (more common in patients following a
stroke) assessment and management in patients who have experienced a stroke or transient ischemic attack. Healthcare
providers should also be aware that stroke patients may also experience central or mixed sleep apnea, but this is much less
common. In addition, many of the screening, assessment and management strategies listed below also apply to patients
with diagnosed sleep apnea who are also at increased risk of a first stroke or transient ischemic attack.
2.8
Obstructive sleep apnea (OSA) is an emerging risk factor for stroke and has also been shown to be present in many
patients following a stroke [Evidence Level B]. Preventative strategies should be in place for people with obstructive
sleep apnea and stroke patients with sleep disturbance symptoms that emerge following stroke [Evidence Level B].
2.8.1 Screening and Assessment for Sleep Apnea
Patients who have experienced a stroke or TIA are more likely to experience obstructive sleep apnea following their
stroke compared to people who have not had a stroke [Evidence level A]. Patients who have experienced a stroke or
TIA should be screened* at all transition points and follow-up visits for the presence of sleep apnea symptoms [Evidence
level A], using a validated sleep apnea screening tool. (* Screening is defined as using a standardized brief clinical
questionnaire to determine sleep patterns, symptoms of OSA, and likelihood that a patient may have sleep apnea – refer
to Implementation Tools section for suggested tools).
i. Patients with sleep apnea post-stroke often do not display typical clinical characteristics of sleep apnea
(Evidence level B); therefore initial screening for sleep apnea using a validated screening questionnaire
should be considered for patients with:
a. recurrent stroke [Evidence level A].
b. fragmented sleep, difficulty sleeping, daytime sleepiness [Evidence level B].
c. increased frequency of nocturia or snoring [Evidence level B].
ii. Patients with symptoms suggestive of sleep apnea on initial screening (as described in 2.8.1.i) should be
referred to a sleep specialist for more detailed assessment and diagnosis [Evidence level C], which may
include a more detailed sleep history, physical assessment, and a formalized sleep study [Evidence level
A].
iii. Sleep apnea screening should be considered in patients with drug resistant hypertension [Evidence Level
B] and atrial fibrillation [Evidence Level C].
Fourth Edition FINAL
Update: September 2012
Page 47 of 69
CBPRSC 2012
2.8.2
Section 2: Prevention
2.8 Assessment and Management of Sleep Apnea
Management of Obstructive Sleep Apnea in Patients with Stroke
Stroke prevention strategies, including specific targeted management strategies for sleep apnea, should be initiated for
patients with confirmed sleep apnea pre or post stroke or TIA, based on objective clinical assessment and
investigations [Evidence Level B].
i. The management of all treatable vascular disease risk factors should be optimized in patients with
confirmed sleep apnea pre or post stroke [Evidence Level B] as described in other sections of these best
practice recommendations including: blood pressure, diet, sodium intake, physical activity, obesity,
alcohol reduction, smoking cessation and antithrombotic therapy.
ii. First line therapies for the treatment of sleep apnea before or following a stroke may include:
a. Avoidance of hypnotic and sedative medications and alcohol (sedatives) (Evidence level B);
b. Positional therapy [Evidence level B]: Patients should be advised to stay off their back where
possible to alleviate apnea-type symptoms when they have received a diagnosis of supine related
sleep apnea [Evidence level B].
c. Weight loss [Evidence level B]
d. Continuous positive airway pressure (C-PAP) [Evidence level B].
e. Dental appliances [Evidence level B] in consultation with dental specialists.
iv. Severity of sleep apnea, presenting symptoms and patient compliance are factors that should be
considered in the selection of treatment modalities [Evidence level B] .
v. Determination of severity of sleep apnea should be based on clinical symptoms, sleep architecture
(patterns), number of respiratory events per hour and oxygen saturation (Evidence level B).
vi. Patients and family members should be provided ongoing education, counseling and support regarding
the signs, symptoms and risks of sleep apnea [Evidence Level B]; compliance with treatment to reduce
stroke recurrence, and increase recovery; and, signs and symptoms of stroke and appropriate actions to
take when any stroke symptoms appear [Evidence Level B].
vii. Refer to available comprehensive Sleep Apnea Guidelines for additional information on the management
of sleep apnea (Refer to Canadian Thoracic Society Guidelines; American Academy of Sleep Medicine).
166,167
2.8.3
2.8.4
There is insufficient evidence to provide specific recommendations with respect to treatment of completely
asymptomatic sleep apnea and the risks of stroke and TIA [Evidence Level C].
i.
Based on expert opinion, the Canadian stroke sleep apnea writing group recommends that treatment be
considered in asymptomatic patients with an AHI > 20 per hour or SaO2 < 90% for greater than 12% of
total recording time [Evidence Level C]. 168
ii.
The overall vascular health status of the patient (e.g., presence of hypertension, coronary artery disease,
etc) should also be considered as a factor in treatment decisions [Evidence Level C].
Paediatric Considerations: There is no direct evidence available to demonstrate a connection in children regarding
sleep apnea and stroke. However, it is recommended that children with stroke be screened for signs and symptoms
suggesting sleep apnea [Evidence Level C], or conditions predisposing them to sleep apnea, such as obesity, sickle
cell disease, severe strokes, or structural airway problems (e.g., enlarged tonsils) [Evidence Level C].169,170
i. Any child with suspected sleep apnea should be referred to a paediatric sleep specialist or respirologist
for further assessment and decision-making regarding appropriate management [Evidence Level C].
Rationale
Sleep apnea is an emerging area of concern in stroke management. Sleep Disorders are an under recognized problem that
predispose people to stroke occurrence and recurrence. Sleep apnea is a modifiable risk factor that can be treated quickly to
alleviate the associated risk of stroke. Although there is a lack of randomized controlled trials, several observational studies have
demonstrated that people with obstructive sleep apnea are 1.6 to 2.7 times more likely to experience a stroke. Treating patients
with obstructive sleep apnea and stroke with continuous positive airway pressure has been shown to have positive effects on
outcomes in the post-stroke setting. Patients with untreated sleep apnea have demonstrated poorer outcomes and increased
mortality rates.
Fourth Edition FINAL
Update: September 2012
Page 48 of 69
CBPRSC 2012
Section 2: Prevention
2.8 Assessment and Management of Sleep Apnea
System Implications
• Implementation of protocols for assessment of sleep disturbances or risk factors for sleep apnea in stroke patients
• Implementation of criteria to prioritize appropriate access to sleep specialists and sleep study labs as required, especially in
under-resourced regions
• Regional service capacity and clearly communicated access criteria for specialized sleep medicine services
• Resources and opportunities for education on sleep apnea and correct use of screening and assessment tools for healthcare
professionals
• Development of patient and family education resources on the awareness of sleep apnea as a risk factor pre and post stroke,
and the importance of screening and treatment adherence
Performance Measures
1. Proportion of patients presenting to hospital with stroke or TIA who have a history of confirmed sleep apnea.
2. Proportion of patients with stroke or TIA who are screened for sleep apnea symptoms before discharge from acute care,
during inpatient or outpatient stroke rehabilitation, and/or during stroke prevention follow-up visits.
3. Proportion of patients who are screened and referred for sleep studies following a stroke or TIA.
4. Proportion of patients referred for sleep studies following a stroke or TIA who receive a confirmed diagnosis of sleep
apnea.
5. Proportion of stroke and TIA patients with confirmed sleep apnea with recurrent stroke or TIA at one year and 5 years
following index stroke or TIA.
6. Quality of life for patients with sleep apnea and their family members (measured at intervals to monitor changes over
time with treatment).
7. Sustained weight loss for overweight or obese patients with stroke and sleep apnea at regular intervals (1 year, 2 years
etc.) following achievement of target weight loss.
Measurement Notes
- Data for these indicators will be in health professional notes and assessment sections of individual charts, and should be
added to electronic health records.
Implementation Resources and Knowledge Transfer Tools
 Tools to screen for OSA
 Berlin Sleep Scale http://www.cpap-supply.com/Articles.asp?ID=178
 STOP BANG Questionnaire http://www.carolinashealthcare.org/documents/
cmcnortheast/STOPforSleep[1].pdf
 Adjusted Neck Circumference (ANC)
 Sleep Apnea Clinical Score http://www.thoracic.org/assemblies/srn/questionaires/sdq.php
 Tools to assess daytime sleepiness:
 Epworth Sleepiness Scale http://www.stanford.edu/~dement/epworth.html
 Tools to assess sleep quality:
 Pittsburg Sleep Quality Index (PSQI)




Canadian Thoracic Society Guidelines:
http://www.respiratoryguidelines.ca/sites/all/files/cts_sleep_guideline_update_2011.pdf
American Academy of Sleep Medicine: http://www.aasmnet.org/Resources/clinicalguidelines/OSA_Adults.pdf;
http://www.aasmnet.org/Resources/clinicalguidelines/040210.pdf
Practice parameters for the respiratory indications for polysomnography in children. Sleep 2011;34(3):379-88.
American Academy of Pediatrics. Clinical practice guideline: diagnosis and management of childhood obstructive sleep
apnea syndrome. http://pediatrics.aappublications.org/content/early/2012/08/22/peds.2012-1671.abstract
Summary of the Evidence
LINK to Evidence Tables For Sleep Apnea and Stroke
Epidemiological studies have shown that the prevalence of obstructive sleep apnea ranges between 17-76%, depending on the characteristics
of the cohorts included in individual studies, and the definitions of sleep apnea applied. OSA is often defined by the number of
apneas/hypopneas a subject experiences per hour of sleep, or the apnea/hypopnea index (AHI), with mild OSA often being defined as >5 AHI
and severe OSA often defined as >30 AHI.
Fourth Edition FINAL
Update: September 2012
Page 49 of 69
CBPRSC 2012
Section 2: Prevention
2.8 Assessment and Management of Sleep Apnea
Several large observational studies with patients with mild-severe sleep apnea have been reported, and about half include comparison groups
between participants with treated and untreated sleep apnea. Study population sizes have ranged from a low of 166 participants to over 5,000
participants and included both males and females. These studies have consistently reported an increased risk of stroke and new vascular
events in patients with OSA. Artz and colleagues (2005; n=1475) reported an odds ratio (OR) of 3.87 for first stroke in moderate-severe OSA
patients (AHI>20) compared to patients with an AHI<5 when controlling for hypertension.168 Similarly, Redline and colleagues (2010; n=5422)
found that the unadjusted OR for sleep apnea and stroke was 2.26 in males and 1.65 in females.171 For men in the highest AHI quartile
(AHI>19), the adjusted hazard ratio for sleep apnea and stroke was 2.86 (95% CI, 1.1–7.4). For men, each one-unit increase in AHI was
associated with a 6% increased risk of stroke. Valham and colleagues (2008; n=392) found patients with sleep apnea (AHI>5) had an
increased risk of stroke compared with patients without sleep apnea (HR of 2.89, 95% CI 1.37 to 6.09, P<0.005), and this was independent of
age, body mass index, gender, left ventricular function, coronary artery intervention, diabetes mellitus, hypertension, previous stroke/transient
ischemic attack, atrial fibrillation, and smoking.172
Sleep disturbances may be further aggravated by stroke or even caused by stroke. Studies have found that patients with sleep apnea are often
older, more obese, have suffered a previous stroke and have a higher prevalence of hypertension compared to patients without sleep apnea.
173-177 Differences between controls and patients with stroke or TIA have been found in habitual snoring (58% v.12%), AHI (27 v.6) and in
minimal oxygen saturation during sleep (82% v. 89).178 Stroke type (ischemic versus hemorrhagic stroke) has been found to be significantly
associated with having an AHI>20 (OR 4.5 [1.2–16.8]) and stroke severity has been associated with poststroke sleep apnea.179
Unrecognized and untreated sleep disorders may adversely influence rehabilitation efforts and outcomes, and increase the risk of stroke
recurrence. Iranzo and colleagues (2002) found that early neurologic worsening in first-ever hemiphseric ischemic stroke patients was
associated with sleep apnea (OR of 8.2; 95% CI, 1.3-51.2, p=0.018) and higher AHI (p=0.011).180 Kaneko and colleagues (2003) found stroke
patients with sleep apnea had worse functional capacity, as measured by the FIM, both at admission and at discharge from a rehabilitation unit,
and that for every 10-unit increase in AHI, the functional independence measure (FIM) score decreased by 2.3.181 Stroke patients with sleep
apnea also spent 30% more time in the hospital compared to patients without sleep apnea (p<0.05). It has also been found that the rate of
stroke recurrence is significantly and independently higher in patients with obstructive sleep apnea (OR=1.52, P<0.05).182
Sleep apnea has been also been associated with drug-resistant hypertension. Demede and colleagues (2011) found that patients with
resistant-hypertension were at higher risk of having sleep apnea compared to non-resistant hypertension patients (OR = 2.46, 95% CI: 1.03–
5.88, P < .05).183 Similarly, Friedman and colleagues (2010) found that AHI severity was greater in patients with drug-resistant hypertensive
than in a controlled hypertension group (AHI: 43.0+5.4 versus 18.1+4.2 events per hour of sleep; P=0.02).184 Likewise, in a logistic regression
model, OSA was strongly and independently associated with drug-resistant hypertension (OR 4.8, 95% CI, 2.0 to 11.7).185
Associations between sleep apnea and atrial fibrillation have also been established. Abe and colleagues (2010) identified associations between
paroxysmal atrial fibrillation incidence and obstructive sleep apnea (P < 0.009) and prevalent Paroxysmal Atrial Fibrillation (P < 0.001)
occurring during sleep.186 Braga and colleagues (2009) found that patients with atrial fibrillation had higher rates of sleep apnea compared to a
control group (81.6% versus 60%, p = 0.03) and that oxygen saturation parameters were significantly worse in patients with atrial fibrillation:
lower SaO2 nadir (81.9% versus 85.3%, p = 0.01); mean SaO2 (93.4% versus 94.3%, p = 0.02); and a longer period of time below 90% (26.4
min versus 6.7 min, p = 0.05).187 In a USA population-based observational study, obstructive sleep apnea and severity measure of obstructive
sleep apnea were identified as univariate predictors of atrial fibrillation (HR 2.18, 95% CI 1.34-3.54)188 Similarly, Stevenson and colleagues
(2008) found that the adjusted odds ratio for the association between atrial fibrillation and sleep disordered breathing (AHI >15) was 3.04 (95%
CI 1.24–7.46, P= 0.02).189 Untreated sleep-apnea was also associated with recurrence of atrial fibrillation after cardioversion procedures
(p=.013).190 In a recent meta-analysis of almost 4,000 patients, 4 studies demonstrated that the presence of obstructive sleep apnea increased
the risk of atrial fibrillation recurrence after catheter ablation, and 2 showed no significant difference using multivariate analysis.191 The authors
found that OSA patients had a 25% greater risk of atrial fibrillation recurrence after catheter ablation compared to controls (RR 1.25, 95% CI
1.08 to 1.45, p= 0.003); however, the heterogeneity test showed significant differences among the individual studies (chi-square =9.77, p =0.08,
I2 =49%).
