You are on page 1of 197

THE JOURNAL OF THE ALZHEIMER’S ASSOCIATION

Volume 3, Issue 4 October 2007

CONTENTS
Editorial 333 Structural neuroimaging in the diagnosis of
dementia
261 Zenith Award Program of the Alzheimer’s Tiffany Chow
Association
Zaven S. Khachaturian 336 Functional neuroimaging in the diagnosis of
dementia
262 Introduction: The Third Canadian Consensus Michael Borrie
Conference on the Diagnosis and Treatment
of Dementia, 2006 341 General risk factors for dementia:
Howard Chertkow A systematic evidence review
Christopher Patterson, John Feightner,
Review Articles Angeles Garcia, Chris MacKnight

266 Mild cognitive impairment and cognitive 348 Primary prevention of dementia
impairment, no dementia: Part A, concept Christopher Patterson, John Feightner,
and diagnosis Angeles Garcia, Chris MacKnight
Howard Chertkow, Ziad Nasreddine, 355 Management of mild to moderate
Yves Joanette, Valérie Drolet, John Kirk, Alzheimer’s disease and dementia
Fadi Massoud, Sylvie Belleville, Howard Bergman David B. Hogan, Peter Bailey, Anne Carswell,
283 Mild cognitive impairment and cognitive Barry Clarke, Carole Cohen, Dorothy Forbes,
impairment, no dementia: Part B, therapy Malcolm Man-Son-Hing, Krista Lanctôt,
Fadi Massoud, Sylvie Belleville, John Kirk, Debra Morgan, Lilian Thorpe
Howard Chertkow, Ziad Nasreddine, 385 Clinical practice guidelines for severe
Yves Joanette, Howard Bergman, Alzheimer’s disease
Morris Freedman Nathan Herrmann, Serge Gauthier, Paul G. Lysy
292 Clinical diagnosis of dementia 398 Management of dementia with a
Alain Robillard cerebrovascular component
299 Neuropsychological testing and assessment Christian Bocti, Sandra Black, Chris Frank
for dementia 404 Disclosure of the diagnosis of dementia
Claudia Jacova, Andrew Kertesz, Mervin Blair, John D. Fisk, B. Lynn Beattie, Martha Donnelly,
John Fisk, Howard H. Feldman Anna Byszewski, Frank J. Molnar
318 Cobalamin and homocysteine in older adults: 411 Ethical considerations for decision making
Do we need to test for serum levels in the for treatment and research participation
work-up of dementia? John D. Fisk, B. Lynn Beattie, Martha Donnelly
Angeles Garcia
418 Genetics and dementia: Risk factors,
325 The role of biologic markers in the diagnosis diagnosis, and management
of Alzheimer’s disease Ging-Yuek Robin Hsiung, A. Dessa Sadovnick
Hyman M. Schipper

Contents continued on next page


Contents continued
428 Toward a revision of criteria for the 444 Commentary on “The Third Canadian
dementias Consensus Conference on the Diagnosis and
Kenneth Rockwood, Rémi W Bouchard, Treatment of Dementia”
Richard Camicioli, Gabriel Léger Bruno Vellas, Emma Reynish, Philippe Robert
446 Commentary on “The Third Canadian
Perspectives Consensus Conference on the Diagnosis
441 Commentary on “The Third Canadian and Treatment of Dementia”—An appraisal
Consensus Conference on the Diagnosis and Paul S. Aisen
Treatment of Dementia:” Clinical guidelines
are not enough—System-wide, population- Association Pages
based programs are needed to improve the
449 Alzheimer’s Association Update
care of patients with Alzheimer’s disease and
related dementias
Howard Fillit

Cover image courtesy of Drs. William Jagust and Ansgar Furst, University of California, Berkeley and Lawrence
Berkeley National Laboratory.
The top pair of images is a patient with frontotemporal lobar degeneration (FTLD), the center pair is a patient with
Alzheimer’s disease (AD), and the bottom pair is a normal older adult. Images on the left column are FDG-PET
superimposed on the MRI. Images on the right column are PET with Pittsburgh compound B (PIB) superimposed on the
MRI. Left column images show frontal hypometabolism in FTLD patient, parietal hypometabolism in AD patient, and
normal metabolism in the normal older adult. Right column images show PIB binding only in the AD patient and not
the FTLD patient or normal adult.
THE JOURNAL OF THE ALZHEIMER’S ASSOCIATION

Alzheimer’s & Dementia (ISSN 1552-5260) is published quarterly by available online may be obtained by purchasing Pay-Per-View access for
Elsevier, 360 Park Avenue South, New York, NY 10010-1710. Months $32 per article on the journal website, www.alzheimersanddementia.org.
of issue are January, April, July, and October. Business Offices: 1600 John
© 2007 The Alzheimer’s Association. All rights reserved. This
F. Kennedy Boulevard, Suite 1800, Philadelphia, PA 19103-2899. Edi-
journal and the individual contributions contained in it are protected
torial Office: 11830 Westline Industrial Dr, St Louis, MO 63146-3318.
under copyright by The Alzheimer’s Association and the following terms
Accounting and Circulation Offices: 6277 Sea Harbor Drive, Orlando,
and conditions apply to their use:
FL 32887-4800.
Photocopying. Single photocopies of single articles may be made for
CUSTOMER SERVICE (orders, claims, online, change of address): personal use as allowed by national copyright laws. Permission of the
Elsevier Periodicals Customer Service, 6277 Sea Harbor Drive, Orlando, Publisher and payment of a fee is required for all other photocopying,
FL 32887-4800. Tel: (800) 654-2452 (U.S. and Canada); (407) 345-4000 including multiple or systematic copying, copying for advertising or
(outside U.S. and Canada). Fax: (800) 225-4030 (U.S. and Canada); (407) promotional purposes, resale, and all forms of document delivery. Special
363-9661 (outside U.S. and Canada). E-mail: elspcs@elsevier.com Ad- rates are available for educational institutions that wish to make photo-
dress changes must be submitted four weeks in advance. copies for non-profit educational classroom use.
Permission may be sought directly from Elsevier’s Global Rights
YEARLY SUBSCRIPTION RATES: United States and possessions: Department in Oxford, UK: phone 215-239-3804 or ⫹44 (0)1865
Individual $105.00; Institution $229.00; Student/Resident: $70.00. All 843830, fax ⫹44 (0)1865 853333, e-mail healthpermissions@elsevier.
other countries (prices include airspeed delivery): Individual $105.00; com. Requests may also be completed online via the Elsevier homepage
Institution $229.00; Student/Resident $70.00. Single Issues $58.00. To (http://www.elsevier.com/permissions).
receive student/resident rate, orders must be accompanied by name of In the USA, users may clear permissions and make payments through
affiliated institution, date of term and the signature of program/residency the Copyright Clearance Center, Inc., 222 Rosewood Drive, Danvers,
coordinator on institution letterhead. Orders will be billed at the indi- MA 01923, USA; phone: (978) 750-8400, fax: (978) 750-4744, and in
vidual rate until proof of status is received. Current prices are in effect for the UK through the Copyright Licensing Agency Rapid Clearance
back volumes and back issues. Service (CLARCS), 90 Tottenham Court Road, London W1P 0LP,
Further information on this journal is available from the Publisher or UK; phone: (⫹44) 20 7631 5555; fax: (⫹44) 20 7631 5500. Other
from this journal’s website (http://www.alzheimersanddementia.org). countries may have a local reprographic rights agency for payments.
Information on other Elsevier products is available through Elsevier’s
website (http://www.elsevier.com). Derivative Works. Subscribers may reproduce tables of contents or
prepare lists of articles including abstracts for internal circulation within
Advertising information. Advertising orders and inquiries can be sent their institutions. Permission of the Publisher is required for resale or
to: USA, Canada, and South America, Hank Blaney, Elsevier, 360 distribution outside the institution.
Park Ave South, New York, NY 10010: phone (212) 633-3648; fax: Permission of the Publisher is required for all other derivative works,
(212) 633-3820; e-mail, h.blaney@elsevier.com. Classified advertising including compilations and translations.
orders and inquiries can be sent to Simone Imbert, Elsevier, 360 Park Ave Electronic Storage or Usage. Permission of the Publisher is required
South, New York, NY 10010: phone (212) 462-1908, fax (212) 633- to store or use electronically any material contained in this journal,
3820, e-mail, s.imbert@elsevier.com. Europe and the rest of the including any article or part of an article.
world, Julie Toop; phone ⫹44 (0) 1865 843016; fax ⫹44 (0) 1865 Except as outlined above, no part of this publication may be repro-
843976; e-mail media@elsevier.com. duced, stored in a retrieval system or transmitted in any form or by any
means, electronic, mechanical, photocopying, recording or otherwise,
Author inquiries. For inquiries relating to the submission of articles
without prior written permission of the Publisher.
(including electronic submission where available), please visit www.
Address permissions requests to: Elsevier Rights Department, at the
elsevier.com/authors. This site also provides the facility to track ac-
fax and e-mail addresses noted above.
cepted articles and set up e-mail alerts to inform you of when an article’s
status has changed, as well as detailed artwork guidelines, copyright Notice. No responsibility is assumed by the Publisher for any injury
information, frequently asked questions, and more. Please see Informa- and/or damage to persons or property as a matter of products liability,
tion for Authors for individual journal requirements. Contact details for negligence or otherwise, or from any use or operation of any methods,
questions arising after acceptance of an article, especially those relating to products, instructions or ideas contained in the material herein. Because
proofs, are provided after registration of an article for publication. Re- of rapid advances in the medical sciences, in particular, independent
prints. For queries about author offprints, e-mail authorsupport@ verification of diagnoses and drug dosages should be made.
elsevier.com. To order 100 or more reprints for educational, commercial, Although all advertising material is expected to conform to ethical
or promotional use, contact the Commercial Reprints Department, (medical) standards, inclusion in this publication does not constitute a
Elsevier Inc., 360 Park Avenue South, New York, NY 10010-1710. Fax: guarantee or endorsement of the quality or value of such product or of
(212) 462-1935; e-mail reprints@elsevier.com. Reprints of single articles the claims made of it by its manufacturer.
THE JOURNAL OF THE ALZHEIMER’S ASSOCIATION