With evidence suggesting sleep apnea is an independent risk factor for stroke and that sleep-disordered breathing may be exacerbated by
stroke, the next question to address is whether treatment of sleep apnea reduces the risk of stroke and other serious vascular events. There
are several treatment modalities that might be considered including positional therapy, surgery, lifestyle modification, dental appliances, and
continuous positive airway pressure (CPAP). Svatikova and colleagues (2011) found that the use of positional therapy to treat sleep apnea in
an ischemic stroke population reduced the absolute percentage of time spent in a supine position while sleeping by 36% (95% CI: 18–55%, P <
0.001) and reduced AHI by a relative 19.5% (95% CI: 4.9–31.9%, P = 0.011). 192
A Cochrane review investigated the use of surgery to treat obstructive sleep apnea.193 A meta-analysis was not possible and the authors found
that the studies identified in the review did not provide evidence to support the use of surgery in sleep apnoea/hypopnoea syndrome, as overall
significant benefit was not demonstrated. Long-term follow-up of patients who undergo surgical correction of upper airway obstruction is
required to aid in identifying suitable treatment and candidates for surgery.
Fourth Edition FINAL
Update: September 2012
Page 50 of 69
CBPRSC 2012
Section 2: Prevention
2.8 Assessment and Management of Sleep Apnea
Continuous positive airway pressure therapy has been shown to significantly reduce the occurrences of Paroxysmal Atrial Fibrillation (P <
0.001);186 improve sleep-related symptoms;194-196 and may prevent the occurrence of new vascular events.197-198 Giles and colleagues
(2006) found CPAP improved Epworth Sleepiness Scale scores compared to controls, reduced AHI compared to oral appliances, improved
sleep efficiency, and improved minimum oxygen saturation.199 Martinez-Garcia and colleagues (2011) found that patients with moderatesevere obstructive sleep apnea (AHI≥20) who could not tolerate CPAP showed an increased adjusted incidence of new ischemic strokes (HR
2.87, 95% CI 1.11-7.71, p=0.03), compared to patients with moderate-severe OSA who tolerated CPAP.200
It has also been reported that CPAP use is associated with improved poststroke outcomes. In a randomized trial, Bravata and colleagues
(2011) found that stroke patients randomized to CPAP treatment had greater median improvement in NIHSS (-3.0) than control patients (-1.0),
p=0.03.201 The greatest improvement was observed in patients who started CPAP therapy within 48 hours of stroke onset. Hsu and colleagues
(2006) found that CPAP use was positively correlated with a better Barthel Index scores (p=0.035) and language scores (ACE subscale)
(p=0.013) amongst stroke patients with sleep apnea.202 Similarly, Parra and colleagues (2011) found that the percentage of patients with
neurological improvement 1 month after stroke was significantly higher in the CPAP group (Rankin scale 90.9 vs. 56.3% (p<0.01); Canadian
scale 88.2 vs. 72.7% (p<0.05)).In a randomized trial, CPAP use was associated with improved blood pressure in patients with drug-resistant
hypertension.203 Patients who used CPAP for more than 5.8 hours a night showed a greater reduction in daytime diastolic BP (-6.12mmHg, CI 1.45; -10.82, P=0.004), 24-h diastolic BP (-6.98mmHg, CI -1.86; -12.1, P=0.009) and 24-h systolic BP (-9.71mmHg, CI -0.20; -19.22, P=0.046).
Studies consistently report, however, that CPAP adherence rates are low among sleep apnea patients.
Brown and colleagues (2005) explored the cost-effectiveness of screening for sleep apnea in patients with acute ischemic stroke.204 A decision
tree modeled 2 alternative strategies: polysonography followed by 3 months of CPAP for those found to have OSA versus no screening. The
primary outcome was the utility gained through screening and treatment in relation to two willingness-to-pay thresholds of $50 000 and $100
000 per quality-adjusted life year (QALY). Screening resulted in an incremental cost-effectiveness ratio of $49 421 per QALY. Screening was a
cost-effective option as long as CPAP treatment improves patient utilities by >0.2 for a willingness-to-pay of $50 000 per QALY. These results
are preliminary and clinical trials are required to further investigate the effectiveness of CPAP and oral appliances in improving stroke outcomes
in patients with obstructive sleep apnea.
Fourth Edition FINAL
Update: September 2012
Page 51 of 69
CBPRSC 2012
Section 2: Prevention
2.9 Management of Smoking Cessation
Best Practice Recommendation 2.9
Management of Smoking Cessation
* Moderate revisions for 2012
All members of the interdisciplinary team should address smoking cessation and a smoke-free environment at every healthcare
encounter for active smokers.205
i.
In all healthcare settings along the stroke continuum (inpatient, ambulatory, and community), patient smoking status
should be identified, assessed and documented [Evidence Level A].
ii.
Provide unambiguous, non-judgmental, and patient-specific 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 cessation attempt–either directly or through referral to appropriate
resources [Evidence Level A].
iv. People who are not ready to quit should be offered a motivational intervention to help enhance their readiness to quit
[Evidence Level B].
v. A combination of pharmacological therapy and behavioural therapy should be considered in all smoking cessation
programs and interventions [Evidence Level A].
vi. The three classes of pharmacological agents that should be considered as first-line therapy for smoking cessation are
nicotine replacement therapy, bupropion, and varenicline [Evidence Level A].
i.
The choice of appropriate pharmacotherapy should take into account the patient’s medical stability, clinical
needs, other medical factors, and patient preferences [Evidence Level C]. Refer to Table 2.9
Pharmacotherapy in Smoking Cessation Treatment.
vii. For admitted stroke patients who are current smokers, protocols should be in place to manage nicotine withdrawal
during hospitalization [Evidence Level B].
viii. There is a lack of clear evidence regarding the timing to initiate nicotine withdrawal/replacement therapy in patients
following a stroke. Expert opinion suggests this may begin as soon as it is medically appropriate, and should take
into consideration the type of stroke event, stroke severity, patient interest, and physician comfort level. [Evidence
Level C].
i.
In cases where the patient is likely to continue to smoke while in hospital or immediately after discharge,
nicotine replacement therapy should be offered as a safer alternative to smoking [Evidence Level C].
ix. The importance of smoking cessation in adolescent and teen populations must be considered a priority and
addressed, especially in young patients who have already experienced a stroke [Evidence Level C].
i.
In teens and young adults, it is recommended that screening for smoking status be carried out in a
confidential, non-judgmental environment without the presence of parents or caregivers where possible
[Evidence Level B].
x. Health care providers working with pediatric populations should counsel patients, parents and caregivers about the
potential harmful effects of early onset smoking and second- hand smoke on the health of their children. [Evidence
Level C].
Note, the term ‘Smoking’ in these recommendations refers to tobacco and other inhaled substances.
Rationale
The Quality of Stroke Care in Canada stroke audit report found that among all Canadians who experienced a stroke in 2008-09,
41% were current smokers, and more prominent in younger adult stroke patients (less than 49 years old).206 The Interstroke
study reported that current smokers had increased risk of stroke, with the impact greater on ischemic stroke compared to
hemorrhagic stroke, and this risk increased with the number of cigarettes smoked per day.54 Also the significant impact of
smoking on stroke was second only to hypertension. The CAN-ADAPTT working group has reported that approximately 17% of
Canadians are current smokers, and a large proportion has been shown to be willing to make a quit attempt.207 Health care
providers have an important role to play in assisting individuals to quit smoking. Moreover, even brief interventions by providers
are known to be effective in increasing the likelihood of a quit attempt by a person who smokes. Clinical practice guidelines are
known to be an important and effective provider tool to close the gap between recommended care and actual care provided4.
Smoking cessation has been found to reverse/reduce stroke risk as duration of being smoke-free increased. Female patients who
have had a stroke are at additional risk for recurrent stroke if they continue to smoke and are taking oral contraception or
estrogen-based hormone replacement therapy.
Fourth Edition FINAL
Update: September 2012
Page 52 of 69
CBPRSC 2012
Section 2: Prevention
2.9 Management of Smoking Cessation
System Implications
•
A focus on arterial health for paediatric cases–such as diet, exercise, non-smoking, avoidance of drugs that increase
stroke risk.
•
Access to risk factor management programs such as smoking cessation programs should be available in all communities,
primary healthcare settings and workplaces.
•
Government actions to restrict smoking in public areas and discourage smoking through legislation and taxation
initiatives.
•
Coordinated efforts among stakeholders such as Heart and Stroke Foundations (national and provincial), the Canadian
Stroke Network, public health agencies, ministries of health and care providers across the continuum to produce patient,
family and caregiver education materials with consistent information and messages on risk factor management.
•
Coordinated process for ensuring access to and awareness of educational materials, programs, activities and other
media related to risk factor management by healthcare professionals, patients and caregivers, including advertising the
availability of educational material, effective dissemination mechanisms and follow-up.
•
Educational resources, that are culturally and ethnically appropriate, are available in multiple languages and that address
the needs of patients with aphasia.
Performance Measures
1. Proportion of patients with documented smoking status recorded on patient record.
2. Proportion of patients with stroke and TIA with a history of cigarette smoking who are given smoking cessation advice
and counseling during acute hospital stay, inpatient and outpatient rehabilitation, and during secondary prevention visits.
3. Proportion of stroke and TIA patients who participate in a smoking cessation program who are smoke-free at 6 months,
one year and two years.
Implementation Resources and Knowledge Transfer Tools
o Canadian Best Practice Recommendations for Stroke Care Smoking Cessation Pharmacology Summary Table
(weblink)
o CAN-ADAPTT Smoking Cessation Guidelines
https://www.nicotinedependenceclinic.com/English/CANADAPTT/Pages/Home.aspx
o CAN-ADAPTT Tool Kit:
http://knowledgex.camh.net/primary_care/toolkits/addiction_toolkit/smoking/Pages/tools_resources.aspx
o Smoking Cessation and the Cardiovascular Specialist: Canadian Cardiovascular Society Position Paper.
http://www.onlinecjc.ca/article/S0828-282X(10)00076-0/fulltext
o QUIT NOW - Health Canada Smoking Cessation: http://www.hc-sc.gc.ca/hc-ps/tobac-tabac/quit-cesser/index-eng.php
o CADTH Smoking Cessation Pharmacology 2011.
http://www.cadth.ca/media/pdf/CADTH_Smoking_Cessation_Summary_for_Health_Care_Providers_e.pdf
o Canadian Public Health Association: http://www.cpha.ca/en/activities/stopsmoking.aspx
o US Tobacco guidelines http://www.surgeongeneral.gov/initiatives/tobacco/index.html
o Skills and tools for clinicians on motivational interviewing (handouts, self-evaluations, checklists):
http://www.motivationalinterview.org/clinicians/Side_bar/skills_maintenence.html
o The Motivational Interviewing Assessment: Supervisory Tools for Enhancing Proficiency
(manual): http://www.motivationalinterview.org/clinical_supervisors/side_bar/mia_step_manual.html
o The Change Book: http://www.motivationalinterview.org/Documents/The_Change_Book_2nd_Edition.pdf
o The Change Book Workbook: http://www.motivationalinterview.org/Documents/
The_Change_Book_2nd_Edition_Workbook.pdf
o Contemplation latter: http://web.fu-berlin.de/gesund/gesu_engl/conner9.htm; http://www.nd.gov.hk/pdf/bdf-2010R2q13-eng.pdf;
o Readiness-ruler - http://www.quitlinenc.com/health-professionals/screening-brief-intervention/counseling-deliverymethods/readiness-rulerHeaviness of Smoking Index: http://www.iarc.fr/en/publications/pdfsonline/prev/handbook12/Tobacco_vol12_appendices.pdf; http://mdquit.org/fax-to-assist/module-2
Fourth Edition FINAL
Update: September 2012
Page 53 of 69
CBPRSC 2012
Section 2: Prevention
2.9 Management of Smoking Cessation
Stages of Change Questionnaires: http://www.uri.edu/research/cprc/Measures/Smoking11.htm;
http://www.livestrong.com/article/332795-stages-of-change-smoking-cessation/
o Tobacco Use questionnaires:
o http://youcanmakeithappen.ca/wp-content/uploads/2011/08/Appendix-B3-Tobacco-Use-Questionnaire.pdf
o http://www.cdc.gov/nchs/data/nhis/tobacco/1997_forward_tobacco_questions.pdf
o http://www.who.int/tobacco/surveillance/en_tfi_tqs.pdf
o http://yss.uwaterloo.ca/yss12/dynamicdoc_app/view/document_files/yss12_english_module_A.pdf
o http://www.anthc.org/chs/wp/tobacco/upload/FINAL-ANMC-Tobacco-use-questionnaire-May-08.PDF
Summary of the Evidence
o
LINK to Evidence Tables for Management of Smoking Cessation
Tobacco smoking remains a significant risk factor for many chronic diseases including cardiovascular disease.208 In Canada, the
Public Health Agency of Canada report on cardiovascular disease found that 15.3 percent of the population over the age of
twelve self-reports being active smokers in 2007; this is an improvement from 19.9 percent reported in 2000.209 Smoking policies
and regulations have made notable progression over that same timeframe. The United States Department of Health and Human
Services has stated that tobacco “presents a rare confluence of circumstances: (1) a highly significant health threat; (2) a
disinclination among clinicians to intervene consistently; and (3) the presence of effective interventions”.210 The World Health
Organization “M-Power” report describes smoking as a global tobacco epidemic.211 In that report, 6 policies were recommended
to reverse the tobacco epidemic, all of which are targeted at the national level. These policies are tobacco use and prevention
policies; protection of people from tobacco smoke; assistance in quitting tobacco use; warnings about the dangers of tobacco;
enforcement of bans on tobacco advertising, promotion and sponsorship; and raising taxes on tobacco.