Volume 3, Issue 4 October 2007

EDITORIAL BOARD
Editor-in-Chief Editorial Advisory Board
Zaven S. Khachaturian Philippe Amouyel, Lille, France
451 Hungerford Drive Carol Barnes, Tucson, AZ
Suite 119-355 Anders Björklund, Lund, Sweden
Rockville, MD 20850 Francois Boller, Paris, France
Tel. 301-294-7201; Fax 301-294-7203 Heiko Braak, Frankfurt, Germany
E-Mail: ADJ@kra.net Jason Brandt, Baltimore, MD
John C. S. Breitner, Seattle, WA
Editors-at-Large Henry Brodaty, Sydney, Australia
Floyd Bloom, San Diego, CA Louis Burgio, Chapel Hill, NC
Paul Greengard, New York, NY Carl Cotman, Irvine, CA
Peggye Dilworth-Anderson, Chapel Hill, NC
Associate Editors Fred Gage, La Jolla, CA
Paul Coleman, Rochester, NY Alison Goate, St Louis, MO
Francoise Forette, Paris, France Michel Goedert, Cambridge, United Kingdom
Virginia Lee, Philadelphia, PA Jeff Kaye, Portland, OR
Marsel Mesulam, Chicago, IL David Knopman, Rochester, MN
John Trojanowski, Philadelphia, PA Jae-Young Koh, Seoul, South Korea
Michael Weiner, San Francisco, CA Lewis Kuller, Pittsburgh, PA
Eckhard Mandelkow, Hamburg, Germany
Eva Mandelkow, Hamburg, Germany
Richard Mayeux, New York, NY
Steven M. Paul, Indianapolis, IN
Ron Petersen, Rochester, MN
Steve Post, Cleveland, OH
Teresa Radebaugh, El Dorado, KS
Joseph Rogers, Phoenix, AZ
Allen Roses, Durham, NC
Barbara Sahakian, Cambridge, United Kingdom
Mary Sano, New York, NY
Lon S. Schneider, Los Angeles, CA
Dennis Selkoe, Boston, MA
Sangram Sisodia, Chicago, IL
Philip Sloane, Chapel Hill, NC
Publication Staff Christine Van Broeckhoven, Antwerp, Belgium
Managing Editor Sandra Weintraub, Chicago, IL
Ara S. Khachaturian Kathleen Welsh-Bohmer, Durham, NC
Tel. 301-309-6730; Fax: 301-309-6724 Bengt Winblad, Stockholm, Sweden
E-mail: ADJ_MgrEd@kra.net

Publisher
Elsevier Inc.
11830 Westline Industrial Dr, St Louis, MO 63146-3318
THE JOURNAL OF THE ALZHEIMER’S ASSOCIATION

Volume 3, Issue 4 October 2007

Alzheimer’s Association
MEDICAL AND SCIENTIFIC ADVISORY COUNCIL
Chair William E. Klunk, MD, PhD Chairs Emeriti
Samuel E. Gandy, MD, PhD University of Pittsburgh School of Marilyn S. Albert, PhD
Thomas Jefferson University Medicine Leonard Berg, MD
Farber Institute for Neurosciences Department of Psychiatry Steven DeKosky, MD
Philadelphia, PA Pittsburgh, PA David Drachman, MD
Robert Katzman, MD
Vice Chair John C. Morris, MD William Markesbery, MD
Ronald C. Petersen, MD, PhD Washington University School of Allen Roses, MD
Mayo Clinic Medicine
Department of Neurology Department of Neurology Alzheimer’s Association
Rochester, MN St. Louis, MO William H. Thies, PhD
Vice President, Medical and
Members Ralph A. Nixon, MD, PhD Scientific Relations
Joseph D. Buxbaum, PhD New York University School of Chicago, IL
Mount Sinai School of Medicine Medicine
Departments of Psychiatry, Departments of Psychiatry and Cell Maria Carrillo, PhD
Neuroscience, Geriatrics and Adult Biology Director, Medical and Scientific
Development New York, NY Relations
New York, NY Chicago, IL
James W. Simpkins, PhD
Peggye Dilworth-Anderson, PhD University of North Texas Health Sam Fazio, PhD
University of North Carolina, Science Center Director, Medical and Scientific
Chapel Hill Department of Pharmacology and Relations
School of Public Health Neuroscience Chicago, IL
Center for Aging and Diversity Fort Worth, TX
UNC Institute on Aging Paula Moore
Chapel Hill, NC Yaakov Stern, PhD Director, Medical and Scientific
Columbia University Relations
Steven H. Ferris, PhD New York, NY Chicago, IL
New York University School of
Medicine Linda Teri, PhD Consultants
Silberstein Institute for Aging and University of Washington
Zaven Khachaturian, PhD
Dementia Seattle, WA
Senior Science Adviser
New York, NY
Potomac, MD
Linda J. Van Eldik, PhD
Claudia H. Kawas, MD Northwestern University Feinberg
University of California, Irvine School of Medicine
Department of Neurology Department of Cell and Molecular
Department of Neurobiology and Biology
Behavior Chicago, IL
Irvine, CA
Alzheimer’s & Dementia 3 (2007) 261

Editorial

Zenith Award Program of the Alzheimer’s Association


The difficulty of finding funds to sustain the expansion of As the major private funding entity in the U. S., the
Alzheimer’s research at a reasonable rate above inflation is Alzheimer’s Association has historically faced the same
not a new problem for the field. Thirty years ago while challenges as the NIA. Like the NIA, the Alzheimer’s As-
developing the nascent fields of brain aging and dementia sociation seeks to develop programs that are attractive to po-
research at the National Institute on Aging (NIA) at the tential donors while addressing the needs of the medical and
National Institutes of Health (NIH), the two perennial chal- scientific programs of the Association. The Zenith Award was
lenges I faced were (1) how to obtain set-aside funds from created by the Alzheimer’s Association in 1991 as a unique
U. S. Congress (which are specifically targeted appropria- program intended not only to attract major gifts and donors but
tions for Alzheimer’s research) and (2) how to attract new also to provide special recognition and support to established
talent with fresh ideas to the field of Alzheimer’s research. investigators with a track record of creativity. A notice of this
Today, dementia research is confronted with the same
award appears in this issue of Alzheimer’s & Dementia, and the
crises as three decades ago but for different reasons. The
details may be found at http://www.alz.org/professionals_
current dilemma in funding stems from the extraordinary
and_researchers_11284.asp.
discrepancy between the available supply of research dol-
During the 16-year history of the Zenith Award, the list
lars and the ever-increasing demand for these funds.
During the early years of developing a national program of recipients of this special grant represents the “who’s
of Alzheimer’s research, the only successful strategy in who” of dementia research. During the same period, the
achieving the dual objectives of increasing the appropria- Zenith Award has played a crucial role in assembling an
tions for Alzheimer’s research and recruiting new investi- impressive cadre of highly dedicated donors that pledge to
gators to the field was to create new funding mechanisms. become Zenith Fellows by donating $1 million each to
The development of special programs (or funding mecha- support the Association’s scientific programs. The unusual
nisms) became the most effective tool for increasing NIA’s aspect of the Zenith Award is that the Fellows as a group
budget by providing Congress specific targets for appropri- make the final selection of recipients—selected from the entire
ations, as well as the means for advancing the Institute’s array of applicants being evaluated for scientific merit— by
programmatic goals. For example, programs such as the the normal peer-review process of the Association.
Alzheimer’s Disease Research Centers (ADRCs), the Con- The prospects for improving funding for Alzheimer’s
sortium to Establish Registries for Alzheimer’s Disease research through the NIH in the foreseeable future are dim.
(CERAD), Alzheimer’s Disease Clinical Studies (ADCS), Therefore, more than ever before, the burden of funding
Leadership and Excellence in Alzheimer’s Disease (LEAD), high risk– high payoff projects falls on the Alzheimer’s
Alzheimer’s Disease Education and Referral (ADEAR) Association. By expanding the Zenith program signifi-
center, and the Cross-Cultural and Epidemiological Studies cantly, the Association can create novel funding mecha-
Initiative were primarily responsible for the dramatic nisms that will attract major gifts to the Association, attract
growth of federal funds allocated to Alzheimer’s research. much needed talent to the field, and cultivate creative new
As an illustration, the LEAD program, at $1 million per ideas for research. In this way the Association will lend
award, increased NIA’s budget by $10 million annually and invaluable speed to our quest for a cure.
increased the visibility and prestige of Alzheimer’s research
nationally as a result of the high-caliber recipients of these
awards. At the same time, this program became a potent Zaven S. Khachaturian
way of recruiting highly talented young investigators to the Editor-in-Chief
field under the tutelage of senior scientists. E-mail address: adj@kra.net

1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.08.003
Alzheimer’s & Dementia 3 (2007) 262–265

Introduction: The Third Canadian Consensus Conference on the


Diagnosis and Treatment of Dementia, 2006*
Howard Chertkowa,b,c,d,**
a
Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis -Jewish General Hospital (Department of
Clinical Neuroscience), McGill University, Montreal, Quebec, Canada
b
Division of Geriatric Medicine, Department of Medicine, Sir Mortimer B. Davis -Jewish General Hospital, McGill University, Montreal,
Quebec, Canada
c
Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
d
Centre de recherche, Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada

In this issue of Alzheimers & Dementia, something tions, gaining approval by at least 80% of the group, which
unique will be presented—a set of wide-ranging recommen- we intend to become the core of dementia care and practice
dations developed by the entire academic community of in our country (and perhaps beyond our borders as well!).
Canada involved in clinical research and care of Alzhei- The Alzheimer’s disease academic community in Can-
mer’s disease and dementia patients. Despite constant re- ada has a tradition of developing and disseminating national
search progress on all fronts and despite ongoing contro- standards for physicians. In 1989 Dr Mark Clarfield, a
versies, these articles prove that there is a large body of geriatrician at McGill University, chaired a Canadian Con-
clinical practice regarding dementia that is commonly ac- sensus Conference on Dementia (CCCD) that led to a series
cepted and agreed on by the authorities in the field, whether of recommendations that were eventually published in the
they are from Neurology, Geriatric Medicine, Geriatric Psy- Canadian Medical Association Journal (CMAJ) [1]. In
chiatry, Neuropsychology, or Family Practice. Because of 1998 a second Canadian Consensus Conference on Demen-
the manageable size and cohesion of our clinical research tia (Second CCCD) was held under the joint direction of Drs
and academic community, it was possible in Canada to have Christopher Patterson and Serge Gauthier to deal with re-
all of our experts exchange viewpoints online and meet cent developments such as the introduction of cholinester-
together in one room. During the course of 1 year, we were
ase inhibitors. The consensus statements agreed to at the
able to hammer out an evidence-based set of recommenda-
conference were published in the CMAJ [2,3], with the
background articles supporting the statements appearing
shortly afterwards in the Canadian Journal of Neurological
*EDITOR’S NOTE: This series of papers from the Third Canadian
Consensus Conference on the Diagnosis and Treatment of Dementia will Sciences (Can J Neurol Sci 2001;28[Suppl 1]:S1–121). They
serve not only to educate family physicians, but also to further standardize have had a significant impact on the care of patients with
guidelines and broaden the research infrastructure. The utility of the exer- dementia in Canada, particularly in terms of giving guid-
cise initiated by the Canadian consortium is necessitated by prospective
ance to practicing physicians and direction to continuing
plans in several parts of the globe to conduct comprehensive population-
based initiatives to identify individuals at risk for dementia and other medical education (CME)/continuing professional develop-
memory disorders. These new super-surveys envision enrolling cohorts ment (CPD) activities. These publications have been widely
with many thousands of volunteers at a time when funding resources are disseminated to Canadian physicians and other stakehold-
diminishing. An important task facing these new initiatives will be an ers. After the Second CCCD, the Consortium of Canadian
increased reliance on general medical practitioners to augment and econ-
omize case detection and risk factor identification in large populations.
Centres for Clinical Cognitive Research (C5R) was charged
Although this is a timely and important idea for the field, the practical with the responsibility of evaluating the impact of the Sec-
applications of the concept will clearly require further deliberations and ond CCCD and organizing the next consensus conference at
validation by other groups. We encourage our readers and other interested an opportune time. The timing of the next meeting would be
individuals to submit commentaries or share their perspective on any of the
recommendations proposed by the Canadian consortium.
based on the pace and significance of advances in the field.
**Corresponding author. Tel.: 514-340-8260; Fax: 514-340-8295. Have there been sufficient advances to warrant a rethink-
E-mail address: howard.chertkow@mcgill.ca ing of diagnostic and therapeutic recommendations directed
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.012
H. Chertkow / Alzheimer’s & Dementia 3 (2007) 262–265 263

at practicing physicians? With my colleagues, I answered in Dr Howard Bergman (Joseph Kaufmann Professor of
the affirmative. On the diagnostic side, for example, there Geriatric Medicine at McGill University; President
have been substantial developments in neuroimaging. The of Canadian Geriatric Society; and director of the
potential role of homocysteine as a risk factor for Alz- Quebec Research Network on Aging)
heimer’s disease (AD) has become recognized. Should Dr Howard Chertkow (Professor of Neurology and
the recommended modest battery of laboratory investi- Geriatric Medicine at McGill University and
gations for patients with seemingly typical AD be altered past president of C5R; Chair of the Steering
because of these and other findings? The recognition that Committee)
vascular risk factors like hypertension and hyperlipid- Dr John W. Feightner (Family physician; Professor of
emia increase the likelihood of AD as well as vascular Family Practice, University of Western Ontario)
dementia offers enhanced opportunities for prevention of Dr Howard Feldman (Professor and Head, Division of
dementia. New diagnoses such as mild cognitive impair- Neurology, University of British Columbia; acted
ment (MCI) have become widely used. Mixed dementias as liaison to the Canadian Neurological Society)
seem to be growing in importance with technical ad- Dr Serge G. Gauthier (Professor of Neurology, Med-
vances in imaging. icine, and Psychiatry at McGill University; chief of
On the therapeutic side, a better understanding of the the Alzheimer Disease Research Unit at the McGill
needs of caregivers and how to support them has arisen. Centre for Studies in Aging)
Novel nonpharmacologic management modalities have Dr Nathan Herrmann (Geriatric psychiatrist; Profes-
been developed and studies, notwithstanding the inherent sor and Director of Geriatric Psychiatry, Sunny-
difficulties of research in this area. Guidelines on dementia brook Hospital, University of Toronto; acted as
and driving have been introduced. The cholinesterase inhib- liaison with the Canadian Academy of Geriatric
itors have become widely prescribed, although their utility Psychiatry)
continues to be debated. Is there still consensus that they Dr David Hogan (Brenda Stafford Foundation Chair
should be offered routinely to patients with mild to moder- and Professor in Geriatric Medicine, University of
ate AD? Do they have a role for those at a severe stage? Calgary)
Memantine has been introduced, and physicians are uncer- Dr Yves Joanette (Neuropsychology and Speech lan-
tain about its place in therapy. Although routine use of guage Pathologist; Directeur, Centre de recherche
vitamin E was not recommended, this micronutrient did de l’Institut universitaire de gériatrie de Montréal,
receive dissenting support at the Second CCCD. It has come Université de Montréal)
under attack as a therapeutic intervention. Dr Christopher J. Patterson (Professor of Geriatric
In 1998 it was believed that “the reduced risk [for AD] Medicine; McMaster University).
associated with long-term estrogen use in epidemiological
studies may provide an additional potential benefit to con- Financial and/or in-kind support was obtained from the
sider when weighing the pros and cons of estrogen therapy.” Canadian Institutes of Health Research, Fonds de la Recher-
The results of the Women’s Health Initiative Memory Study che en Santé du Québec, Alzheimer Society of Canada,
[4,5] seemingly dashed this hope. There are new data C5R, Canadian Neurological Society, Canadian College of
concerning a number of other therapies designed to pre- Family Physicians, Canadian Academy of Geriatric Psychi-
vent, delay the onset, and/or slow the progression of a atrists, Quebec Research Network on Aging, and Canadian
dementia. Geriatric Society. Pharmaceutical companies could make
There emerged a general agreement last year that another unrestricted contributions of $10,000 each and attend the
meeting had clearly become necessary to review the field consensus meeting as observers only. The total contribution
and determine the current consensus among physicians and of pharmaceutical sources to the conference was less than
other health care providers who deal with patients with a 50% of the cost of putting on the consensus conference. All
dementia. As then president of C5R, it fell to me to provide grants were received in the form of unrestricted grants, and
leadership to the process that culminated in the Third no organization or company had any input into the form or
Canadian Consensus Conference on the Diagnosis and Treat- content of the recommendations.
ment of Dementia (CCCDTD3), which was held in March The Steering Committee met frequently by teleconfer-
2006 in Montreal. The organization, planning, and structure of ence to establish the objectives and methods and plan for the
the conference adhered as far as possible to the guidelines consensus meetings and to set up working groups. Partici-
established by the AGREE collaboration (The AGREE Col- pants on the working groups were chosen on the basis
laboration: Appraisal of guidelines for research and evaluation of known expertise in dementia or a related area, reputation
[AGREE] instrument; www.agreecollaboration.org). for delivering high quality work quickly, recognition as a
A Steering Committee of nationally eminent experts in national opinion leader, and/or being acknowledged as both
different aspects of dementia was formed. Members (listed open-minded and collaborative. Participants agreed to abide
alphabetically) were the following: by the rules established by the Steering Committee for
264 H. Chertkow / Alzheimer’s & Dementia 3 (2007) 262–265

carrying out their task. Conflicts of interest were declared (I) Evidence obtained from at least one properly ran-
beforehand, with written declarations provided by all domized controlled trial
participants. (II-1) Evidence obtained from well-designed con-
The topic areas were as follows: trolled trials without randomization, or
(II-2) Evidence obtained from well-designed cohort or
Topic 1: Concept, utility, and management of MCI case-control analytic studies preferably from more
and cognitive impairment no dementia (CIND) than one center or research group, or
(pharmacologic and nonpharmacologic manage- (II-3) Evidence obtained from comparisons between
ment) (H. Chertkow, coordinator) times or places with or without the intervention.
Topic 2: Criteria for “dementia”, AD, “mixed dementia Dramatic results in uncontrolled experiments are
states” and others (Kenneth Rockwood, coordinator) included in this category.
Topic 3: Dementia diagnosis and differential diagnosis (III) Opinions of respected authorities based on clin-
of dementia for the primary care practitioner and ical experience, descriptive studies, or reports of
consultant: Clinical, laboratory, imaging, markers expert committees.
(Howard Feldman, coordinator)
Topic 4: Assessment and management of risk factors The strength of recommendations was graded by using
(genetic and environmental) and primary preven- the following system used in the previous CCCD [2,8]:
tion strategies (Christopher Patterson, coordinator)
A: There is good evidence to support this maneuver.
Topic 5: Management of mild to moderate AD (David
B: There is fair evidence to support this maneuver.
Hogan, coordinator)
C: There is insufficient evidence to recommend for or
Topic 6: Management of severe AD (Serge Gauthier
against this maneuver, but recommendations might
and Nathan Hermann, coordinators)
be made on other grounds.
Topic 7: Management of dementia with a cerebrovascu-
D: There is a fair evidence to recommend against this
lar component (Christian Bocti and Sandra Black,
procedure.
coordinators)
E: There is good evidence to recommend against this
Topic 8: Ethical issues (eg, the use of placebos in re-
procedure.
search, end of life decisions, terminal care, giving the
diagnosis) (John Fisk, coordinator) Ideally, as in the previous CCCD [8], grade A and E
Topic 9: Genetic aspects of dementia (Robin Hsiung recommendations were supported by Level 1 evidence, but
and Dessa Sadovnick, coordinators) this could not always be the case; sometimes Level 2 evi-
dence was deemed sufficient. Note that a C recommendation
After carrying out appropriate literature search(es), does not imply that the maneuver was useless or harmful;
each working group prepared background article(s) and there was simply insufficient evidence to make a stronger
discussed/agreed to a series of draft recommendations in recommendation. In the background articles, the grading
their assigned topic area. The article(s) and the recommen- and strength of supporting evidence are given.
dations were then posted on an on-line website. Feedback Participants were required to read the background arti-
on them was solicited from all participants. This process cles prepared by each working group and vote on recom-
extended for several months. mendations. All recommendations receiving at least 80%
Literature searches were carried out in both PubMed and voting support were considered to have achieved consensus.
Embase going back to 1996, the end date of the literature Those not achieving this degree of approval were either
review for the Second CCCD. These searches were supple- modified (with the amended recommendation then brought
mented by articles selected from the authors’ files. A pre- forward for further discussion and approval at the March
liminary list of potential articles was obtained through 2006 meeting) or (rarely) were noted in the results but stated
these searches. The list was reduced by two individuals to have achieved only weak consensus.
on each subcommittee to relevant articles on the basis of A 2-day meeting of all participants was held in Montreal.
methodologic quality, presentation of original data, and At that meeting a brief overview was provided by each
non-overlap with other reports. For therapy, a standard- working group. Then there was a presentation followed by
ized analysis of the quality of the evidence was under- discussion and a vote on each modified recommendation.
taken, integrating a rating of the quality of the report by Voting was open and transparent, and the process for for-
using the Jadad scale [6] and such quantitative aspects as mulating recommendations was outlined in detail before the
statistical significance and magnitude of effects (www. conference. Several recommendations had to be reworked
naturalstandards.com). to finally meet approval by the group.
The quality of the available evidence was graded (I, II, or A total of 214 separate recommendations were posted on
III) by using the following system adapted from the Cana- the website. One hundred forty-seven recommendations
dian Task Force on Preventive Health Care [7]. were ultimately approved with strong consensus on the Web
H. Chertkow / Alzheimer’s & Dementia 3 (2007) 262–265 265