Research has demonstrated that current smokers who smoke 20 or more cigarettes per day have an associated increase of
stroke risk approximately 2 to 4 times that of non-smokers. 212-216 Overall, given that an estimated 25 percent of adults are active
smokers, approximately 18 percent of strokes may be attributed to active smoking.217
Smoking acts as a risk factor in a dose-dependent fashion, such that heavy smokers have more risk than light smokers, who in
turn have more risk than nonsmokers.218,219 Results of a recent study demonstrated that the relative risk for ischemic stroke
associated with smoking fewer than 20 cigarettes per day was 1.56 when compared with non-smokers and 2.25 when 20 or more
cigarettes were smoked per day.220,221 Reported relative risks for hemorrhagic stroke among smokers followed a similar pattern.
Within a male population, smoking fewer than 20 cigarettes was associated with a 1.6- fold increase for intracerebral hemorrhage
and a 1.8-fold increase for subarachnoid hemorrhage compared with non-smokers. 220,221 When the rate of smoking increased to
20 cigarettes or more, the associated risk increased to 2.1 and 3.2 for intracerebral hemorrhage and subarachnoid hemorrhage,
respectively. A study conducted within a female subject population yielded a similar pattern of risk.220
Risk associated with current cigarette smoking is greatest in the middle years and declines with age.91 The Cardiovascular STudy
in the ELderly (CASTEL) reported that the relative risk associated with current smoking compared with current non-smokers was
1.60 for fatal stroke.222 Mortality was particularly high among current smokers who had been smoking for 40 or more years (7.2%
v. 1.8% for non-smokers, p < 0.01).
There is a growing body of evidence examining smoking cessation interventions in adolescents and parental groups to protect
young children from second-hand smoke. Next to alcohol, the most commonly used substance among college students is
tobacco. 223 Audrian-McGovern and colleagues (2011) found in adolescents, those who received motivational interview-based
interventions were about 60% less likely to try to quit smoking than those who received structured brief advice (OR: 0.41 [CI:
0.17– 0.97]).224 However, adolescents who received motivational interviewing showed a greater reduction in cigarettes smoked
per day than adolescents who received structured brief advice (5.3 vs. 3.3 fewer cigarettes per day). Parental quit-strategies that
took place in hospitals, pediatric clinical settings, well-baby clinics, and family homes were found to successfully increase
parental quit rates.225 Children aged 4-17 were found to benefit significantly from parental smoking cessation.
There are several non-pharmacotherapy interventions that have been shown to be effective smoking cessation interventions. In a
recent Cochrane Review, it was found that smoking cessation interventions that used behavioural therapy strategies, either solely
or in combination with pharmacotherapy, produced a significant effect in favour of the intervention (RR=1.43, 95% CI 1.03-1.98,
p=0.032).226 Similarly, Cabezas and colleagues (2011) examined the effects of a behavioural based smoking cessation
Fourth Edition FINAL
Update: September 2012
Page 54 of 69
CBPRSC 2012
Section 2: Prevention
2.9 Management of Smoking Cessation
intervention strategy on abstinence at one year.227 The intervention included brief motivational interviews for smokers at the
precontemplation–contemplation stage, brief intervention for smokers in preparation–action who do not want help, intensive
intervention with pharmacotherapy for smokers in preparation–action who want help and reinforcing intervention in the
maintenance stage. Control group involved usual care. The 1-year continuous abstinence rate at follow-up was 8.1% in the
intervention group and 5.8% in the control group (P = 0.014). The odds of quitting of the intervention versus control group was
1.50 (95% confidence interval = 1.05–2.14).
Ussher and colleagues (2012) performed a Cochrane Review to determine whether exercise-based interventions were an
effective smoking cessation tool.228 They found three studies that showed significantly higher abstinence rates in a physically
active group versus a control group at end of treatment.
A systematic review and meta-analysis of smoking cessation therapies found that Bupropion trials were superior to controls at
one year (12 RCTs, OR1.56, 95% CI, 1.10–2.21, P = 0.01) and at three months (OR 2.13, 95% CI, 1.72–2.64).229 Two RCTs
evaluated the superiority of bupropion versus NRT at one year (OR 1.14, 95% CI, 0.20–6.42). P =< 0.0001) and also at
approximately 3 months (OR 3.75, 95% CI, 2.65–5.30). Three RCTs evaluated the effectiveness of varenicline versus bupropion
at 1 year (OR 1.58, 95% CI, 1.22–2.05) and at approximately three months (OR 1.61, 95% CI, 1.16–2.21). Using indirect
comparisons, varenicline was superior to NRT when compared to placebo controls (OR 1.66, 95% CI 1.17–2.36, P = 0.004) or to
all controls at one year (OR 1.73, 95% CI 1.22–2.45, P = 0.001). This was also the case for three-month data. Adverse events
were not systematically different across studies. A study that examined the effects of bupropion for relapse prevention found that
it was associated with a higher point-prevalence for smoking abstinence compared to placebo (p=.007), and this was
independent of past history of depression.230
Fourth Edition FINAL
Update: September 2012
Page 55 of 69
CBPRSC 2012
Section 2: Prevention
2.9 Management of Smoking Cessation
Table 2.9: Pharmacotherapy for Smoking Cessation in Patients with Stroke and TIA
This table provides a summary of the pharmacotherapeutic properties, side effects, drug interactions and other important information on the current
medications available for use in Canada. This table should be used as a reference guide by health care professionals when selecting an appropriate
agent for individual patients. Patient compliance, patient preference and/or past experience, side effects, and drug interactions should all be taken
into consideration during decision-making, in addition to other information provided in this table and available elsewhere regarding these
medications.
Initial Treatment Length
Time to Peak Effect
Nicotine patch
8-12 weeks
Requires 2-3 days to
get maximal serum
levels
Nicotine gum
4-36 weeks
After 20-30 min of
chewing
Nicotine inhaler
12-24 weeks
Within 15 minutes after
forced inhalation for 20
minutes
Nicotine Lozenge
4-24 weeks
After 20-30 min of
sucking
As an aid to smoking cessation
Indications
24 Hour patch: 21 mg
for 3 to 6 weeks, then
14 mg for 2 to 4 weeks
then7 mg for 2 to 4
weeks.
Usual
Dosing
16 Hour patch: 15mg
for 6 weeks then10mg
for 2 weeks then 5mg
for 2 weeks
<25 cig/d or smokes
>30 min upon waking: 2
mg
>25 cig/d or smokes
<30 min upon waking: 4
mg
Week 1-6; 1 piece q12h (at least 9/d)
Week 7-9: 1 piece q24h
Week 10-12: 1 piece
q4-8h
Weeks 1-12: 6-12
cartridges per day then
gradually reduce as
able.
(min 6/d for first 3-6
weeks)
Stop when reduced to
1-2 per day
Max: 12 cartridges per
day
Stop when reduced to
1-2 per day
Max: 20-30 pieces per
day
Fourth Edition FINAL
Update: September 2012
Polacrilex:
Smokes >30 min upon
waking: 2mg
Smokes <30 min upon
waking: 4mg
Bitartarate:
< 20 cig/d: 1 mg
> 20 cig/d: 2 mg
Bupropion
7-12 weeks
1-2 weeks
Varenicline
12 -24weeks
1-2 weeks
As an aid to smoking
cessation, major
depressive disorder,
seasonal affective
disorder
As an aid to smoking
cessation
150 mg once daily x 3
days then 150 mg BID
x 7-12 weeks. Begin 12 weeks prior to
selected quit date
0.5 mg once daily x 3
days then 0.5 mg BID x
4 days then 0.5-1 mg
BID x 12 weeks. Begin
1-2 weeks prior to
selected quit date.
Week 1-6; 1 lozenge
q1-2h
Week 7-9: 1 piece q24h
Week 10-12: 1 piece
q4-8h
Stop when reduced to
1-2 per day
Max: 30 mg/day
Page 56 of 69
CBPRSC 2012
Special Dosing Notes
Side Effects
Effect of Food and
Other Administration
Notes
Section 2: Prevention
2.9 Management of Smoking Cessation
Nicotine patch
Nicotine gum
Nicotine inhaler
Nicotine Lozenge
Smokers are precise in the way they titrate their smoking to maintain nicotine levels, and dosing should
be titrated and personalized accordingly. A common issue is under dosing NRT in heavier smokers.
Dosing guide: 1 cigarette = 1 mg nicotine. E.g., if smoke 2 packs per day, offer 2 x 21mg patches plus
gum or inhaler for cravings. In the “Reduce to Quit” approach, patients may continue to smoke while on
the patch as they are receiving nicotine via the patch/gum/lozenge/inhaler and should be smoking fewer
cigarettes, which is the goal.
Bupropion
Must titrate dose when
discontinuing
Varenicline
Upward titration to
reduce nausea from
drug
Headache, GI upset,
dizziness, nausea,
disturbed sleep, rash at
site
Headache, GI upset,
hiccups, disturbed
sleep, sore jaw
Irritation of throat
and nasal passages,
sneezing, coughing
especially in those with
bronchospastic
disease, hiccups
GI upset, mouth/throat
soreness, hiccups
Dry mouth, insomnia,
agitation, vivid dreams,
unease. Risk of seizure
is 1/1000 (risk factors
include those with
seizure or eating
disorders)
Nausea, insomnia,
abnormal/vivid dreams.
Health Canada warning
for psychiatric effects
Do not cut patch,
causes rapid
evaporation rending
product useless. Rotate
patch site to avoid skin
irritation.
Recent food and
beverage impairs
release of nicotine.
Avoid food and drink 15
min before or while
using gum (30 min for
caffeine/acidic
products). Not regular
chewing gum; use bite,
chew, park technique.
Not a true inhaler (is a
vaporizer) so best
effect with continuous
puffing; nicotine
absorbed from oral
mucosa. Cold
temperatures can
decrease absorption
rate.
Recent food and
beverage impairs
release of nicotine.
Avoid food and drink 15
min before or while
using lozenge.
Sustained release
product; do not crush or
chew.
Drug Interactions
Nicotine itself is not subject to cytochrome P-450 interactions. Tobacco smoke however leads to potent
induction of CYP1A1 and 1A2. When smoking is discontinued, the substrate drug may require a dosage
decrease over a period of several days. CYP1A1, 1A2 substrates include: theophylline, clozapine,
olanzapine, fluvoxamine, TCAs (partial substrate).
Inhibits CYP2D6, 2B6
substrate, avoid with
MAOI
Increased adverse
effects if combined with
NRT
Contraindications/Cauti
ons
Life-threatening
arrhythmias, severe
angina,
atopic/eczematous
dermatitis or other skin
conditions (e.g.
psoriasis)
Seizure disorder,
anorexia, bulimia, use
of MAOI in 14 days,
patients undergoing
abrupt discontinuation
of alcohol, sedatives
and benzodiazepines
Depression, suicidal
ideation, schizophrenia,
bipolar other major
depressive disorders
Fourth Edition FINAL
Life-threatening
arrhythmias, severe
angina
Life-threatening
arrhythmias, severe
angina
Update: September 2012
Life-threatening
arrhythmias, severe
angina
Page 57 of 69
CBPRSC 2012
Use in Special
Populations
Combination Therapy?
Section 2: Prevention
2.9 Management of Smoking Cessation
Nicotine patch
Nicotine gum
Nicotine inhaler
Nicotine Lozenge
Cardiovascular/Stroke Patients: Demonstrated safety in stable cardiovascular disease (possible
exceptions are unstable angina, recent MI, unstable arrhythmia, acute heart failure). Commonly
used in many inpatient settings as symptoms of nicotine withdrawal can begin within 1 hour. It is
considered by many experts as far safer than continued smoking.
Pregnancy/Breastfeeding/Adolescents: While data are limited in pediatrics and
pregnant/breastfeeding women, NRT is generally considered safer than smoking in these
populations and should be considered. Offer the lowest effective dose of a short-acting nicotine
product to minimize nicotine exposure.
Can use with oral
agents, gum, inhaler or
lozenges. Evidence
suggests better
abstinence rates with
combination over
monotherapy.
Can use with oral agents or patch. Evidence suggests better abstinence rates
with combination over monotherapy.
Partially replaces nicotine delivered by cigarettes
Mechanism of Action
Approximate $ per
month
Fourth Edition FINAL
$100
$75-200
(6-20 pieces/d)
$175- 350
(6-12 cartridges/d)
Update: September 2012
$100-250
(6-12 lozenges/d)
Bupropion
May be used in
pregnant women,
especially those with
depression. May be
considered in
adolescents or
breastfeeding women.
Requires dose
adjustment in
renal/hepatic disease.
Varenicline
Data not available in
pregnancy/lactation.
May be considered in
adolescents. Requires
dose adjustment in
renal disease (if CrCl
<30mL/min, max 0.5
mg BID).
Can use with
varenicline or NRT
(nicotine replacement
therapy). Addition of
patch significantly
increases long term
cessation compared
with patch alone.
Monitor for treatment
emergent hypertension
when NRT is combined
with bupropion.
Not fully understood.
Likely due to inhibition
of dopamine and
norepinephrine uptake.
Can use with bupropion
or NRT (although
increased adverse
effects with NRT).
$60
Partial agonist at
nicotinic acetylcholine
receptor, causing
decreased dopamine
release and activation
of mesolimbic reward
system.
$125
Page 58 of 69
CBPRSC 2012
Section 2: Prevention
References
CANADIAN BEST PRACTICE RECOMMENDATIONS FOR STROKE CARE
CHAPTER 2: PREVENTION OF STROKE
REFERENCES
1.
Health promotion glossary. Geneva: World Health Organization; 1998: page 4.
Available: http://www.who.int/healthpromotion/about/HPR%20Glossary%201998.pdf
2.
Goldstein LB, Bushnell CD, Adams RJ, Appel LJ, Braun LT, Chauturvedi S, Creager MA, Culebras A, Eckel RH, Hart RG, Hinchey JA, Howard VJ,
Jauch EC, Levine SR, Meschia JF, Moore WS. Guidelines for the primary prevention of stroke: a guideline for healthcare professionals from the
American Heart Association/American Stroke Association. Stroke. 2011 February; 42(2): 517-584.
3.
Henney JE, Taylor CL, and Boon CS, Editors. Strategies to reduce sodium intake in the United States. Institute of Medicine of the National
Academies 2010; Washington DC: National Academies Press.
4.
Canadian Society for Exercise Physiology. Canadian physical activity guidelines. 2012 Scientific Statements. Accessed at
/www.csep.ca/CMFiles/Guidelines/CanadianPhysicalActivityGuidelinesStatements_E_2012.pdf
5.
Kokubo Y. Traditional risk factor management for stroke: a never-ending challenge for health behaviors of diet and physical activity. Curr Opin
Neurol, 2012;25:11-17.
6.
Gillman MW, Cupples LA, Gagnon D, et al. Protective effect of fruits and vegetables on development of stroke in men. JAMA 1995;273:1113-7.
7.
He FJ, Nowson CA, MacGregor GA. Fruit and vegetable consumption and stroke: meta-analysis of cohort studies. Lancet 2006;367:320-6.
8.
Li Y, He Y, Lai J, Wang D, Zhang J, Fu P, Yang X, Qi L. Dietary patterns are associated with stroke in Chinese adults. J Nutr, 2011;141:1834-1839.
9.
Mahe G, Ronziere T, Laviolle B, Golfier V, Cochery T, De Bray J-M, Paillard F. An unfavorable dietary pattern in associated with symptomatic
ischemic stroke and carotid atherosclerosis. J Vasc Surg, 2010;52:62-68.
10.
Agnoli C, Krogh V, Grioni S, Sieri S, Palli D, Masala G, Sacerdote C, Vineis P, Tumino R, Frasca G, Pala V, Berrino F, Chiodini P, Mattiello A,
Panico S. A priori-defined dietary patterns are associated with reduced risk of stroke in a large Italian cohort. J Nutr, 2011;141:1552-1558.
11.
Joshipura KJ, Ascherio A, Manson JE, et al. Fruit and vegetable intake in relation to risk of ischemic stroke. JAMA 1999;282:1233-9.
12.
Liu S, Manson JE, Lee IM, et al. Fruit and vegetable intake and risk of cardiovascular disease: the Women’s Health Study. Am J Clin Nutr
2000;72:922-8.
13.
Alderman MH. The Cochrane review of sodium and health. Am J Hypertens, 2011;24(8):854-856.
14.
Joffres MR, Campbell NRC, Manns B, Tu K. Estimate of the benefits of a population-based reduction in dietary sodium additives on hypertension
and its related health care costs in Canada. Canadian Journal of Cardiology. 2007 May; 23(1): 437-443.
15.
Penz ED, Joffres MD, Campbell NRC. Reducing dietary sodium and decreases in cardiovascular disease in Canada. Canadian Journal of
Cardiology 2008; 24(6): 497-501.
16.
Graudal NA, Hubeck-Graudal T, Jurgens G. Effects of low sodium diet versus high sodium diet on blood pressure, renin, aldosterone,
catecholamines, cholesterol, and triglyceride. Cochrane Database of Systematic Reviews 2011, Issue 11. Art.No.: CD004022.
17.
Gardener H, Rundek T, Wright CB, Elkind MSB, Sacco, RL. Dietary Sodium and Risk of Stroke in the Northern Manhattan Study. Stroke.
2012;43:1200- 1205.)
18.
He FJ, Appel LJ, Cappucio FP, de Wardener HE, MacGregor GA. Does reducing salt intake increase cardiovascular mortality? Kidney
International. 2011 August; doi: 10.1038/ki.2011.246.
19.
He FJ, MacGregor GA. Salt reduction lowers cardiovascular risk: meta-analysis of outcome trials. The Lancet. 2011 July; Vol. 378: p.380-381.
20.
Frohlich ED. The Salt Conundrum: A Hypothesis. Hypertension, 2007;50:161-166,
21.
Aleksandrova K, Pischon T, Weikart C. Urinary Sodium Excretion and Cardiovascular Mortality. The Journal of the American Medical Association
(JAMA). 2011 September; 306(10).
22.
Strazzullo P, D'Elia L, Kandala NB, Cappuccio FP. Salt intake, stroke, and cardiovascular disease: meta-analysis of prospective studies. BMJ.
2009 Nov 24;339:b4567. doi: 10.1136/bmj.b4567.
23.
Appel L, Frohlich ED, Hall JE, Pearson TA, Sacco RL, Seals DR, Sacks FM, et al. the Importance of Population-Wide Sodium Reduction as a
Means to Prevent Cardiovascular Disease and Stroke. Circulation, 2011; 123: 1138-1143.
24.
Saito I, Iso H, Kokubo Y, Inoue M, Tsugane S. Body mass index, weight change and risk of stroke and stroke subtypes: the Japan public health
center-based prospective (JPHC) study. International Journal of Obesity, 2011;35:283-291.
25.
Bazzano LA, Gu D, Whelton MR, Wu X, Chen C-S, Duan X, Chen J, Chen J-c, He j. Body mass index and risk of stroke among Chinese men and
women. Ann Neurol, 2010;67:11-20.
26.
Yatsuya H, Toyoshima H, Yamagishi K, Tamakoshi K, Taguri M, Harada A, Ohashi Y, Kita Y, Naito Y, Yamada M, Tanabe N, Iso H, Ueshima H, for
the Japan Arteriosclerosis Longitudinal Study (JALS) Group. Body mass index and risk of stroke and myocardial infarction in a relatively lean
population. Circ Cardiovasc Qual Outcomes, 2010;3:498-505.
27.
Hu G, Tuomilehto J, Silventoinen K, Sarti C, Mannisto S, Jousilahti P. Body mass index, waist circumference, and waist-hip ratio on the risk of total
and type-specific stroke. Arch Intern Med, 2007;167(13):1420-1427.
Fourth Edition FINAL
Update: September 2012
Page 59 of 69
CBPRSC 2012
Section 2: Prevention
References
28.
Winter Y, Rohrmann S, Linseisen J, Lanczik O, Ringleb PA, Hebebrand J, Back T. Contribution of obesity and abdominal fat mass to risk of stroke
and transient ischemic attacks. Stroke, 2008;39:3145-3151.
29.
Bodenant M, Kuulasmaa K, Wagner A, Kee F, Palmieri L, Ferrario MM, Montaye M, Amouyel P, Dallongeville J, for the MORGAM project.
Measures of abdominal adiposity and the risk of stroke. The monica risk, genetics, archiving and monograph (MORGAM) study. Stroke,
2011;42:2872-2877.
30.
Dalton M, Cameron AJ, Zimmet PZ, Shaw JE, Jolley D, Dunstan DW, Welborn TA, on behalf of the AUSDIAB Steering Committee. Waist
circumference, waist-hip ration and body mass index and their correlation with cardiovascular disease risk factors in Australian adults. Journal of
Internal Medicine, 2003;254:555-563.
31.
Janssen I, Katzmarzyk PT, Ross R. Waist circumference and not body mass index explains obesity-related health risk. Am J Clin Nutr,
2004;79:379-384.
32.
Yau WY, Hankey GJ. Which dietary and lifestyle behaviours may be important in the aetiology (and prevention) of stroke? Journal of Clinical
Neuroscience, 2011;18:76-80.
33.
Zhu S, Heshka S, Wang Z, Shen W, Allison DB, Ross R, Heymsfield SB. Combination of BMI and waist circumference for identifying
cardiovascular risk factors in whites. Obes Res, 2004;12:633-645.
34.
Clark VR, Greenberg B, Harris TS, Carson BL. Body Mass Index And Waist Circumference Predictors Of Cardiovascular Risk In African
Americans . Ethnicity & Disease, Volume 22, Spring 2012
35.
Reimers CD, Knapp G, Reimers AK. Exercise as stroke prophylaxis. Dtsch Arztbl Int, 2009;106(44):715-721.
36.
Lee CD, Folsom AR, Blair SN. Physical activity and stroke risk: a meta-analysis. Stroke 2003;34:2475-81.
37.
Jurkiewicz MT, Marzolini S, Oh P. Adherence to a home-based exercise program for individuals after stroke. Top Stroke Rehabil, 2011;18(3):277284.
38.
Reynolds K, Lewis B, Nolen JD, et al. Alcohol consumption and risk of stroke: a meta-analysis. JAMA, 2003;298:579-588
39.
Mazzaglia G, Britton AR, Altmann DR, et al. Exploring the relationship between alcohol consumption and non-fatal or fatal stroke: a systematic
review. Addiction, 2001;96:1743-1756.
40.
Truelsen T, Gronbaek M, Schnohr P, et al. Intake of beer, wine, and spirits and risk of stroke: the Copenhagen city heart study. Stroke
1998;29:2467
41.
Kiechl S, Willeit J, Rungger G, et al. Alcohol consumption and atherosclerosis: What is the relation? Prospective results from the Bruneck Study.
Stroke 1998;29:900
42.
Sacco RL. Identifying patient populations at high risk of stroke. Neurology 1998;51(3 Suppl):S27
43.
Lidegaard O, Lokkegaard E, Jensen A, Skovlund CW, Keiding N. Thrombotic stroke and myocardial infarction with hormonal contraception. N Engl
J Med, 2012;366:2257-2266.
44.
Lidegaard O, Nielsen LH, Skovlund CW, Skjeldestad FE, Lokkegaard E. Risk of venous thromboembolism from use of oral contraceptives
containing different progesterones and oestrogen doses: Danish cohort study, 2001-9. BMJ, 2011;343:d6423.
45.
Lidegaard OL, Nielsen LH, Wessel C, Lokkegaard E. Venous thrombosis in users of non-oral hormonal contraception: follow-up study, Denmark
2001-10. BMJ, 2012;344:e2990.
46.
Bath PMW, Gray LJ. Association between hormone replacement therapy and subsequent stroke: a meta-analysis. BMJ, 2005;doi:10.1136
47.
Renoux C, Dell’Aniello S, Garbe E, Suissa S. Transdermal and oral hormone replacement therapy and the risk of stroke: a nested case-control
study. BMJ, 2010;340:2519
48.
Cole JA, Norman H, Doherty M, Walker AM. Venous thromboembolism, myocardial infarction, and stroke among transdermal contraceptive system
users. Obstet Gynecol, 2007;109:339-346.
49.
Yang L, Kuper H, Sandin S, Margolis KL, Chen Z, Adami H-O, Weiderpass E. Reproductive history, oral contraceptive use, and the risk of ischemic
and hemorrhagic stroke in a cohort of middle-aged Swedish women. Stroke, 2009;40:1050-1058.
50.
Hypertension Canada. Canadian hypertension education program. Available: http://hypertension.ca/chep/.
51.
Campbell NRC, PoirierL, Tremblay G, Lindsay P, et al. Canadian Hypertension Education Program: The Science Supporting New 2011 CHEP
Recommendations With an Emphasis on Health Advocacy and Knowledge Translation. Can J Cardiol 2011;27:407-414.
52.
Launer LJ, Masaki K, Petrovitch H, et al. The association between midlife blood pressure levels and late-life cognitive function. The Honolulu–Asia
Aging Study. JAMA 1995;274:1846-51.
53.
Tu K, Chen Z, Lipscombe L. Prevalence and incidence of hypertension from 1995–2005: a population-based study. CMAJ 2008;178:1429-35.
54.
O’Donnell, MJ., Denis, X., Liu, L., Zhang, H., Chin, SL., Rao-Melacini, P., Rangarajan, S., Islam, S., Pais, P., McQueen, MJ., Mondo, C.,
Damasceno, A., Lopez-Jaramillo, P., Hankey, GJ., Dans, AL., Yusoff, K., Truelsen, T., Diener, HC., Sacco, RL., Ryglewicz, D., Czlonkowska, A.,
Wang, X. & Yusuf, S. Risk factors for ischaemic and intracerebral haemorrahagic stroke in 22 countries (the INTERSTROKE study): A case-control
study. The Lancet, 2010; 376(9735): 112-123.
55.
Du X, McNamee R, Cruickshank K. Stroke risk from multiple risk factors combined with hypertension: a primary care based case-control study in a
defined population of northwest England. Ann Epidemiol 2000;10:380-8.
56.
Chobanian AV, Bakris GL, Black HR, et al. The Seventh Report of the Joint National Committee on Prevention, Detection, Evaluation, and
Treatment of High Blood Pressure: the JNC 7 report. JAMA 2003;289:2560-72.
Fourth Edition FINAL
Update: September 2012
Page 60 of 69
CBPRSC 2012
Section 2: Prevention
References
57.
Lewington S, Clarke R, Qizilbash N, et al.; Prospective Studies Collaboration. Age-specific relevance of usual blood pressure to vascular mortality:
a meta-analysis of individual data for one million adults in 61 prospective studies. Lancet 2002; 360:1903-13.
58.
Weber MA. Managing the patient at risk for a second stroke. J Hypertens Suppl 2005;23(S1):S41-S47
59.
Bestehorn, K., Wahle, K. & Kirch, W. Stroke risk screening of adults with hypertension: Prospective cross-sectional study in primary care. Clinical
Drug Investigation, 2008; 28(5): 281-289.
60.
Girerd, X. & Giral, P. Risk stratification for the prevention of cardiovascular complications of hypertension. Current Medical Research and Opinion,
2004; 20(7): 1137-1142.
61.
Lee M, Saver JL, Chang B, et al. Presence of baseline prehypertension and risk of incident stroke: a meta-analysis. Neurology, 2011;77:13301337.
62.
Ovbiagele B, Diener H-C, Yusuf S, Martin RH, Cotton D, Vinisko R, Donnan GA, Bath PM, for the PROFESS Investigators. Level of systolic blood
pressure within the normal range and risk of recurrent stroke. JAMA, 2011;306(19):2137-2144.
63.
Musini, VM., Tejani, AM., Bassett, K. & Wright, JM. Pharmacotherapy for hypertension in the elderly. Cochrane Database of Systematic Reviews
2009, Issue 4: CD000028.
64.
Goldstein LB, Adams R, Alberts MJ, et al. Primary prevention of ischemic stroke. A guideline from the American Heart Association/American Stroke
Association Stroke Council. Stroke 2006;37:1583-633.