or after subsequent modification and voting at the meeting. time will tell, however, whether we will improve both the
Four other recommendations achieved weak consensus. The care provided by Canadian physicians and the outcomes of
others were rejected (or modified and incorporated into patients with a dementia.
other recommendations).
The Steering Committee for the CCCDTD3 decided Author Disclosures
early in its deliberations that the work done should (1)
address the needs of family physicians and (2) address the Howard Chertkow has received support from Pfizer Can-
needs of specialists. To satisfy the first goal, a liaison with ada (Advisory Board, Speaker, Grant Recipient), Neuro-
the College of Family Physicians of Canada was estab- chem Inc. (Advisory Board), and Lundbeck Canada (Advi-
lished. A representative (Dr John W. Feightner) of the sory Board, Speaker).
College sat on the organizing committee, and several family
practitioners (Drs Martha Donnelly and Chrisopher Frank) Acknowledgments
were designated by the College to attend the meeting. Fur-
thermore, a family practitioner sat as an adviser on each My lab support staff of Victor Whitehead, Shelley So-
working group. That physician was charged with reviewing loman, Randi Pilon, and Kathy D’Souza provided invalu-
the recommendations from a family practice perspective to able support at all stages of this process. Dr. Anca Raus
ensure their relevance and practicality. It is hoped that this carried out many of the literature services. MedPlan Com-
will help ensure the relevance and practicality of the rec- munications Inc. provided outstanding meeting and web
ommendations agreed to. support for this process, including a sophisticated online
Translating, disseminating, and implementing our rec- voting platform.
ommendations received particular emphasis. During the last
half-day of the meeting, a session chaired by Dr Morris References
Freedman focused on how to further our goals in the area of
knowledge translation. At this session Dr Freedman was [1] Clarfield M. Assessing dementia: the Canadian Consensus. CMAJ
elected to chair an implementation committee that will fo- 1991;144:851–3.
cus on disseminating the final set of recommendations. [2] Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW, Feldman
Work is being done at a number of levels: H, et al. The recognition, assessment and management of dementing
disorders: conclusions from the Canadian Consensus Conference on De-
1. Publication of the 18 background articles in this issue mentia. CMAJ 1999;160(Suppl):S1–15.
[3] Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW,
of Alzheimer’s & Dementia.
Feldman H, et al. Canadian Consensus Conference on Dementia: a
2. Development of a series of five case-based articles for physician’s guide to using the recommendations. CMAJ 1999;160:
the CMAJ that would highlight the most important 1738 – 42.
recommendations. These will focus on (1) risk factors [4] Shumaker SA, Egault C, Rapp SR, Thal L, Wallace RB, Ockene JK, et
and prevention of AD, (2) MCI, (3) investigating al. Estrogen plus progestin and the incidence of dementia and mild
cognitive impairment in postmenopausal women: the Women’s Health
dementia, (4) mild and moderate dementia, and (5)
Initiative Memory Study: a randomized controlled trial. JAMA 2003;
severe dementia. 289:2651– 62.
3. Development of a tool kit/practice aids for distribu- [5] Shumaker SA, Legault C, Kuller L, Rapp SR, Thal L, Lane DS, et al.
tion to Canadian family practitioners. Conjugated equine estrogens and incidence of probable dementia and
4. Production of course material for distribution to peer mild cognitive impairment in postmenopausal women: Women’s
Health Initiative Memory Study. JAMA 2004;291:2947–58.
and specialist teachers across Canada. The hope is
[6] Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan
that this material will be used for small group, inter- DJ, et al. Assessing the quality of reports of randomized clinical trials:
active educational activities developed for Canadian is blinding necessary? Control Clin Trials 1996;17:1–12.
practitioners. [7] Woolf SH, Battista RN, Anderson GM, Logan AG, Wang E. Assessing
5. Assessment of the impact of our recommendations on the clinical effectiveness of preventive maneuvers: analytic principles
and systematic methods in reviewing evidence and developing clinical
clinical practice and patient outcomes.
practice recommendations—a report by the Canadian Task Force on
These activities of the implementation committee are the Periodic Health Examination. J Clin Epidemiol 1990;43:891–905.
[8] Patterson C, Gauthier S, Bergman H, Cohen C, Feightner JW, Feldman
currently underway. H, et al. The recognition, assessment and management of dementing
In conclusion, the CCCDTD3 was a time-consuming but disorders: conclusions from the Canadian Consensus Conference on
a rewarding enterprise for all those who participated. Only Dementia. Can J Neurol Sci 2001;28(Suppl 1):S3–16.
Alzheimer’s & Dementia 3 (2007) 266 –282

Review Articles

Mild cognitive impairment and cognitive impairment, no dementia:


Part A, concept and diagnosis
Howard Chertkowa,b,c,d,*, Ziad Nasreddinee, Yves Joanetted,f, Valérie Droletd, John Kirkb,
Fadi Massoudg, Sylvie Bellevilled,h, Howard Bergmana,b
a
Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital,
McGill University, Montreal, Quebec, Canada
b
Division of Geriatric Medicine, Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal,
Quebec, Canada
c
Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
d
Centre de recherche, Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
e
Département de médecine, service de neurologie, Hôpital Charles LeMoyne and Université de Sherbrooke, Montreal, Quebec, Canada
f
Faculté de Médicine, Université de Montréal, Montréal, Quebec, Canada
g
Service de Gériatrie, Centre Hospitalier de l’Université de Montréal, and Département de Médecine, Université de Montréal, Montréal,
Quebec, Canada
h
Department de Psychologie, Université de Montréal, Montréal, Quebec, Canada

Abstract Mild cognitive impairment (MCI) and cognitive impairment, no dementia (CIND) are contro-
versial emerging terms that encompass the clinical state between elderly normal cognition and
dementia. This article reviews recent work on the classification of MCI and CIND, their prognosis,
and diagnostic approaches and presents evidence-based recommendations approved at the meeting
of the Third Consensus Conference on the Diagnosis and Treatment of Dementia held in Montreal
in March 2006. New short tools such as the Montreal Cognitive Assessment are making it easier for
family physicians to confidently attach the label of MCI.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Mild cognitive impairment; Alzheimer’s disease; Dementia; Diagnosis; Cognitive impairment, no dementia

1. Introduction In this article we will review the concept of MCI as well as


cognitive impairment, no dementia (CIND), their prognosis,
Dementia, mild cognitive impairment (MCI), and normal the search for useful prognostic markers that predict pro-
aging represent a continuum of cognitive states encountered gression to dementia, and current developments in treatment
in the elderly. As our ability to treat dementia such as of MCI. The recommendations presented here were ap-
Alzheimer’s disease (AD) improves, there is corresponding proved by at least 80% of the votes cast by the 45 experts
increase in our interest in mild cognitive decline that might who attended the CCCDTD3.
presage AD and dementia. This interest is motivated by
patient requests for prognosis and treatment. Treating phy-
sicians are currently at sea in a wealth of research informa- 2. Methods
tion and a dearth of practical suggestions for patient man-
An evidence-based review was carried out to search for
agement. This report constitutes a background article for the
references for CIND and MCI by using the terms “Ques-
Third Canadian Consensus Conference on the Diagnosis
tionable dementia” OR “Age-associated cognitive decline”
and Treatment of Dementia (CCCDTD3), held March 2006.
OR “Cognitive impairment” OR “Cognitive impairment no
Dementia” OR “Mild Cognitive Impairment” as primary
*Corresponding author. Tel.: 514-340-8260; Fax: 514-340-8295. search terms. These were combined with the secondary
E-mail address: howard.chertkow@mcgill.ca search terms “concept,” “criteria,” “diagnosis,” “therapy,”
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.013
H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 267