65.
Gueyffier F, Boissel JP, Boutitie F, et al. Effect of antihypertensive treatment in patients having already suffered from stroke. Gathering the
evidence. The INDANA (INdividual Data ANalysis of Antihypertensive intervention trials) Project Collaborators. Stroke 1997;28:2557-62.
66.
Rodgers A, MacMahon S, Gamble G, et al. Blood pressure and risk of stroke in patients with cerebrovascular disease. The United Kingdom
Transient Ischaemic Attack Collaborative Group. BMJ 1996;313:147.
67.
Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Effects of ramipril on cardiovascular and microvascular outcomes in people
with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Lancet 2000; 355:253-9.
68.
PROGRESS Collaborative Group. Randomised trial of a perindopril-based bloodpressure- lowering regimen among 6,105 individuals with previous
stroke or transient ischaemic attack. Lancet 2001;358:1033-41.
69.
Dahlöf B, Devereux R, de Faire U, et al. The Losartan Intervention For Endpoint reduction (LIFE) in Hypertension study: rationale, design, and
methods. The LIFE Study Group. Am J Hypertens 1997;10:705-13.
70.
Schrader J, Lüders S, Kulschewski A, et al. The ACCESS Study: evaluation of Acute Candesartan Cilexetil Therapy in Stroke Survivors. Stroke
2003;34: 1699-703.
71.
Lithell H, Hansson L, Skoog I, et al. The Study on Cognition and Prognosis in the Elderly (SCOPE): principal results of a randomized double-blind
intervention trial. J Hypertens 2003;21:875-86.
72.
Yusuf S, Teo KK, Pogue J, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med 2008;358:1547-59.
73.
Beckett N, Peters R, Tuomilehto J, Swift C, Sever P, Potter J, McCormack T, Forette F, Gil-Extremera B, Dumitrascu S, Staessan JA, Thijs L,
Bulpitt C, for the HYVET Study Group. Immediate and late benefits of treating very elderly people with hypertension: results from active treatment
extension to hypertension in the very elderly randomised controlled trial. BMJ, 2012:344:d7541.
74.
Bulpitt CJ, Beckett NS, Peters R, Leonetti G, Gergova V, Fagard R, Burch LA, Banya W, G, Fletcher AE. Blood pressure control in the
hypertension in the very elderly trial (HYVET). Journal of Human Hypertension, 2012;26:157-163.
75.
Beckett NS, Peters R, Fletcher AE, et al. Treatment of hypertension in patients 80 years of age and older. N Engl J Med 2008;358:1887-98.
76.
Launer LJ, Masaki K, Petrovitch H, et al. The association between midlife blood pressure levels and late-life cognitive function. The Honolulu–Asia
Aging Study. JAMA 1995;274:1846-51.
77.
Cholesterol Treatment Trialists’ (CTT) Collaborators. Efficacy of cholesterol-lowering therapy in 18 686 people with diabetes in 14 randomised trials
of statins: a meta-analysis. The Lancet 2008; 371(9607): 117-125.
78.
Genest J, McPherson R, Frohlich J, Anderson T, et al.. 2009 Canadian Cardiovascular Society Canadian guidelines for the diagnosis and
treatment of dyslipidemia and prevention of cardiovascular disease in the adult – 2009 recommendations. Canadian Journal of Cardiology, 2009;
25 (10):567-579.
79.
Amarenco, P. & Labreuche, J. Lipid management in the prevention of stroke: review and updated meta-analysis of statins for stroke prevention.
The Lancet Neurology 2009; 8(5): 453-463.
80.
O’Regan C, Wu P, Arora P, et al. Statin therapy in stroke prevention: a metaanalysis involving 121 000 patients. Am J Med 2008;121:24-33.
81.
Corvol JC, Bouzamondo A, Sirol M, et al. Differential effects of lipid-lowering therapies on stroke prevention: a meta-analysis of randomized trials.
Arch Intern Med 2003;163:669-76.
82.
Collins R, Armitage J, Parish S, et al. Effects of cholesterol-lowering with simvastatin on stroke and other major vascular events in 20536 people
with cerebrovascular disease or other high-risk conditions. Lancet 2004;363:757-67.
83.
Amarenco P, Bogousslavsky J, Callahan A 3rd, et al.; Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) Investigators.
High-dose atorvastatin after stroke or transient ischemic attack. N Engl J Med 2006;355:549-59.
84.
Amarenco P, Labreuche J, Lavallee P, et al. Statins in stroke prevention and carotid atherosclerosis: systematic review and up-to-date metaanalysis. Stroke 2004;35:2902-9.
85.
Goldstein LB, Amarenco P, Szarek M, et al. Hemorrhagic stroke in the Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL)
study. Neurology 2008;70: 2364-70.
86.
European Stroke Organization (ESO) Executive Committee; ESO Writing Committee. Guidelines for management of ischaemic stroke and
transient ischaemic attack 2008. Cerebrovasc Dis 2008;25:457-507.
Fourth Edition FINAL
Update: September 2012
Page 61 of 69
CBPRSC 2012
Section 2: Prevention
References
87.
Idris I, Thomson GA, Sharma JC. Diabetes mellitus and stroke. Int J Clin Pract 2006;60:48-56.
88.
Karapanayiotides T, Piechowski-Jozwiak B, van Melle G, et al. Stroke patterns, etiology, and prognosis in patients with diabetes mellitus.
Neurology 2004;62: 1558-62.
89.
Lehto S, Rönnemaa T, Pyörälä K, et al. Predictors of stroke in middle-aged patients with non-insulin-dependent diabetes. Stroke 1996;27:63-8.
90.
Shepherd J, Barter P, Carmena R, et al. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with
coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006; 29:1220-6.
91.
Gerstein HC, Miller ME, et al. Effects of intensive glucose lowering in type 2 diabetes. (ACCORD). N Engl J Med 2008;258:2545-59.
92.
ADVANCE Collaborative Group. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med
2008;358:2560-72.
93.
Hirsh J, Guyatt G, Albers GW, Harrington R, Schunemann HJ, American College of Chest Physicians. Executive summary: American College of
Chest Physicians evidence-based clinical practice guidelines (8th edition). Chest 2008; 133(6 Suppl): 71S-109S.
94.
Soman, T., Rafay, MF., Hune, S., Allen, A., MacGregegor, D. & deVeber, G. The risks and safety of clopidogrel in pediatric arterial ischemic stroke.
Stroke 2006; 37: 1120-1122.
95.
Yip S, Benavente O. Antiplatelet agents for stroke prevention. Neurotherapeutics, 2011;8:475-487.
96.
Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial
infarction, and stroke in high risk patients. BMJ 2002;324:71-86.
97.
Schwartz NE, Albers GW. Is there a role for combinations of antiplatelet agents in stroke prevention? Curr Treat Options Neurol 2007;9:442-50.
98.
O’Donnell MJ, Hankey GJ, Eikelboom JW. Antiplatelet therapy for secondary prevention of noncardioembolic ischemic stroke: a critical review.
Stroke 2008;39: 1638-46.
99.
Verro P, Gorelick PB, Nguyen D. Aspirin plus dipyridamole versus aspirin for prevention of vascular events after stroke or TIA: a meta-analysis.
Stroke 2008;39: 1358-63.
100.
Hankey GJ. Antiplatelet therapy for the prevention of recurrent stroke and other serious vascular events: a review of the clinical trial data and
guidelines. Curr Med Res Opin 2007;23:1453-62.
101.
De Schryver ELLM, Algra A, van Gijn J. Dipyridamole for preventing stroke and other vascular events in patients with vascular disease. Cochrane
Database Syst Rev 2007;(3):CD001820.
102.
Diener HC, Cunha L, Forbes C, et al. European Stroke Prevention Study. 2. Dipyridamole and acetylsalicylic acid in the secondary prevention of
stroke. J Neurol Sci 1996;143:1-13.
103.
Hankey GJ, Sudlow CLM, Dunbabin DW. Thienopyridine derivatives (ticlopidine, clopidogrel) versus aspirin for preventing stroke and other serious
vascular events in high vascular risk patients. Cochrane Database Syst Rev 2000;(1):CD001246.
104.
Halkes, PHA., Gray, LJ., Bath, PMW., Diener, H-C., Guiraud-Chaumeil, B., Yatsu, FM., & Algra, A. Dipyridamole plus aspirin versus aspirin alone
in secondary prevention after TIA or stroke: a meta-analysis by risk. J Neurol Neurosurg Psychiatry 2008; 79:1218-1223.
105.
Diener HC, Sacco R, Yusuf S; Steering Committee; PRoFESS Study Group. Rationale, design and baseline data of a randomized, double-blind,
controlled trial comparing two antithrombotic regimens (a fixed-dose combination of extended-release dipyridamole plus ASA with clopidogrel) and
telmisartan versus placebo in patients with strokes: the Prevention Regimen for Effectively Avoiding Second Strokes Trial (PRoFESS). [published
erratum in Cerebrovasc Dis 2008;25:192]. Cerebrovasc Dis 2007;23:368-80.
106.
ESPRIT Study Group, Halkes PH, van Gijn J, et al. Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial origin
(ESPRIT): randomised controlled trial. Lancet 2006;367:1665-73.
107.
Bhatt DL, Fox KA, Hacke W, et al. Clopidogrel and aspirin versus aspirin alone for the prevention of atherothrombotic events. N Engl J Med
2006;354:1706-17.
108.
Diener HC, Bogousslavsky J, Brass LM, et al. Aspirin and clopidogrel compared with clopidogrel alone after recent ischaemic stroke or transient
ischaemic attack in high-risk patients (MATCH): randomised, double-blind, placebo-controlled trial. Lancet 2004;364:331-7.
109.
CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). Lancet
1996;348:1329-39.
110.
The SPS3 Investigators. Effects of Clopidogrel Added to Aspirin in Patients with Recent Lacunar Stroke. New England Journal of Medicine. 2012
August; 367:817.
111.
Paediatric Stroke Working Group. Stroke in childhood: clinical guidelines for diagnosis, management and rehabilitation. London (UK): Royal
College of Physicians of London; 2004. Available: www.rcplondon.ac.uk/pubs/books/childstroke/childstroke_guidelines.pdf
112.
Lanthier S, Kirkham FJ, Mitchell LG, et al. Increased anticardiolipin antibody IgG titers do not predict recurrent stroke or TIA in children. Neurology
2004;62:194-200.
113.
Roach ES, Golomb MR, Adams R, et al. Management of stroke in infants and children: a scientific statement from a special writing group of the
American Heart Association Stroke Council and the Council on Cardiovascular Disease in the Young. Stroke 2008;39:2644
114.
Soman T, Rafay MF, Hune S, Allen A, MacGregor D, deVeber G.
The risks and safety of clopidogrel in pediatric arterial ischemic stroke. Stroke. 2006 Apr;37(4):1120-2.
115.
Mertens L, Eyskens B, Boshoff D, Gewillig M. Safety and efficacy of clopidogrel in children with heart disease. J Pediatr, 2008;153:61-64.
116.
Li JS, Yow E, Berezny KY, Bokesch PM, Takahashi M, Graham TP, Sanders SP, Sidi D, Bonnet D, Ewert P, Jennings LK, Michelson AD. Dosing
of clopidogrel for platelet inhibition in infants and young children: primary results of the platelet inhibition in children on clopidogrel (PICOLO) trial.
Circulation, 2008;117:553-559.
Fourth Edition FINAL
Update: September 2012
Page 62 of 69
CBPRSC 2012
Section 2: Prevention
References
117.
Skanes AC, Healey JS, Cairns JA, et al. Focused 2012 Update of the Canadian Cardiovascular Society Atrial Fibrillation Guidelines:
Recommendations for Stroke Prevention and Rate/Rhythm Control. Canadian Journal of Cardiology, 2012;28:125-136.
118.
Gage, BF., Waterman, AD., Shannon, W., Boechler, M., Rich, MW. & Radfor, MJ. Validation of clinical classification schemes for predicting stroke:
results from the national registry of atrial fibrillation. Journal of the American Medical Association 2001; 285(22): 2864-2870.
119.
Lip GY, Halperin JL. Improving stroke risk stratification in atrial fibrillation. American Journal of Medicine, 2010: 123(6): 844-848.
120.
Hobbs FDR, Fitzmaurice DA, Mant J, et al. A randomized controlled trial and cost-effectiveness study of systematic screening (targeted and total
population screening) versus routine practice for the detection of atrial fibrillation in people aged 65 and over. The SAFE study. Health Technology
Assessment. 2005; 9:1-74.
121.
Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham study. Stroke. 1991; 22: 983-88.
122.
Kimura K, Minematsu K, Yamaguchi T, for the Japan Multicenter Stroke Investigators’ Collaboration: Atrial fibrillation as a predictive factor for
severe stroke and early death in 15,831 patients with acute ischemic stroke. J Neurol Neurosurg Psychiatry 2005; 76: 679– 683.
123.
Gladstone D, et al. Potentially preventable strokes in high-risk patients with atrial fibrillation who are not adequately anticoagulated. Stroke. 2009;
40: 235-40.
124.
Connolly, SJ., Ezekowitz, MD., Yusuf, S., et al. And the RE-LY Steering Committee and Investigators. Dabigatran versus Warfarin in patients with
atrial fibrillation. NEJM 2009; 361: 1139-1151.
125.
Patel, et.al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. New England Journal of Medicine, 2011; 365:883-891.
126.
Granger et. Al. Apixaban versus warfarin in patients with atrial fibrillation. New England Journal of Medicine, 2011; 365(11): 981-992.
127.
Hylek EM, Go AS, Chang Y, et al. Effect of intensity of oral anticoagulation on stroke severity and mortality in atrial fibrillation. N Engl J Med.
2003; 349: 1019-26.
128.
Gage BF. Can we rely on RE-LY? N Engl J Med. 2009; 361: 1200-2.
129.
Birman-Deych E, Radford MJ, Nilasena DS, Gage BF. Use and effectiveness of warfarin in Medicare beneficiaries with atrial fibrillation. Stroke.
2006; 37: 1070-4.
130.
Hankey, GJ. At last, a RE-LYable alternative to warfarin for atrial fibrillation. International Journal of Stroke 2009; 4: 454-455.
131.
Connolly SJ, Eikelboom J, Joyner C, Diener HC, Hart R, Golitsyn S, Flaker G, Avezum A, Hohnloser SH, Diaz R, Talajic M, Zhu J, Pais P, Budaj A,
Parkhomenko A, Jansky P, Commerford P, Tan RS, Sim KH, Lewis BS, Van Miegham W, Lip GY, Kim JH, Lanas-Zanetti F, Gonzalez-Hermosillo
A, Dans AL, Munawar M, O’Donnell M, Lawrence J, Lewis G, Afzal R, Yusuf S, AVERROES Steering Committee and Investigators. Apixaban in
patients with atrial fibrillation. The New England Journal of Medicine, 2011; 364(9):806-817.
132.
Hart RG, Pearce LA, Aguilar MI. Meta-analysis: antithrombotic therapy to prevent stroke in patients who have nonvalvular atrial fibrillation. Ann
Intern Med 2007; 146:857-67.
133.
Connolly S, Pogue J, Hart R, et al.; ACTIVE Writing Group of the ACTIVE Investigators. Clopidogrel plus aspirin versus oral anticoagulation for
atrial fibrillation in the Atrial fibrillation Clopidogrel Trial with Irbesartan for prevention of Vascular Events (ACTIVE W): a randomised controlled trial.
Lancet 2006;367: 1903-12.
134.
ACTIVE Investigators. Effect of clopidogrel added to aspirin in atrial fibrillation. N Engl J Med. 2009; 360: 2066-78.
135.
Mant J, Hobbs R, Fletcher K, et al.; BAFTA Investigators; Midland Research Practices Network (MidReC). Warfarin versus aspirin for stroke
prevention in an elderly community population with atrial fibrillation (the Birmingham Atrial Fibrillation Treatment of the Aged Study, BAFTA): a
randomised controlled trial. Lancet 2007;370:493-503.
136.
EAFT (European Atrial Fibrillation Trial) Study Group. Secondary prevention in non-rheumatic atrial fibrillation after transient ischemic attack or
minor stroke. Lancet 1993;342:1255-62.
137.
Morocutti C, Amabile G, Fattapposta F, et al. Indobufen versus warfarin in the secondary prevention of major vascular events in nonrheumatic atrial
fibrillation. SIFA (Studio Italiano Fibrillazione Atriale) Investigators. Stroke 1997;28:1015-21
138.
Saxena R., Koudstaal PJ. Anticoagulants versus antiplatelet therapy for preventing stroke in patients with nonrheumatic atrial fibrillation and a
history of stroke or transient ischemic attack. Cochrane Database of Systematic Reviews 2004, Issue 4. Art. No.: CD000187.
139.
Berge E, Sandercock PAG. Anticoagulants versus antiplatelet agents for acute ischaemic stroke. Cochrane Database of Systematic Reviews 2002,
Issue 4. Art. No.: CD003242. DOI: 10.1002/14651858.CD003242.
140.
Hansen, ML., Sorensen, R., Clausen, MT., Fog-Petersen, ML., Raunso, J., Gladsboll, N., Gislason, GH., Folke, F., Anderson, SS., Schramm, TK.,
Abildstrom, SZ., Poulsen, HE., Kober, L. & Torp-Pedersen, C. Risk of bleeding with single, dual, or triple therapy with warfarin, aspirin, and
clopidogrel in patients with atrial fibrillation. Arch Intern Med 2010; 170(16): 1433-1441.
141.
Halliday A, Mansfield A, Marro J, Peto C, Peto R, Potter J, Thomas D. Prevention of disabling and fatal strokes by successful carotid
endarterectomy in patients without recent neurological symptoms: randomised controlled trial. Lancet. 2004; 363: 1491–1502.
142.
Halliday A, Harrison M, Hayter E, Kong X, Mansfield A, Marro J, Pan H, Peto R, Potter J, Rahimi K, Rau A, Robertson S, Streifler J, Thomas D;
Asymptomatic Carotid Surgery Trial (ACST) Collaborative Group.10-year stroke prevention after successful carotid endarterectomy for
asymptomatic stenosis (ACST-1): a multicentre randomised trial. Lancet. 2010 Sep 25;376(9746):1074-84.
143.
Chimowitz MI, Lynn MJ, Derdeyn CP, et al. Stenting versus aggressive medical therapy for intracranial arterial stenosis. N Engl J Med,
2011;365:993-1003.
144.
Ferguson GG, Eliasziw M, Barr HWK, et al.; North American Symptomatic Carotid Endarterectomy Trial (NASCET) Collaborators. The North
American Symptomatic Carotid Endarterectomy Trial: surgical results in 1415 patients. Stroke 1999;30:1751-8.
Fourth Edition FINAL
Update: September 2012
Page 63 of 69
CBPRSC 2012
Section 2: Prevention
References
145.
Randomised trial of endartertectomy for recently symptomatic carotid stenosis: final results of the MRC European Carotid Surgery Trial (ECST).
Lancet 1998; 351:1379-87.
146.
Mayberg MR, Wilson SE, Yatsu F, et al. Veterans Affairs Cooperative Studies Program 309 Trialist Group. Carotid endarterectomy and prevention
of cerebral ischemia in symptomatic carotid stenosis. JAMA 1991;266:3289-94.
147.
Rothwell PM, Eliasziw M, Gutnikov SA, et al.; Carotid Endarterectomy Trialists’ Collaboration. Analysis of pooled data from the randomised
controlled trials of endarterectomy for symptomatic carotid stenosis. Lancet 2003;361:107-16.
148.
Biller J, Feinberg WM, Castaldo JE, et al. Guidelines for carotid endarterectomy: a statement for healthcare professionals from a special writing
group of the Stroke Council, American Heart Association. Circulation 1998;97:501-9.
149.
Findlay JM, Tucker WS, Ferguson RO, et al. Guidelines for the use of carotid endarterectomy: current recommendations from the Canadian
Neurosurgical Society. CMAJ 1997;157:653-9.
150.
Bond R, Rerkasem K, Rothwell PM. Systematic review of the risks of carotid endarterectomy in relation to the clinical indication for and timing of
surgery. Stroke 2003;34:2290-301.
151.
Gladstone D, Oh J, Fang J, Lindsay P, Tu J, Silver F and Kapral M. Urgency of carotid endarterectmy for secondary stroke prevention results
from the Registry of the Canadian Stroke Network. Stroke 2009;40;8;2776
152.
Chaturvedi S, Bruno A, Feasby T. Carotid endarterectomy — an evidence-based review: report of the Therapeutics and Technology Assessment
Subcommittee of the American Academy of Neurology. Neurology 2005;65:794-801.
153.
Alamowitch S, Eliasziw M, Barnett HJM; North American Symptomatic Carotid Endarterectomy Trial (NASCET); ASA Trial Group; Carotid
Endarterectomy (ACE) Trial Group. The risk and benefit of endarterectomy in women with symptomatic internal carotid artery disease. Stroke
2005;36:27-31.
154.
Rothwell PM, Eliasziw M, Gutnikov SA, et al.; Carotid Endarterectomy Trialists’ Collaboration. Endarterectomy for symptomatic carotid stenosis in
relation to clinical subgroups and timing of surgery. Lancet 2004;363:915-24.
155.
Asymptomatic Carotid Atherosclerosis Study (ACAS) Group. Endarterectomy for asymptomatic internal carotid artery stenosis. JAMA
1995;273:1421-8.
156.
Mackey AE, Abrahamowicz M, Langlois Y, et al. Outcome of asymptomatic patients with carotid disease. Asymptomatic Cervical Bruit Study
Group. Neurology 1997;48:896-903.
157.
Gladstone DJ, Sahlas DJ. Should people with asymptomatic carotid artery stenosis undergo endarterectomy for primary stroke prevention? CMAJ
2004;171:726-7.
158.
Adams HP Jr, del Zoppo G, Alberts MJ, et al. Guidelines for the early management of adults with ischemic stroke. A guideline from the American
Heart Association/ American Stroke Association Stroke Council, Clinical Cardiology Council, Cardiovascular Radiology and Intervention Council,
and the Atherosclerotic Peripheral Vascular Disease and Quality of Care Outcomes in Research Interdisciplinary Working Group. Stroke
2007;38:1655-711.
159.
Kennedy J, Quan H, Ghali WA, et al. Importance of the imaging modality in decision making about carotid endarterectomy. Neurology
2004;62:901-4.
160.
Fairhead JF, Mehta Z, Rothwell PM. Population-based study of delays in carotid imaging and surgery and the risk of recurrent stroke. Neurology
2005;65:371-5.
161.
Jörg E, Roland L and Featherstone M. Randomized controlled trials comparing endarterectomy and endovascular treatment for carotid artery
stenosis. A Cochrane Systematic Review. Stroke 2009;40;1373
162.
Meier, P., Knapp, G., Tamhane, U., Chaturvedi, S. & Gurm, HS. Short term and intermediate term comparison of endarterectomy versus stenting
for carotid artery stenosis: systematic review and meta-analysis of randomised controlled clinical trials. BMJ 2010; 340:c467.
163.
ICSS investigators. Carotid artery stenting compared with endarterectomy in patients with symptomatic carotid stenosis (International Carotid
Stenting Study): an interim analysis of a randomised controlled trial. The Lancet, 2010; 375(9719): 985-997.
164.
Brott, TG., Hobson, RW., Howard, G. Et al for the CREST Investigators. Stenting versus endarterectomy for treatment of carotid-artery stenosis. N
Engl J Med 2010; 363:11-23.
165.
Carotid Stenting Trialists Collaboration. Short-term outcome after stenting versus endarterectomy for symptomatic carotid stenosis: a preplanned
meta-analysis of individual patient data. The Lancet, 2010;376: 1062 – 1073.
166.
Fleetham J, Ayas N, Bradley D, Fitzpatrick M, et al for the Canadian Thoracic Society Sleep Disordered Breathing Committee. Canadian
Thoracic Society 2011 guideline update: Diagnosis and treatment of sleep disordered breathing. Canadian Respiratory Journal, 2011;18(1):25-47.
167.
Epstein LJ, Kristo D, Strollo PJ Jr, Friedman N, Malhotra A, Patil SP, Ramar K, Rogers R, Schwab RJ, Weaver EM, Weinstein MD; Adult
Obstructive Sleep Apnea Task Force of the American Academy of Sleep Medicine. Clinical Guideline for the Evaluation, Management and Longterm Care of Obstructive Sleep Apnea in Adults. Journal of Clinical Sleep Medicine, 2009; 5(3):263-76.
168.
Artz M, Young T, Finn L, Skatrud JB, Bradley TD. Association of sleep-disordered breathing and the occurrence of stroke. American Journal of
Respiratory and Critical Care Medicine, 2005;172(11):1447-1451.
169.
American Academy of Pediatrics. Clinical practice guideline: diagnosis and management of childhood obstructive sleep apnea syndrome.
Pediatrics 2002;109(4):704-712 .
170.
Practice parameters for the respiratory indications for polysomnography in children. Sleep 2011;34(3):379-88.
171.
Redline S, Yenokyan G, Gottlieb DJ, Shahar E, O’Connor GT, Resnick HE, Diener-West M, Sanders MH, Wolf PA, Geraghty EM, Tauqueer A,
Lebowitz M, Punjabi NM. Obstructive sleep apnea-hypopnea and incident stroke: the sleep heart health study. American Journal of Respiratory
and Critical Care Medicine, 2010;182(2):269-277.
172.
Valham F, Mooe T, Rabben T, et al. Increased risk of stroke in patients with coronary artery disease and sleep apnea: a 10-year follow-up.
Circulation 2008; 118:955–960.
Fourth Edition FINAL
Update: September 2012
Page 64 of 69
CBPRSC 2012
Section 2: Prevention
References
173.
Dziewas R, Humpert M, Hopmann B, Kloska SP, Ludemann P, Ritter M, Dittrich R, Ringelstein EB, Young P, Nabavi DG. Increased prevalence of
sleep apnea in patients with recurring ischemic stroke compared with first stroke victims. Journal of Neurology, 2005;252(11):1394-1398.
174.
Harbison J, ford GA, James OFW, Gibson GJ. Sleep-disordered breathing following acute stroke. QJM- Monthly Journal of the Association of
Physicians, 2002;95(11):741-747.
175.
Hui DSC, Choy DKL, Wong LKS, Ko FWS, Li TST, Woo TST, Woo J, Kay R. Prevalence of sleep-disordered breathing and continuous positive
airway pressure compliance: results in Chinese patients with first-ever ischemic stroke. Chest, 2002;122(3):852-860.
176.
Johnson KG, Johnson DC. Frequency of sleep apnea in stroke and TIA patients: a meta-analysis. Journal of Clinical Sleep Medicine,
2010;6(2):131-137.
177.
Joo BE, Seok HY, Yu SW, Kim BJ, Park KW, Lee DH, Jung KY. Prevalence of sleep-disordered breathing in acute ischemic stroke as determined
using a portable sleep apnea monitoring device in Korean subjects. Sleep and Breathing, 2011;15(1):77-82.
178.
Bassetti C, Aldrich MS, Chervin RD, Quint D. Sleep apnea in patients with transient ischemic attack and stroke: a prospective study of 59 patients.
Neurology, 1996;47(5):116-1173.
179.
Cadilhac DA, Thorpe RD, Pearce DC, Barnes M, Rochford PD, Tarquinio N, Davis SM, Donnan GA, Pierce RJ. Sleep disordered breathing in
chronic stroke survivors. A study of the long term follow-up of the SCOPES cohort using home based polysomnography. Journal of Clinical
Neuroscience, 2005;12(6):632-637.
180.
Iranzo A, Santamaria J, Berenguer J, Sanchez M, Chamorro A. Prevalence and clinical importance of sleep apnea in the first night after cerebral
infarction. Neurology, 2002;58(6):911-916.
181.
Kaneko Y, Hajek VE, Zivanovic V, Raboud J, Bradley TD. Relationship of sleep apnea to functional capacity and length of hospitalization
following stroke. Sleep, 2003;26(3):293-297.
182.