or “management.” Searches were carried out in both 3. CIND, MCI, and transition cognitive states of the
PubMed and Embase going back to 1996 (the last date of elderly
the Second Canadian Consensus Conference on Treatment
The Canadian Study of Health and Aging (CSHA) has
of Dementia searches). This search was supplemented by
estimated the prevalence of dementia to be 8% in the pop-
articles in the authors’ files. The articles chosen were broken
ulation older than age 65 years [3]. The best estimates of
down further into articles dealing with (1) the concept,
lifetime risk of AD range from 14.5% to 26.2% of the
prognosis, and pathology of the entities, (2) diagnosis, (3)
population [4]. With the accepted criteria of McKann et al
nonpharmacologic therapies, and (4) pharmacologic thera- [5], most physicians feel fairly confident now in diagnosing
pies. A preliminary list of 1,729 potential articles was ob- AD and dementia. Significant impairment in two or more
tained through this search. The list was reduced to 314 domains of cognition, significant impairment in day-to-day
articles on the basis of methodologic quality, presentation of functional activities, and lack of other explanations for the
original data, and non-overlap with other reports. These decline are the mainstays of dementia diagnosis. The main
included 80 articles regarding diagnosis, 28 regarding phar- limitation in diagnosing dementia is now finding the requi-
macologic therapy, 16 related to nonpharmacologic therapy, site time to carry out the assessment!
and the rest related to concept, natural history, and progno- What is more challenging are the patients who do not
sis of the terms being considered. have dementia, but who are not “normal”. First, the concept
For therapy, a standardized analysis of the quality of the of “normality” in an elderly person is itself controversial.
evidence was undertaken, integrating a rating of the quality Some researchers stress that maintenance of the same level
of the report by using the Jadad scale [1] and such quanti- of performance shown by a young person is the goal, and
tative aspects as statistical significance and magnitude of one definition, that of age-associated memory impairment
effects (see also www.naturalstandard.com). (AAMI) [6], compares the performance of elderly subjects
Recommendations were rated on the basis of rules of with that of younger persons. Up to 80% of individuals in
evidence developed by the Canadian Task Force on the their 80s will fall into the AAMI category by demonstrating
periodic health examination [2]. Ratings for recommenda- memory performance at least 1 standard deviation (SD)
tions were ranked at the following levels: below mean test values for younger subjects [7]. Longitu-
(I) Evidence obtained from at least one properly random- dinal follow-up shows this group to be quite heterogeneous,
ized controlled trial containing individuals preserving their cognition and a sub-
(II) group deteriorating toward dementia [8]. The argument that
such a label captures a legitimate target group for drug
(1) Evidence obtained from well-designed controlled tri- therapies that prevent cognitive decline [9] has proved con-
als without randomization, or troversial. In essence, many of the individuals falling within
(2) Evidence obtained from well-designed cohort or the AAMI group are normal. This underlines the majority
case control analytic studies, preferably from more view that normality must be determined with respect to a
than one center or research group, or particular age group. Cognitive aging is thus considered a
normal phenomenon and not a pathologic condition. An
(3) Evidence obtained from comparisons between times
older individual whose memory performance and day-to-
or places with or without the intervention. Dramatic
day function is “about the same as others his or her age”
results in uncontrolled experiments are included in
[10] is arguably normal.
this category.
Note that normality does not require total absence of
disease (three fourths of normal elderly subjects take a
(III) Opinions of respected authorities based on clinical medication, for instance) but performance at the same level
experience, descriptive studies, or reports of expert com- as others their age. This assessment is largely subjective and
mittees. is influenced by the nuance of questions. When asked “do
Recommendations are graded as follows: you have memory problems?”, one study found that 67% of
elderly subjects replied in the affirmative. When the ques-
A: There is good evidence to support this maneuver. tion was rephrased “Do you have memory problems relative
B: There is fair evidence to support this maneuver. to most people your age?”, only 6% replied positively [10].
C: There is insufficient evidence to recommend for or Retesting elderly on memory tests during a period of 2 years
against this maneuver, but recommendations might will reveal that about one third of them are exhibiting some
be made on other grounds. subtle but measurable memory decline over time [11]. On
D: There is a fair evidence to recommend against this the revised Wechsler Adult Intelligence Scale IQ scale, an
procedure. 85-year-old can achieve an IQ score of 100 by answering
E: There is good evidence to recommend against this only half as many questions correctly as a 21-year-old!
procedure. Hence, to claim that an elderly person shows “cognitive
268 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

impairment,” it is important to be comparing that person appeared quite heterogeneous [18]. A subclassification of
with a group of individuals similar in age. CIND included amnestic 25.1%, vascular cognitive impair-
While supporting this concept of normal cognitive aging ment 18.1%, psychiatric 17.2%, neurologic 7.3%, medical/
in terms of clinical assessment, it must be noted that there toxic metabolic 3.5%, mixed 7.6%, and not specified 19.0%.
are recent results that call this notion into serious question. Another drawback is that there is no agreement on how the
Sliwinski et al [12] derived measures of normal cognitive cutoff scores should be set or which tests should be used.
aging from a study group, but they then followed those The Kungsholmen Project [19] used the label CIND, but
individuals for many years, during which a significant por- they based the diagnosis on scoring at least 1 SD below the
tion of their normal elderly group progressed to MCI or age and education specific mean on the Mini-Mental State
dementia. They examined the effects of preclinical demen- Examination (MMSE) score derived from a normative
tia on estimates of normal cognitive function in the aged. group [20].
After excluding preclinical dementia cases, they found that A different approach has been used in the Washington
many changes previously deemed normal cognitive aging University longitudinal cohort study of memory loss. These
simply disappeared. The results indicate that by failing to individuals were referred to an Alzheimer Disease Research
exclude preclinical dementia, conventional normative stud- Center and classified according to severity of decline as-
ies underestimate the mean, overestimate the variance, and sessed by a semistructured interview of the client/patient
overestimate the effect of age on cognitive measures. With and the caregiver, scored in a scale, the Clinical Dementia
a different approach, Bennett et al [13] assessed 134 elderly Rating scale, developed in 1982 [21]. Degree of ability in
individuals in a longitudinal study who had been classified areas such as memory, orientation, social and functional
as normal and who soon thereafter died and came to au- activities, and hobbies were assessed and combined to give
topsy. They found that more than one third met National a single numeric score. In this scale, 0 indicated normal
Institute on Aging (NIA)–Reagan pathologic criteria for function with no loss, 1.0 indicated significant loss almost
intermediate likelihood of AD, and these individuals actu- always diagnostic of dementia, and a transition level, 0.5
ally scored about one fourth standard unit lower on tests of (termed questionable dementia), indicated some very mild
episodic memory. Again, cognitive aging was being driven loss that might or might not be classified as dementia. It has
by preclinical AD. been pointed out, however, that you can be CDR 0.5 and
In the CSHA, subjects in a population study were cate- nevertheless meet diagnostic criteria for AD and dementia,
gorized as no cognitive loss, CIND, or dementia [3,14]. thus making this grouping significantly different from MCI.
CSHA defined CIND on the basis of a consensus conference What is important to note is that primacy is given to a
of physician, nurse, and neuropsychologist, integrating all detailed and expert history taken from patient and family,
available information from clinical and neuropsychological focusing on sometimes subtle features of decline and loss
assessment. Mild objective memory problems falling be- (rather than a particular cutoff score on any test) as the
tween these two poles of normality and dementia are a critical feature. Morris et al [22] have argued that this
common phenomenon in older people; the CSHA docu- approach is valid and accurate and takes into account an
mented a 16.8% prevalence of cognitive impairment with- individual-oriented concept of degenerative disease that is
out dementia in the elderly [15,16]. Individuals falling into more sensitive than neuropsychological testing. Many (but
this category exhibited an increased risk of death and even- not all) CDR 0.5 subjects would be classified as MCI. The
tual dementia (about 50% after a 5-year follow-up period) expertise to apply this approach might be limited to only a
[17]. CIND as a label has the benefit that inclusion depends few experts in the field.
on a conclusion simply that there is cognitive impairment, There are other global measures, such as Reisberg’s
without exclusions related to the degree of decline, presence Global Deterioration Scale [23], that also focus on “wors-
of functional impairment, or suppositions regarding under- ening” from a previous level. Most MCI individuals score as
lying etiology. The CSHA label does not appear to have Stage 3, earliest subtle deficits, on the Global Deterioration
been based solely on psychometric cutoff scores, although Scale, but it is not clear that the two concepts are inter-
psychometric tests were administered. changeable [24]. Both share the view that they are defining
A number of limitations, however, are inherent in using MCI as a predementia stage.
this term. One drawback is that an individual might be A somewhat different approach focuses on patients’ and
considered as having CIND as a result of lifelong static families’ subjective complaints of memory and cognitive
brain damage, mental retardation, schizophrenia, and a loss as the starting point. A person’s degree of complaints
range of pathologies that would be obvious to the treating might not be equivalent to their actual objective degree of
physicians. The prognosis and diagnostic approach to CIND decline, of course. Clearly, elderly subjects might complain
would not be as relevant to the treating family physician or of memory loss as a result of anxiety, dementia in family
specialist, who is focused on patients complaining of a new members or friends, or mild depression. At the same time,
decline in memory. When CIND as a label was applied to a studies show that memory complaints in elderly people
cohort derived from referral Memory Clinics, the causes should no longer be considered merely as an innocent age-
H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 269

related phenomenon or a symptom of depression. Instead, another neurodegenerative disease, such as primary pro-
these complaints deserve to be taken seriously, at least as a gressive aphasia. Alternatively, they might be exhibiting
possible early sign of actual decline that could ultimately evidence of vascular damage (now termed vascular MCI or
lead to dementia [25]. At the same time unless there is vascular cognitive impairment). We and others have found
objective supporting evidence of real memory loss, the that most subjects initially labeled amnestic single MCI by
prognosis is relatively benign [26,27]. the clinician in fact exhibit subtle deficits in other neuro-
Historically, this conception of subjective complaint of psychological and reaction time tests [36,37]. The clinician
cognitive loss, accompanied by objective supporting evi- should not be surprised when an MCI individual performs
dence of loss, was originally suggested by Levy and an slightly abnormally on placement of hands on clock draw-
International Psychogeriatric Association working group ing. The validity of these subtypes, in terms of presumed
[28]. They termed it age-associated cognitive decline different prognoses for developing AD, is quite unproven at
(AACD). Currently, the most widespread term in use to this point. The details of how a subgroup should be applied
characterize this group is MCI, derived from the World are somewhat vague, as are which tests to use and how to
Health Organization and adapted by a number of centers apply them. It is not at all clear that practicing clinicians
[29 –31]. MCI is a clinical label that includes elderly sub- need to delineate these subgroups in any formal manner, or
jects with short-term or long-term memory impairment and that these subgroups impact significantly on prognosis [38].
with no significant daily functional disability. The original In fact, there have been numerous slightly different opera-
diagnosis of MCI required subjective report of cognitive tionalized criteria that have appeared in the literature.
decline from a former level, gradual in onset, and present In our experience, use of different norms and different
for at least 6 months. This subjective complaint is supple- tests, however, can significantly alter inclusion across sub-
mented by objective evidence of memory and learning de- groups [39,40]. A recent European Consortium on AD rec-
cline, with other cognitive domains remaining “generally ommended that general medical practitioners should focus
intact” [32]. There was no clear delineation as to how the on establishing the presence of an MCI syndrome, whereas
presence of memory loss was to be established. In all cases,
specialists could focus on the second stage of establishing
this term excluded individuals with significant depression,
the MCI subgroup and the third stage of analyzing the
delirium, mental retardation, or other psychiatric disorders
etiologic subtype (ie, degenerative, vascular, traumatic) in
likely responsible for the impairment. If the memory loss
each case [41].
was severe and accompanied by significant functional im-
MCI is a controversial concept and is now the focus of
pairment, then the individual met clinical criteria for de-
natural history studies, biomarker studies, along with AD
mentia and was no longer MCI. The concept was intended
prevention studies. The MCI stage might be the optimum
to capture and classify patients who seem to have a cogni-
stage at which to intervene with preventative therapies. It
tive problem that one would be loathe to label as normal and
yet clearly is not severe enough to qualify as dementia. will increasingly become a label used by neurologists, ger-
Modifications of the MCI concept have developed during iatricians, and family physicians who treat elderly cogni-
the past decade. Although Petersen et al [31,33] stressed tively impaired individuals. Current interest is largely pred-
that MCI should be based on clinical judgment, they men- icated on the fact that patients classified as MCI have a high
tioned that most MCI subjects fell 1.5 SDs below norms on rate of conversion to dementia, particularly to AD. It is now
memory tests. Subsequently it has become usual to assign becoming clear that many MCI subjects have AD pathology
an objective level of impairment as performance either 1.0 in its earliest stages and will progress to AD if given
SD or usually 1.5 SDs below published norms for a memory sufficient follow-up. In most clinic-based studies of MCI,
test. Furthermore, there has been a push for greater speci- about 44% of individuals meeting the criteria for this diag-
ficity in terms of using three or four patterns of subgroups. nosis convert to AD during a 3-year follow-up, giving an
One subgroup would be elderly individuals whose deficits annual conversion rate of about 15% per year [42]. What is
are restricted to memory, a so-called amnestic or amnestic controversial is whether all MCI individuals progress to AD
single MCI [33,34]. Petersen et al [31] describe most MCI with time. In a 10-year follow-up in a Maastricht Memory
subjects as showing only memory impairment, and these Clinic, Visser et al [43] found the risk of progression to
amnestic MCI are more likely to go on to AD. Other dementia to be 48% in 64 MCI subjects. In one Canadian
subgroups (amnesic multiple) are characterized by impair- group of 89 MCI subjects accumulated in a Memory Clinic
ment extending beyond memory to include subtle but mea- setting, it appeared that about 25% would not progress even
surable changes in executive function, attention, naming 10 years after onset of memory problems [44]. In population-
[35]. A third subgroup would involve individuals with im- based studies, the prognosis of mild cognitive deficits seems
pairment in a single non-memory cognitive domain, while much less ominous. Ritchie et al [45] found that only 22%
memory remains normal. These could sometimes also in- of such a group went on to a degenerative dementia during
volve multiple non-memory domains as a fourth subgroup 8 years of follow-up. In contrast, Morris et al [46], with
[31]. Presumably such subjects are more likely to go on to CDR 0.5 as their target group, have argued that with suffi-
270 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