Wierzbicka A, Rola R, Wichniak A, Richter P, Ryglewicz D, Jernajczyk W. The incidence of sleep apnea in patients with stroke or transient
ischemic attack. Journal of Physiology and Pharmacology, 2006;57(Suppl4):385-390.
183.
Demede M, Pandey A, Zizi F, Bachmann R, Donat M, McFalrane SI, Jean-Louise G, Ogedegbe G. Resistant hypertension and obstructive sleep
apnea in the primary-care setting. Int J Hypertens, 2011;2011:3409-3429.
184.
Friedman O, Bradley TD, Ruttanaumpawan P, Logan AG. Independent association of drug-resistant hypertension to reduced sleep duration and
efficiency. Am J Hyperten, 2010;23(2):174-179.
185.
Goncalves SC, Martinez D, Gus M, de Abreau-Silva EO, Bertoluci C, Dutra I, Branchi T, Moreira LB, Ruchs SC, de Oliveira AC, Fuchs FD.
Obstructive sleep apnea and resistant hypertension: a case-control study. Chest, 2007;132(6):1858-1862.
186.
Abe H, Takahashi M, Yaegashi H, Eda S, Tsunemoto H, Kamikozawa M, Koyama J, Yamazaki K, Ikeda U. Efficacy of continuous positive airway
pressure on arrhythmias in obstructive sleep apnea patients. Heart Vessels, 2010;25:63-69.
187.
Braga B, Poyares D, Cintra F, Guilleminault C, Cirenza C, Horbach S, Macedo D, Silva R, Tufik S, De Paola AA.. Sleep-disordered breathing and
chronic atrial fibrillation. Sleep Medicine, 2009;10(2):212-6.
188.
Gami AS, Hodge DA, Herges RM, Olson EJ, Nykodym J, Kara T, Somers VK. Obstructive sleep apnea, obesity, and the risk of incident atrial
fibrillation. Journal of the American College of Cardiology, 2007;49(5):565-571.
189.
Stevenson IH, Teichtahl H, Cunnington D, Ciavarella S, Gordon I, Kalman JM. Prevalence of sleep disordered breathing in paroxysmal and
persistent atrial fibrillation patients with normal left ventricular function. European Heart Journal, 2008;29:1662-1669.
190.
Kanagala R, Murali NS, Friedman PA, Ammash NM, Gersh BJ, Ballman KV, Shamsuzzaman ASM, Somers VK. Obstructive sleep apnea and the
recurrence of atrial fibrillation. Circulation, 2003;107:2589-2594.
191.
Ng CY, Liu T, Shehata M, Stevens S, Chugh SS, Wang X. Meta-analysis of obstructive sleep apnea as predictor of atrial fibrillation recurrence after
catheter ablation. Am J Cardiol, 2011;108:47-51.
192.
Svatikova A, Chervin RD, Wing JJ, Sanchez BN, Migda EM, Brown DL. Positional therapy in ischemic stroke patients with obstructive sleep apnea.
Sleep medicine, 2011;3:262-266.
193.
Sundaram S, Lim J, Lasserson TJ. Surgery for obstructive sleep apnoea in adults. Cochrane Database of Systematic Reviews, 2009;4:DOI
10.1002/14651858.CD005308.pub2.
194.
Broadley SA, Jorgensen L, Cheek A, Salonikis S, Taylor J, Thompson PD, Antic R. Early investigation and treatment of obstructive sleep apnoea
after acute stroke. Journal of Clinical Neuroscience, 2007;14(4):328-333.
195.
Patel SR, White DP, Malhotra A, Stanchina ML, Ayas NT. Continuous positive airway pressure therapy for treating sleepiness in a diverse
population with obstructive sleep apnea results of a meta-analysis. Archives of Internal Medicine, 2003;163(5):565-571.
196.
Wessendorf TE, Wang Y-M, Thilmann AF, Sorgenfrei U, Konietzko N, Teschler H. Treatment of obstructive sleep apnoea with nasal continuous
positive airway pressure in stroke. European Respiratory Journal, 2001;18(4):623-629.
197.
Bravata DM, Concato J, Fried T, Ranjbar N, Sadarangani T, McClain V, Struve F, Zygmunt L, Knight HJ, Lo A, Richerson GB, Gorman M, Williams
LS, Brass LM, Agostini J, Mohsenin V, Roux F, Yaggi K. Auto-titrating continuous positive airway pressure for patients with acute transient
ischemic attack: a randomized feasibility trial. Stroke, 2010;41(7):1464-1470.
198.
Martinez-Garcia MA, Galiano-Blancar R, Roman-Sanchez P, Soler-Cataluna JJ, Cabero-Salt L, Salcedo-Maiques E. Continuous positive airway
pressure treatment in sleep apnea prevents new vascular events after ischemic stroke. Chest, 2005;128(4):2123-2129.
199.
Giles TL, Lasserson TJ, Smith B, White J, Wright JJ, Cates CJ. Continuous positive airway pressure for obstructive sleep apnoea in adults.
Cochrane Database of Systematic Reviews, 2006;3:DOI 10.1002/14651858.CD001106.pub3.
200.
Martinez-Garcia MA, Campos-Rodriguez F, Soler-Cataluna JJ, Catalan-Serr P, Roman-Sanchez P, Montserrat JM. Increased incidence of nonfatal cardiovascular events in stroke patient with sleep apnea. Effect of CPAP treatment. A 7-year follow-up study. European Respiratory Journal,
2011; September 29 doi: 10.1183/09031936.00011311
Fourth Edition FINAL
Update: September 2012
Page 65 of 69
CBPRSC 2012
Section 2: Prevention
References
201.
Bravata DM, Concato J, Fried T, Ranjbar N, Sadarangani T, McClain V, Struve F, Zygmunt L, Knight HJ, Lo A, Richerson GB, Gorman M, Williams
LS, Brass LM, Agostini J, Mohsenin V, Roux F, Yaggi K. Continuous positive airway pressure: Evaluation of a novel therapy for patients with acute
ischemic stroke. Sleep, 2011;34(9):1271-1277.
202.
Hsu CY, Vennelle M, Li HY, Engleman M, Dennis MS, Douglas NJ. Sleep-disordered breathing after stroke: a randomized controlled trial of
continuous positive airway pressure. Journal of Neurology, Neurosurgery, and Psychiatry, 2006;77(10):1143-1149.
203.
Parra O, Sanchez-Armengol A, Bonnin M, Arboix A, Campos-Rodriguez F, Perez-Ronchel J, Duran-Cantolla J, De La Torre G, Gonzalez Marcos
JR, de la Pena M, Carmen Jimenez M, Masa F, Casado I, Luz Alonso M, Macarron JL. Early treatment of obstructive apnoea and stroke outcome:
a randomized controlled trial. European Respiratory Journal, 2011;37(5):1128-1136.
204.
Brown DL, Chervin RD, Hickenbottom SL, Langa KM, Morgenstern LB Screening for obstructive sleep apnea in stroke patients: a costeffectiveness analysis. Stroke, 2005;36(6):1291-1293.
205.
Fiore MC, Jaen CR, Baker TB, et al. Treating Tobacco Use and Dependence: 2008 Update. Clinical Practice Guideline. Rockville, MD: U.S.
Department of Health and Human Services. Public Health Service. May 2008.
206.
Quality of Stroke Care in Canada Report. 2011, Canadian Stroke Network: Ottawa Canada. www.strokebestpractices.ca
207.
CAN-ADAPTT (January 2011). Canadian Practice-Informed Smoking Cessation Guideline: Summary Statements. Toronto, Canada: Canadian
Action Network for the Advancement, Dissemination and Adoption of Practice-informed Tobacco Treatment, Centre for Addiction and Mental
Health.
208.
National Institute for Health and Clinical Excellence (NICE). Brief interventions and referral for smoking cessation in primary care and other
settings. London (UK): National Institute for Health and Clinical Excellence; 2006 Mar. 36 p.
209.
Public Health Agency of Canada. Tracking heart disease and stroke in Canada. 2009. Available:www.phac-aspc.gc.ca.
210.
United States Department of Health and Human Services. Ending the Tobacco Epidemic. Progress towards a healthier nation. Report August
2012. http://www.hhs.gov/ash/initiatives/tobacco/tobaccoprogress2012.pdf
211.
World Health Organization. WHO report on the global tobacco epidemic 2008: the MPOWER package. Geneva: The Organization; 2008.
212.
Wolf PA, D’Agostino RB, Kannel WB, et al. Cigarette smoking as a risk factor for stroke. The Framingham Study. JAMA 1988;259:1025-9.
213.
Kawachi I, Colditz GA, Stampfer MJ, et al. Smoking cessation in relation to total mortality rates in women. A prospective cohort study. Ann Intern
Med 1993;119:992-1000.
214.
Flemming KD, Brown RD Jr. Secondary prevention strategies in ischemic stroke: identification and optimal management of modifiable risk factors.
Mayo Clin Proc 2004;79:1330-40.
215.
Robbins AS, Manson JE, Lee IM, et al. Cigarette smoking and stroke in a cohort of US male physicians. Ann Intern Med 1994;120:458-62.
216.
Asthana A, Piper ME, McBride PE, Ward A, Fiore MC, Baker TB, Stein JH. Long-term effects of smoking and smoking cessation on exercise stress
testing: three-year outcomes from a randomized clinical trial. Am Heart J, 2012;163:81-87.
217.
Goldstein LB, Adams R, Becker K, et al. Primary prevention of ischemic stroke: a statement for healthcare professionals from the Stroke Council of
the American Heart Association. Stroke 2001;32:280 99.
218.
Hankey GJ. Smoking and risk of stroke. J Cardiovasc Risk 1999;6:207-11.
219.
Bonita R, Duncan J, Truelsen T, et al. Passive smoking as well as active smoking increases the risk of acute stroke. Tob Control 1999;8:156-60.
220.
Kurth T, Kase CS, Berger K, et al. Smoking and risk of hemorrhagic stroke in women. Stroke 2003;34:2792-5.
221.
Kurth T, Kase CS, Berger K, et al. Smoking and the risk of hemorrhagic stroke in men. Stroke 2003;34:1151-5.
222.
Mazza A, Pessina AC, Pavei A, et al. Predictors of stroke mortality in elderly people from the general population. The CArdiovascular STudy in the
ELderly (CASTEL). Eur J Epidemiol 2001;17:1097-104.
223.
Butler KM, Fallin A, Ridner SL. Evidence-based smoking cessation for college students. Nurs Clin N Am, 2012;47:21-30.
224.
Audrain-McGovern J, Stevens S, Murray PJ, Kinsman S, Zuckoff A, Pletcher J, Moss D, Baumritter A, Kalkhuis-Beam S, Carlson E, Rodriguez D,
Wileyto EP. The efficacy of motivational interviewing versus brief advice for adolescent smoking behaviour change. Pediatrics, 2011;128:e101-111.
225.
Rosen LJ, Noach MB, Winickoff JP, Hovell MF. Parental smoking cessation to protect young children: a systematic review and meta-analysis.
Pediatrics, 2012;129:141-152.
226.
Carson KV, Brinn MP, Peters M, Veale A, Esterman AJ, Smith BJ. Interventions for smoking cessation in Indigenous populations. Cochrane
Database of Systematic Reviews 2012, Issue 1. Art.No.: CD009046.
227.
Cabezas C, Advani M, Puente D, Rodriguez-Blanco T, Martin C, ISTAPS Study Group. Effectiveness of a stepped primary care smoking cessation
intervention: cluster randomized clinical trial (ISTAPS study). Addiction, 2011;106:1696-1706.
228.
Ussher MH, Taylor A, Faulkner G. Exercise interventions for smoking cessation. Cochrane Database of Systematic Reviews 2012, Issue 1. Art.
No.: CD002295.
229.
Wu, P., Wilson, K., Dimoulas, P. & Mills, EJ. Effectiveness of smoking cessation therapies: a systematic review and meta-analysis. BMC Public
Health 2006; 6: article number 300.
230.
Cox, LS., Patten, CA., Niaura, RS>, Decker, PA., Rigotti, N, Sachs, DPL., Sonia Buist, A., & Hurt, RD. Efficacy of bupropion for relapse prevention
in smokers with and without history of major depression. Journal of General Internal Medicine, 2004; 19(8): 828-834
Fourth Edition FINAL
Update: September 2012
Page 66 of 69
CBPRSC 2012
Section 2: Prevention
References
Clinical Practice Guidelines Related to the Prevention of Stroke
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
Anderson T, Campbell N, Carpentier A, Frohlich J, Genest J, McPherson R, …Ur E. 2009 Canadian Cardiovascular Society/Canadian guidelines for
the diagnosis and treatment of dyslipidemia and prevention of cardiovascular disease in the adult – 2009 recommendations. Canadian Journal of
Cardiology. 2009 October; 25(10): 567-579. Available at:
http://www.ccs.ca/download/consensus_conference/consensus_conference_archives/2009_Dyslipidemia-Guidelines.pdf.
Adams RJ, Albers G, Alberts MJ, Benavente O, Furie K, Goldstein LB, … Schwamm LH, American Heart Association, American Stroke Association.
Update to the AHA/ASA recommendations for the prevention of stroke in patients with stroke and transient ischemic attack. Stroke 2008
May;39(5):1647-52. Available at: http://stroke.ahajournals.org/content/39/5/1647.full.pdf+html.
American Academy of Family Physicians (AAFP). Summary of recommendations for clinical preventive services. Leawood (KS): American Academy
of Family Physicians (AAFP); 2011. Available at: http://guidelines.gov/content.aspx?id=36873.
American Academy of Pediatrics. Clinical practice guideline: diagnosis and management of childhood obstructive sleep apnea syndrome. Pediatrics
2002;109(4):704-712 .
Albers GW, Guyatt G, Harrington R, Hirsh J, Schunemann HJ. Antithrombotic and Thrombolytic Therapy: American College of Chest Physicians
Evidence-Based Clinical Practice Guidelines (8th Edition). Chest 2008 June; 133(6): 113-122. Available at:
http://www.chestnet.org/accp/guidelines/antithrombotic-and-thrombolytic-therapy-8th-edition.
American Dietetic Association (ADA). Pediatric weight management evidence-based nutrition practice guideline. Chicago (IL): American Dietetic
Association (ADA); 2007 Jun. Various p. Available at: http://guidelines.gov/content.aspx?id=12821.
Aurora RN, Chowdhuri S, Ramar K, Bista SR, Casey KR, Lamm CI, … Zak RS. The treatment of central sleep apnea syndromes in adults: practice
parameters with an evidence-based literature review and meta-analyses. Sleep 2012;35(1):17-40. Available at:
http://www.journalsleep.org/ViewAbstract.aspx?pid=28393.
Aurora RN, Auerbach SH, Karippot A, Lamm CI, Morgenthaler TI, Ramar K, … Zak RS. Practice parameters for the respiratory indications for
polysomnography in children. Sleep 2011;34(3):379-88. [150 references] Available at: http://www.journalsleep.org/ViewAbstract.aspx?pid=28075.
Bederson JB, Connolly ES Jr, Batjer HH, Dacey RG, Dion JE, Diringer MN, … American Heart Association. Guidelines for the management of
aneurysmal subarachnoid hemorrhage: a statement for healthcare professionals from a special writing group of the Stroke Council, American Heart
Association. Stroke 2009 Mar;40(3):994-1025. Available at: http://guidelines.gov/content.aspx?id=15560.
CAN-ADAPTT (January 2011). Canadian Practice-Informed Smoking Cessation Guideline: Summary Statements. Toronto, Canada: Canadian Action
Network for the Advancement, Dissemination and Adoption of Practice-informed Tobacco Treatment, Centre for Addiction and Mental Health.
Canadian Cardiovascular Society. Guidelines Library. 2010 and 2012 Atrial Fibrillation Guidelines. Available at:
http://www.ccs.ca/guidelines/cc_library_e.aspx.
Canadian Diabetes Association. 2008 clinical practice guidelines for the prevention and management of diabetes in Canada. Canadian Journal of
Diabetes, 2008;32(1). Available at: http://www.diabetes.ca/for-professionals/resources/2008-cpg/.
Chaturvedi S, Bruno A, Feasby T, Holloway R, Benavente O, Cohen SN, …Wilterdink J. Carotid endarterectomy--an evidence-based review: report of
the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology 2005 Sep 27;65(6):794-801.
Available at: http://www.aan.com/professionals/practice/guidelines/pda/Carotid_Endarterectomy.pdf.
Hypertension Canada. Canadian Hypertension Education Program (CHEP) Recommendations, 2012. Available at: http://www.hypertension.ca/cheprecommendations.
Cincinnati Children's Hospital Medical Center. Best evidence statement (BESt). Screening of hypertension in pediatric patients with diabetes.
Cincinnati (OH): Cincinnati Children's Hospital Medical Center; 2011. Available at: http://guidelines.gov/content.aspx?id=33572.
Clinical guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. American Academy of Sleep Medicine
2009. Available at: http://www.guideline.gov/content.aspx?id=15298&search=%22stroke%22+and+%22sleep+apnea%22
National Institute for Health and Clinical Excellence. Clopidogrel and modified-release dipyridamole for the prevention of occlusive vascular events
(review of technology appraisal guidance 90, 2010). 2010 December. Available at: http://www.nice.org.uk/TA210.
Coull BM, Williams LS, Goldstein LB, Meschia JF, Heitzman D, Chaturvedi S, …Saver JL. Anticoagulants and antiplatelet agents in acute ischemic
stroke: report of the Joint Stroke Guideline Development Committee of the American Academy of Neurology and the American Stroke Association (a
division of the American Heart Association). Neurology 2002 Jul 9;59(1):13-22. Available at:
http://www.guidelines.gov/content.aspx?id=4101&search=anticoagulants+AND+hemorrhage.
Department of Veteran Affairs, Department of Defense. VA/DoD clinical practice guideline for the management of diabetes mellitus. Washington (DC):
Department of Veteran Affairs, Department of Defense; 2010 Aug:146 p. Available at: http://guideline.gov/content.aspx?id=24192.
European Society of Cardiology (ESC), European Association for Cardiovascular Prevention and Rehabilitation (EACPR), Council on Cardiovascular
Nursing, European Association for Study of Diabetes (EASD), International Diabetes Federation Europe (IDF-Europe), European Stroke Initiative
(EUSI), … European Atherosclerosis Society (EAS). European guidelines on cardiovascular disease prevention in clinical practice. Fourth Joint Task
Force of the European Society of Cardiology and other societies [trunc]. Eur J Cardiovasc Prev Rehabil 2007 Sep;14 Suppl 2:S1-113. Available at:
http://guideline.gov/content.aspx?id=12541.
Finnish Medical Society Duodecim. Treatment of dyslipidaemias. In: EBM Guidelines. Evidence-Based Medicine [Internet]. Helsinki, Finland: Wiley
Interscience. John Wiley & Sons; 2010 Aug 3. Available at: http://guideline.gov/content.aspx?id=24712.
Fiore MC, Jaen CR, Baker TB, Bailey WC, Curry SJ, Dorfman SF, … Wewers ME. Treating Tobacco Use and Dependence: 2008 Update. Clinical
Practice Guideline. Rockville, MD: U.S. Department of Health and Human Services. Public Health Service. May 2008. Available at:
http://www.ahrq.gov/clinic/tobacco/treating_tobacco_use08.pdf.
Goldstein LB, Adams R, Becker K, Furberg CD, Gorelick PB, Hademenos G, … del Zoppo DJ. Primary prevention of ischemic stroke: a statement for
healthcare professionals from the Stroke Council of the American Heart Association. Stroke 2001;32:280 99. Available at:
http://stroke.ahajournals.org/content/32/1/280.full.
Gorelick PB, Scuteri A, Black S, Black SE, DeCarli C, Greenberg SM, … Seshadri S. Vascular contributions to cognitive impairment and dementia: a
statement for healthcare professionals from the American heart association/American stroke association. Stroke, 2011;42:2672-2713. Available at:
http://stroke.ahajournals.org/content/42/9/2672.abstract.
Institute for Clinical Sytems Improvement. Health Care Guideline: Diagnosis and Treatment of Obstructive Sleep Apnea. June 2008. Available at:
http://www.icsi.org/sleep_apnea/sleep_apnea__diagnosis_and_treatment_of_obstructive_.html.
Fourth Edition FINAL
Update: September 2012
Page 67 of 69
CBPRSC 2012
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
Section 2: Prevention
References
Institute for Clinical Systems Improvement (ICSI). Diagnosis and management of type 2 diabetes mellitus in adults. Bloomington (MN): Institute for
Clinical Systems Improvement (ICSI); 2010 Jul. 112 p. Updated version; April 2012
http://www.icsi.org/diabetes_mellitus__type_2/management_of_type_2_diabetes_mellitus__9.html.
Institute for Clinical Systems Improvement (ICSI). Healthy lifestyles. Bloomington (MN): Institute for Clinical Systems Improvement (ICSI); 2011 May.
68 p. Available at: http://guidelines.gov/content.aspx?id=34021.
Institute for Clinical Systems Improvement (ICSI). Hypertension diagnosis and treatment. Bloomington (MN): Institute for Clinical Systems
Improvement (ICSI); 2010 November. Available at: http://www.icsi.org/hypertension_4/hypertension_diagnosis_and_treatment_4.html.
Institute for Clinical Systems Improvement (ICSI). Preventive services for adults. Bloomington (MN): Institute for Clinical Systems Improvement (ICSI);
2011 Sep. 90 p. Available at: http://www.icsi.org/preventive_services_for_adults/preventive_services_for_adults_4.html.
Management of Asthma Working Group. VA/DoD clinical practice guideline for management of asthma in children and adults. Washington (DC):
Department of Veteran Affairs, Department of Defense; 2009. 126 p. Available at: http://www.healthquality.va.gov/asthma/ast_2_full.pdf.
Scottish Intercollegiate Guidelines Network. Management of patients with stroke or TIA: assessment, investigation, immediate management and
secondary prevention. A national clinical guideline. Edinburgh (Scotland): Scottish Intercollegiate Guidelines Network (SIGN); 2008. 103 p. Available
at: http://www.sign.ac.uk/pdf/sign108.pdf.
Management of Stroke Rehabilitation Working Group. VA/DoD clinical practice guideline for the management of stroke rehabilitation. Washington
(DC): Veterans Health Administration, Department of Defense, The American Heart Association/ American Stroke Association; 2010. 150 p. Available
at: http://www.healthquality.va.gov/stroke/stroke_full_221.pdf.
National Guideline Clearinghouse (NGC). Guideline summary: EFNS guideline on neuroimaging in acute stroke. Report of an EFNS task force. In:
National Guideline Clearinghouse (NGC) Rockville (MD): cited 2006 Dec. Available at: http://www.guidelinecentral.com/guidelinelibrary/summaries/ngc-5498-efns-guideline-neuroimaging-acute-stroke.
National Blood Pressure and Vascular Disease Advisory Committee. Guide to management of hypertension 2008: Assessing and Managing Raised
Blood Pressure in Adults. Canberra: National Heart Foundation of Australia; 2008. Available at:
http://www.heartfoundation.org.au/SiteCollectionDocuments/HypertensionGuidelines2008to2010Update.pdf.
National Clinical Guideline Centre. Hypertension. Clinical management of primary hypertension in adults. London (UK): National Institute for Health
and Clinical Excellence (NICE); 2011 August. Available at: http://www.nice.org.uk/nicemedia/live/13561/56007/56007.pdf.
National Collaborating Centre for Chronic Conditions. Stroke. Diagnosis and initial management of acute stroke and transient ischaemic attack (TIA).
London (UK): National Institute for Health and Clinical Excellence (NICE); 2008 Jul. 37 p. Available at:
http://www.nice.org.uk/nicemedia/pdf/CG68NICEGuideline.pdf.
National Institute for Health and Clinical Excellence (NICE). Brief interventions and referral for smoking cessation in primary care and other settings.
London (UK): National Institute for Health and Clinical Excellence (NICE); 2006 Mar. 36 p. Available at: http://www.guidelinecentral.com/guidelinelibrary/summaries/ngc-5214-brief-interventions-and-referral-smoking-ce.
National Institute for Health and Clinical Excellence (NICE). Smoking cessation services in primary care, pharmacies, local authorities and
workplaces, particularly for manual working groups, pregnant women and hard to reach communities: Public health guidance; no. 10. London (UK):
National Institute for Health and Clinical Excellence (NICE); 2008 Feb. 87 p. Available at: http://www.nice.org.uk/nicemedia/pdf/PH010guidance.pdf.
National Institute for Health and Clinical Excellence (NICE). Continuous positive airway pressure for the treatment of obstructive sleep
apnoea/hypopnoea syndrome: NICE technology appraisal guidance 139. London (UK): National Institute for Health and Clinical Excellence; 2010
November. Available at: http://www.nice.org.uk/nicemedia/pdf/TA139Guidance.pdf.
National Institute for Health and Clinical Excellence (NICE). Dabigatran etexilate for the prevention of stroke and systemic embolism in atrial
fibrillation: NICE technology appraisal guidance TA249. London (UK): National Institute for Health and Clinical Excellence; 2012 March. Available at:
http://publications.nice.org.uk/dabigatran-etexilate-for-the-prevention-of-stroke-and-systemic-embolism-in-atrial-fibrillation-ta249 (Accessed: June,
2012).
National Institute for Health and Clinical Excellence (NICE). Rivaroxaban for the prevention of stroke and systemic embolism in people with atrial
fibrillation: NICE technology appraisal guidance 256. Available at: http://www.nice.org.uk/nicemedia/live/13746/59295/59295.pdf (Accessed: June,
2012).
Public Health Agency of Canada. National consultation on physical activity guidelines – Final Report. 2011 January. Available at: http://www.phacaspc.gc.ca/hp-ps/hl-mvs/pa-ap/03paap-eng.php.
Aurora RN, Zak RS, Karippot A, Lamm CI, Morgenthaler TI, Auerbach SH, …Ramar K. Practice parameters for the respiratory indications for
polysomnography in children. Sleep 2011;34(3):379-88. Available at:
http://guidelines.gov/content.aspx?id=33568&search=Chronic+respiratory+disease+in+perinatal+period+.
Goldstein LB, Adams R, Alberts MJ, Appel LJ, Brass LM, Bushnell CD, …Sacco RL. Primary prevention of ischemic stroke: a guideline from the
American Heart Association/American Stroke Association Stroke Council. American Heart Association/ American Stroke Association; 2006 May.
Available at: http://stroke.ahajournals.org/content/37/6/1583.full.pdf+html.
Scottish Intercollegiate Guidelines Network (SIGN). Management of diabetes: A national clinical guideline. Edinburgh (Scotland): Scottish
Intercollegiate Guidelines Network (SIGN); 2010 Mar. 170 p. (SIGN publication; no. 116). Available at: http://www.sign.ac.uk/pdf/sign116.pdf.
Scottish Intercollegiate Guidelines Network. Management of Obstructive Sleep Apnoea/Hypopnoea Syndrome in Adults: Guideline No. 73. Healthcare
Improvement Scotland; 2003 July. Available at: http://www.sign.ac.uk/GUIDELINES/fulltext/73/index.html.
Secondary Prevention. In: clinical guidelines for stroke management 2010. Melbourne (Australia): National Stroke Foundation; 2010 Sep. p. 68-76.
Available at: http://guideline.gov/content.aspx?id=24172.
Singapore Ministry of Health. Stroke and transient ischaemic attacks. Assessment, investigation, immediate management and secondary prevention.
Singapore: Singapore Ministry of Health; 2009 Jul. 64 p. Available at: http://guideline.gov/content.aspx?id=15541.
U.S. Preventive Services Task Force. Counseling and interventions to prevent tobacco use and tobacco-caused disease in adults and pregnant
women: U.S. Preventive Services Task Force reaffirmation recommendation statement. Ann Intern Med; 2009 April;150(8):551-5. Available at:
http://annals.org/article.aspx?articleid=744446.
Canadian Diabetes Association. Vascular Protection in People With Diabetes: Canadian Diabetes Association Clinical Practice Guidelines Expert
Committee. 2008 Clinical Practice Guidelines. Available at:
http://www.diabetes.ca/documents/2008CPG/24%20VASCULAR%20PROTECTION%20IN%20PEOPLE%20WITH%20DIABETES-S102-S106.pdf.
Fourth Edition FINAL
Update: September 2012
Page 68 of 69