Table 1 the diagnosis and is useful for randomized clinical trials, but
General criteria for MCI it restricts the diagnosis of MCI to centers having easy
Subjective complaint of memory loss access to neuropsychological evaluation. Furthermore, as
Objective impairment of memory Morris et al [22] have pointed out, these cutoff scores lose
Generally preserved other cognitive ability
the strength of individual assessment that emphasizes de-
Preserved basic day-to-day functioning
No other obvious medical neurologic or psychiatric explanation for the cline from a previous level of performance rather than a
memory problems single test assessment.
Individual does not meet criteria for dementia Similarly, Petersen et al [31,33] had initially suggested
NOTE. Modified from Petersen and Smith, 1999. that non-memory domains be “generally intact” in MCI, and
in terms of bedside mental status testing, this is a reasonable
criterion to apply. In response to this, Ritchie and Touchon
cient time, most or all subjects in this category would [49,50] applied this notion that to satisfy the criteria for
deteriorate to dementia, specifically AD. MCI, all other domains besides verbal delayed memory
Much of the disagreement might center on different must be within 1.0 SD of the age-adjusted norm. Unfortu-
diagnostic criteria being used to assemble cohorts of sub- nately, when such constraints were firmly applied, there
jects for study and different means of collecting subjects. were few putative MCI subjects who still existed. In fact,
There are multiple different operational definitions of detailed cognitive and neuropsychological testing shows
groups presented or discussed as MCI (Table 2). Depending that a range of subtle “low normal” test results is the
on the diagnostic criteria used, the prevalence and incidence majority finding in groups of MCI individuals [51]. These
of MCI vary by a factor of 8 times. The CSHA term CIND neuropsychological test abnormalities, however, are not so
[16] would have encompassed virtually all MCI individuals, evident with the much cruder bedside testing carried out by
along with numerous subjects who would not have met the medical clinicians. Because one major goal of using the
MCI criteria (some had alcoholism, large strokes, or mental terms MCI and dementia is to allow general physicians to
retardation, for instance). It is estimated that one half to two better assess, diagnose, and treat patients they see, the di-
thirds of subjects with CIND would meet criteria for MCI. agnostic criteria might be more meaningful when viewed
But is that an improvement? In fact, is one definition supe- with respect to clinical mental status assessment and simple
rior to the others? bedside testing, rather than being dependent on more accu-
We would argue that these concepts are designed for rate (and often unavailable) neuropsychological criteria.
different goals and have different strengths. A definition like There are other problems with overreliance on quantita-
CIND that can be satisfied by objective test cutoff scores is tive criteria for diagnosing MCI. It is a fact that many
better designed for community and population studies. elderly individuals who would be rated as cognitively intact
What it lacks, however, is ecologic validity in the Memory are in fact substantially impaired on tasks that tap into a
Clinic or clinical setting. In the doctor’s office, patients are range of cognitive systems [52]. Statistically speaking, there
presenting with a memory complaint. Thus, definitions such are normal subjects who will sit for their whole lives at the
as MCI that begin with the individual’s complaint are more bottom of any gaussian curve of neuropsychological results,
likely to correspond with clinical reality for a physician and these individuals must not be falsely labeled as MCI.
[47]. There are several weak points regarding the MCI Cultural, educational, and attentional factors (ie, impaired
definition, however. One is the lack of agreed on criteria for attention as a result of stress, anxiety, or depression) can
determining objective memory loss, the key feature of MCI. also impact on neuropsychological testing. Thus, clinical
In the clinic, one might accept impaired three-word recall as judgment from a physician or psychologist is essential in
sufficient evidence of memory impairment in a hitherto high assessing whether an MCI label is warranted. Neuropsycho-
functioning individual now complaining of memory loss. logical tests can be used, however, to support such clinical
An alternative approach favored by psychological research- decision making.
ers, however, is to insist that MCI must be operationalized One critical element in the diagnosis of dementia is
to be meaningful [48]. This is generally done by including significant impairment in functional ability. Unfortunately,
only individuals exhibiting abnormal performance at least 1 there are few agreed on criteria for evaluation of function or
SD or even 1.5 SDs below the norm for age-appropriate what significant functional impairment entails. About 31%
populations on standardized verbal memory tests [32,45]. of MCI subjects likely exhibit subtle functional changes;
This attempt to establish statistical cutoff criteria for MCI, having to maintain written lists of things might well be
while well-motivated, is fraught with difficulty; who de- considered a functional alteration [53]. The key thing is that
cides which memory tests to be used, which norms, which such changes should only affect higher functions and not
cutoff point? Can memory tests be adequate for culturally represent significant impairment, although this is crudely
and educationally heterogeneous populations? Are the defined. In fact, one group defining MCI suggests that
norms reliable for a particular cultural setting? Rigorous use impairment in complex functions should be an inclusion
of psychometric cutoff points provides more reliability to criterion for MCI.
Table 2
Different criteria for MCI
Study Reference Complaint Non-memory Functional Indication of Normal Tests Used to Assess Objective Cutoffs That Were
Required? Cognitive Deficits Deficits Allowed? Intellectual Functioning Used in the Definition
Allowed?

Petersen et al [30], 1999 Y Not indicated N Y WAIS-R, WMS-R, Auditory Verbal Learning n/a
Test, Wide-Range Achievement Test-III,
MMSE, DRS, Free and Cued Selective
Reminding Test, Boston Naming Test,
Controlled Oral Word Association Test,
Category Fluency Procedures
Morris et al [46], 2001 Not indicated Y Y Not indicated CDR CDR ⫽ 0.5
Ritchie et al [49], 2001 Y N N Y Examen cognitive par ordinateur (ECO) n/a

H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282


Bennett et al [94], 2002 N Y Not indicated Not indicated Extensive neuropsychological battery Population-specific cutoff
scores used
Darby et al [122], 2002 Y N N Not indicated CERAD word list recall ⬍6
Hänninen et al [123], 2002 N Y N MMSE ⱖ20 CDR CDR ⫽ 0.5
Buschke Selective Reminding Test (total ⱖ1.5 SD below healthy
immediate recall during 6 trials ⫹ delayed subsample cutoff
recall)
Visual reproduction Test (WMS) (immediate
and delayed recall)
Logical Memory Test (WMS) (immediate
and delayed recall)
Boston Naming Test (abbreviated)
Verbal Fluency Test
Trail making Test (A⫹B)
MMSE
Kabani et al [124], 2002 Y Y N Not indicated CDR CDR ⫽ 0.5
Tests of explicit memory ⱖ1 SD below age-adjusted
norms
Larrieu et al [99], 2002 Y Y Y MMSE ⬍1 SD from Benton Visual Retention test ⱖ1.5 SD below age &
age & education education adjusted means
adjusted cohort
means
Busse et al [125], 2003 Examined as a Y N Not indicated Structured Mental Status (SIDAM) ⬎1 SD below age & education
clinical variable associated norms on subtests
of SIDAM
Fisk and Rockwood [63], 2003 Examined as a N Examined as a Y Benton Visual Retention test; Buschke Cued No cutoffs set; diagnosis based
clinical variable clinical variable recall test, Digit Span, and auditory verbal on clinical consensus
learning test; WAIS-R Similarities,
Comprehension, Digit Symbol, Block
design; Token test; COWA; Animal
naming.
Lambon et al [126], 2003 Y N Y MMSE ⬎24 Logical Memory (immediate) ⱖ1.5 SD below norms
DRS (memory)

271
Table 2

272
Continued
Study Reference Complaint Non-memory Functional Deficits Indication of Normal Tests Used to Assess Objective Cutoffs That Were
Required? Cognitive Deficits Allowed? Intellectual Functioning Used in the Definition
Allowed?

Lopez et al [127], Necessary for Y Y Not indicated n/a ⱖ1.5 SD below a sample of
2003 “probable” MCI, 250 unimpaired subjects
but not
“possible” MCI
Farlow et al [128], — Y Y Not indicated CDR CDR ⫽ 0.5
2004 (InDDEx NYU Paragraph recall ⬍9
study) HAM-D ⬍13
HAM-D item 1 ⱖ1
Feldman et al [129], N Not indicated Not indicated Not indicated CDR CDR ⬍1

H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282


2004 NYU paragraph recall ⬍9
Ganguli et al [34], Y Not indicated N MMSE ⬎24 CERAD 10 word list delayed recall ⬎1 SD below mean of cohort
2004
Grundman et al [130], Y Not indicated Y MMSE ⱖ24 Logical Memory II ⱖ8 (⫹16 y education)
2004 (ADCS-MIS Hamilton Dep. Rating Scale ⬍4 (8 to 15 y education)
study) ⱖ2 (0 –7 y education)
⬎12
Royall et al [37], 2004 Not indicated Examined as a Y Not indicated MMSE, CLOX 1,2 (clock drawing test), COWA ⱖ1.5 SD below average score
clinical variable (verbal fluency), EXIT 25 (executive of the sample on each
interview), Trail A ⫹ B, CVLT (verbal measure
memory), DRS: MEM
Solfrizzi et al [131], N Y Y MMSE ⬍1.5 SD from Babcock Story Recall Test Score in the lowest 10th
2004 age & education percentile of the cohort
adjusted means
DeJager et al [132], N N Y Not indicated CERAD 10-word list (free and delayed recall), ⱖ1.5 SD below norms
2005 Verbal Paired Associates (cued recall test),
Pattern and spatial recognition from the
CANTAB, CANTAB Paired Associates
Learning (6-items), The Placing Test (TPT)
Devanand et al [133], Y Y Y MMSE ⱖ22 MMSE Delayed recall ⱖ2 of 3 items recalled
2005 or or ⱖ1 SD below norms
if Spanish speaking Selective Reminding Task ⱖ10 points below WAIS-R
with education ⬍5 y or verbal IQ score
MMSE ⱖ18 WAIS-R performance IQ score If no deficits on objective tests,
memory complaint ⫹
informant’s confirmation of
decline and functional decline
Geslani et al [134], Y Not indicated N WAIS-R similarities or CVLT ⱖ1.5 SD below age & sex
2005 digit symbol ⬍0.5 adjusted norms
SD from mean
Kumar et al [135], Y Not indicated N MMSE ⬎25 CVLT ⬍2
2005
Nasreddine et al [112], Y Y Y Y Rey Auditory Verbal Learning Test At least 1 SD below norms
2005 2 subtests of WMS
(delayed visual reproduction and logical
memory)
Table 2
Continued
Study Reference Complaint Non-memory Functional Deficits Indication of Normal Tests Used to Assess Objective Cutoffs That Were Used in the
Required? Cognitive Deficits Allowed? Intellectual Functioning Definition
Allowed?

H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282


Purser et al [136], 2005 N Y Examined as SPMSQ ⬍4 20-item immediate word recall test ⬍4
clinical variable
Visser et al [137], 2005 Not indicated Y Y Not indicated Global Deterioration Scale 3
Blessed Dementia Rating scale ⱖ0.5 on first 8 items if GDS ⫽ 2
20-item story recall Cutoff based on centile score estimations
for LM, RAVLT, NYU paragraph
recall
Gal Int-11 Not indicated Y Y Not indicated CDR CDR ⫽ 0.5
(as described in Visser et al NYU Paragraph recall ⬍11
[137], 2005)
Ampakine study (as Not indicated Y Y MMSE ⬎24 CDR CDR ⫽ 0.5
described in Visser Logical Memory II ⱕ12 (⫹16 y education)
et al [137], 2005) Geriatric Dep. Scale ⱕ4 (8 to 15 y education)
ⱕ2 (0 –7 y education)
⬎6
Piracetam study (as Not indicated Y Y Not indicated CDR CDR ⫽ 0.5
described in Visser et al Logical Memory I ⬍10
[137], 2005) Logical Memory II ⱖ5 pts below LMI
HDRS ⬎17
Rofecoxib Not indicated Y Y MMSE ⬎23 CDR CDR ⫽ 0.5
study (as described in Visser RAVLT total learning ⬍38
et al [137], 2005) HDRS ⬍12
Verghese et al [138], 2006 Y Y N Verbal IQ ⬎84 Blessed test 5-item memory phrase ⱖ3 errors (ⱖ1.5 SD below norms)

Abbreviations: CANTAB, Cambridge Neuropsychological Test Automated Battery; COWA, controlled oral word memory test; CVLT, California Verbal Learning Test; DRS: MEM, Dementia Rating
Scale: Memory Subscale; GDS, Global Deterioration Scale; HAMD, Hamilton depression scale; HDRS, Hamilton Depression Rating Scale; LM, logical memory; RAVLT, Rey Auditory Verbal Learning
Test; SPMSQ, Short Portable Mental Status Questionnaire; WAIS-R, revised Wechsler Adult Intelligence Scale; WMS-R, Wechsler memory scale–revised.

273
274 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

There are many diagnostic issues that arise in character- related to memory loss. This up-regulation was speculated
izing MCI. Should patients with mild depression be ex- to be compensatory in nature [61]. This is important infor-
cluded from consideration? Some studies find depression in mation in gauging the likelihood that MCI subjects will
up to 60% of individuals with MCI who go on to AD, and respond to treatment with cholinesterase inhibitors.
presence of depression might in fact be a useful prognostic
sign in MCI individuals [54]. For the above reasons, it is
5. Prognosis in MCI
usual to encounter subtle but significant heterogeneity in
MCI patients [55]. MCI is perhaps best viewed as a some- Is it possible to delineate at initial presentation which
what heterogeneous clinical syndrome that includes many MCI subjects are most likely to progress to AD during a
subjects in a dementia “prodrome state” who might some limited period of time such as 5 years? Numerous attempts
day be distinguishable as such. have been made to delineate prognostic markers in MCI,
most involving rather small samples of subjects followed
4. The Neuropathology of MCI for limited periods of time. Daly et al [62] studied 123
subjects recruited through the Boston media who all met
Part of the confusion concerning MCI is based on our criteria of being CDR 0.5 on the CDR Scale. After 3 years
evolving understanding of the brain processes occurring in of follow-up, only 19% had progressed to dementia (AD),
the condition. It is clear that pathologically at least, some of 2% progressed as a result of strokes, and 5% had become
the MCI subjects are showing the very earliest stages of AD normal. It was found that most of their group was milder in
[56]. This is characterized by neurofibrillary tangles, usually terms of cognitive deficits than other studies, and that this
in the hippocampus as well as the entorhinal cortex. When explained the lower progression/conversion to AD rate.
AD ensues, the pathology has spread to the lateral temporal Daly et al found that whereas 4% of subjects scoring a CDR
cortex as well. Senile plaques occur only later on. Markes- sum of boxes of 1.0 (almost normal cognition) went on to
bery et al [57] reported the findings of 10 individuals dying progress, 67% of those with a total box score of 3.0 (almost
with amnestic MCI, compared with AD and normal con- demented already) progressed to AD. This demonstrated a
trols. The group showed increased neocortical neuritic not surprising effect of severity; within the broad range of
plaque density compared with normal controls, virtually as individuals labeled with mild memory loss, those most
severe as the AD brains. A study of individuals with CDR impaired to begin with were more likely to progress during
0.5 impairment showed similar findings [46]. These findings follow-up.
fit with the notion that many and indeed most MCI individ- A study by Fisk et al [63] has called into question the
uals will go on to develop clinical dementia caused by AD. need to define MCI with subjective as well as objective
The problems with this conclusion are three-fold. First, it is measures. With a variety of MCI definitions and subjects
known that some individuals who are cognitively normal derived from the CSHA, they found that changes of defini-
will also show AD pathology at autopsy [58]. In the study tion changed the prevalence of MCI but did not change the
by Knopman et al [58] of 39 cognitively normal elderly proportion who progressed to dementia during a period of 5
individuals, four met NIA-RI criteria for intermediate like- years, which remained about 8% to 10% per year. In addi-
lihood of AD, seven cases met Consortium to Establish a tion, all groups included a subgroup who had improved after
Registry for Alzheimer Disease (CERAD) criteria for pos- 5 years.
sible AD, and 19 met Khachaturian criteria for AD. Simi- A number of studies have looked at the impact of APOE
larly, Bennett et al [59] found that between one third and genotype on risk of developing AD and progression of
one half of normal elderly brains had a significant amount of cognitive decline. The results are rather complex. APOE e4
AD pathology. Second, it is also likely that MCI individuals genotype has been found to be a significant risk factor for
with a more benign outlook will not be presenting for CIND in the Canadian ACCORD study [64]. In several
autopsies. There is no way to ascertain that those presenting studies the presence of APOE e4 in cognitively impaired
for autopsy are representative of the entire group. Finally, a elderly subjects was also associated with a greater chance of
clinical-pathologic study of MCI from the Religious Orders memory decline during a period of 3 years [65,66]. Petersen
Study [59] showed that individuals dying with MCI were et al [32] suggested that the MCI individual’s APOE status
intermediate between normal and AD not only in terms of might be useful in prognostication. In their study, Petersen
plaques and tangles but also in terms of cerebral infarcts and et al found a major effect of APOE e4 load on progression
Lewy body pathology. This stresses the nonspecificity of to AD from MCI. In other studies, APOE genotype ap-
MCI in terms of dementia etiology. peared to have little impact on the degree of decline [67].
Of great interest now that MCI brains are being sec- A series of studies have looked at other predictors of
tioned, there appears to be no real increase in choline acetyl- cognitive decline (ie, changing from normal to MCI) or
transferase (ChAT) activity in the cortex at the MCI or early progression (from normal or MCI) to AD/dementia. One
AD stage [60]. In one recent study, levels of hippocampal recent study targeted high homocysteine levels as another
ChAT activity were increased in MCI subjects, although not biologic risk factor, but this remains to be confirmed [68].
H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 275

High levels of low-density lipoprotein or a high ratio of velop AD a good number of years before the diagnosis is
low-density lipoprotein/high-density lipoprotein were asso- officially made [91]. (2) Individuals with MCI or CIND are
ciated with increased risk of later diagnosis of AD in the at increased risk of dementia [92–95]. (3) At the same time,
Baltimore Longitudinal Study [69]. Serum markers for in- a nontrivial percentage of MCI individuals do not progress
flammation such as interleukin 6 have also been found to be to dementia and even appear to become normal over time
associated with cognitive decline in healthy elderly men and [62,92,94,95].
women [70]. To what extent these biochemical markers,
alone or together, can be used in individual prognostication
remains to be elucidated.
6. Conclusions regarding the concept of MCI
Imaging markers might be useful in prognostication in
MCI, but these have mainly been used in academic research MCI as a clinical entity continues to be debated. Morris
settings. On magnetic resonance imaging scans, the formal et al [46,96] argue that there is no such independent entity
measurement of hippocampal volumes [71] and the rate of as MCI, simply AD at various levels of severity, with some
change of global or hippocampal volumes [72,73] are pow- individuals who have AD not even meeting criteria for
erful tools to predict progression to AD. Even earlier alter- dementia. What will change in the future, they argue, is the
ations in entorhinal cortex can be detected with positron threshold at which we are willing to attach a diagnosis of
emission tomography [74,75], and these can be potentially AD based on clinical information and biomarkers.
useful in predicting progression to AD even when cognition Another approach is to view MCI as a high risk group for
is normal [76]. Although visual inspection of standard sin- both death and development of dementia in the future [97].
gle photo emission computed tomography (SPECT) brain This latter underlines the fact that pathologically, many
images is not useful in prognostication [77], the sophisti- MCI individuals already have common conditions respon-
cated quantification of SPECT images might allow fairly ac- sible for age-related dementia, including AD, Lewy body
curate prognostication [78]. Promising reports have emerged disease, and cerebral infarction [59].
with use of magnetic resonance spectroscopy [79] in predicting A rather different perspective emerges from several com-
onset of AD in patients with MCI. Altered patterns of positron munity studies of MCI. In a Pennsylvania study of the
emission tomography activation can be detected during atten- elderly, 4% were found to meet operational criteria for MCI.
tional tasks [80] and with functional magnetic resonance im- About one fourth of these reverted to normal during 10
aging during memory tasks in MCI individuals [81]. These years of follow-up [34]. Similarly, a French population
could potentially be used for prognostication. Imaging remains study found the MCI diagnosis to be remarkably unstable,
a very active field of diagnostic investigation that should pro- with a significant proportion reverting to normal during
vide helpful indicators in the coming years. follow-up [98]. In a cohort of MCI individuals derived from
Neuropsychological and cognitive measures have been the CSHA, fully one third were considered to have no
assessed for their predictive capacity. It is unclear whether cognitive impairment after 5 years. In the PAQUID study in
cognitive tests are robust enough to allow prediction at the France, a similar instability of diagnosis and high reversion
individual patient level, as opposed to large subject groups. rate to normal were noted [99]. Ritchie and Touchon [50]
At the same time, MCI individuals who complain, exhibit concluded that it would be erroneous to consider MCI as a
global cognitive impairment, and exhibit episodic memory clear early stage of AD or dementia.
deficits are at highest risk for progression to AD [82]. We This view is not limited to population studies, however.
have found that MCI subjects with any disorientation to Heterogeneity in the prognosis of MCI during the long term
time, or even subtle problems with clock drawing, are more emerges in cohorts collected from Memory Clinics in both
likely to progress to AD [44]. In fact, the presence of any Europe [100] and Canada [101]. The MCI diagnostic cate-
quantifiable abnormalities beyond memory constitutes a risk gory appears to contain many individuals who will ulti-
for progression from MCI to dementia [83]. Batteries of mately progress to AD (or other dementias), along with a
different formal neuropsychological tests have been used subgroup, usually about one fourth of patients seen in Mem-
together with the objective of stratifying risk of progression ory Clinic settings, who will not progress.
to AD [84 – 87]. Tests of delayed verbal recall and executive Should MCI be considered as a distinct entity? The
function appear to have the best discriminating power for construct of MCI has been criticized as being difficult to
prediction. Although promising, these tests are not suffi- operationalize [102,103]. Some authors insist that better
ciently robust to prognosticate in all MCI cases. approaches must be found, perhaps on the basis of change
Currently a long series of other biomarkers are under scores and measures of cognitive decline over time
investigation: smell testing [88], blood heme oxygenase 1 [102,104]. Some have urged criteria to be developed for
[89], CSF-tau, and A beta 42 [90] to note only a few. “prodromal/predementia Alzheimer Disease” [95,105]. The
What the majority of such studies conclude is largely conclusions of different groups stand in considerable oppo-
consistent and consists of three findings. (1) There appear to sition; work by Morris et al [46] suggests that individuals
be notable and measurable deficits in individuals who de- with MCI already have mild dementia, whereas population
276 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

studies and clinic studies of others stress the heterogeneity dementia and death that has been documented
and unpredictability of the outcome of MCI subjects. (Grade B, Level 2).
There is a danger, however, of “throwing out the baby
with the bath water.” Attaching a diagnostic label to indi-
viduals who are neither (in the opinion of their physician) 8. Diagnosis of MCI and screening for MCI by
normal nor demented accurately captures the clinical di- physicians
lemma and uncertainty [106]. We know enough to advise
such patients on increased risk, arrange for closer follow-up, The question as to how physicians should approach the
and even investigate for underlying causes of memory loss. diagnosis of MCI or CIND is an important one. Is there an
The group has already been targeted for possible secondary agreed or accepted recommended way to diagnose MCI, or
prevention therapy [107–109]. This, it would seem, is pref- does the approach vary depending on the definition used, eg,
erable to the alternative, mislabeling them as normal and MCI vs CIND? In 2001, the Quality Standards Subcommit-
misinforming the families as to a benign prognosis. tee of the American Academy of Neurology recommended
that MCI should be detected. There was sufficient evidence
suggesting that subjects with MCI were at increased risk for
7. Recommendations approved at the CCCDTD3 developing dementia or AD when compared with similarly
meeting regarding concept of MCI: aged individuals in the general population. Screening in-
struments such as the MMSE were found useful, as were
1. Physicians should be aware that most dementias neuropsychological batteries [31].
might be preceded by a recognizable phase of mild Until 2001, there were no specific cognitive screening
cognitive decline. Physicians should be familiar with instruments to detect MCI. Although the MMSE was con-
the concept of MCI (or CIND) as a high risk state for sidered useful, it had low sensitivity to detect MCI because
decline and dementia (Grade B, Level 3). most subjects scored in the normal range on the test [33].
2. There is currently inadequate evidence to recommend Since that time, several screening instruments have been
one term or label (MCI, CIND) over another (Grade developed to help screen for MCI (Table 3). We will look at
B, Level 3). the instruments currently available, validated in MCI,
3. There is inadequate evidence to advise MCI patients CIND, and CDR 0.5.
and their families that the patient is already exhibiting The following screening instruments have shown prom-
signs of dementia or to treat MCI as equivalent to ising results with well-designed studies based on the MCI
dementia (Grade C, Level 2). criteria of Petersen et al [33]:
4. There is fair evidence that physicians should The DemTect [110] evaluates immediate and delayed
closely monitor individuals who have MCI or recall of a word list, number transcoding, verbal fluency,
CIND because of the known increased risk of both and reverse digit span. It was validated with 363 English-

Table 3
Brief cognitive screening tests for detection of MCI
Test First Author Community vs Subjects, Normal/MCI Cognitive Time to Administer Sensitivity/Specificity
Memory Clinic Domain(s) (min) (%)

CWL Shankle [118], 2005 Memory clinic and 119/95 Memory 15–20 (estimate) 82/91, 94/89 with CA
community statistical technique
MoCA Nasreddine [112], 2005 Memory clinic 90/94 Multiple 10 90/87
DemTect Kalbe [110], 2004 Memory clinic 145/97 Multiple 8–10 80/92
CMC Xu [115], 2002 Memory clinic 351 subjects. Multiple ⱕ15 (estimate) 83/80
retrospective design
RI-48-Adams Ivanoiu [111], 2005 Memory clinic 38/25 Memory 20–25 77/100
CDT Yamamoto [113], 2004 Memory clinic 41/48 Clock only ⱕ5 (estimate) 75/76
ECO Artero [116], 2003 Memory clinic 60/308 Multiple 5–10 73/99
Extended delayed Loewenstein [48], 2000 Memory clinic 52/24 Memory ⱕ20 (estimate) 70/84
recall MMSE
Mini-Cog Borson [119], 2005 Memory clinic 140/71 Multiple 3 55/83
STMS Tang-Wai [117], 2003 Memory clinic 788/29 Multiple ⱕ10 (estimate) AUC, 0.82; ⬍80/⬍80
Letter F and Canning [121], 2004 Memory clinic 46/25 (CIND) Language 3 (estimate) F: 48/57, Animal: 44/96
Animal fluency
6-item screener Callahan [120], 2002 Memory clinic and 708/44 Memory and 1.5 97/49
community orientation
ABCS Molloy [114], 2005 Memory clinic 111/124 Multiple 3 AUC, 0.7

Abbreviations: ABCS, AB Cognitive Screen; RI-48: Rappel Indicé.


H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 277

speaking subjects (97 MCI, 121 AD, 145 normal controls to 6 years (average, 3.89 years). Accuracy was determined on
[NC]). The test achieved a sensitivity of 80% and specificity the retrospective capability of the test to detect MCI on the
of 92% to detect MCI and separate it from NC. Positive basis of final diagnosis of AD with follow-up. With specificity
predictive value (PPV) and negative predictive value (NPV) set at 80%, sensitivity was 83%. PPV was 46%, and NPV was
were not reported. MCI subjects were recruited in a memory 96%. Administration time was not reported [115].
clinic. The DemTect can be administered in 8 to 10 minutes. The following screening instruments were studied by
Rappel-indice-48-Adams (RI-48) [111] evaluates de- using a prospective predictive accuracy design.
layed cued-recall of 48 items. It was tested in 107 French- The Examen Cognitif par Ordinateur (ECO) evaluates
speaking subjects (25 MCI, 22 AD, 22 subjective memory multiple cognitive domains, including working memory,
complaints), 38 NC). The test achieved a sensitivity of 77% verbal and visuospatial memory, implicit memory, language
and specificity of 100% to detect MCI and separate it from skills, verbal fluency, visuospatial skills, and attention. It
NC. MCI subjects were recruited in a memory clinic. The was determined to be a good predictive test for determining
RI-48 can be administered in 20 to 25 minutes. which subjects (308 with cognitive complaints, 60 without
The Montreal Cognitive Assessment (MoCA) [112] cognitive complaints) will convert to AD in a 2-year pro-
evaluates delayed recall, verbal fluency, visuospatial skills, spective study. Three subtests (delayed story recall, verbal
clock drawing, executive functions, calculation, abstraction, fluency, visuospatial construction) when combined achieved a
language, orientation, attention, and concentration. It was sensitivity of 73% and specificity of 99% to predict conversion
validated in French and English with 277 subjects (94 MCI, to AD. The test subtests could be administered in 5 to 10
93 AD, 90 NC). The test achieved a sensitivity of 90% and minutes [116].
specificity of 87% to detect MCI and separate it from NC. The Short Test of Mental Status (STMS) evaluates de-
The PPV was 89%, and the NPV was 91%. MCI subjects layed recall, orientation, attention, learning, calculation,
were recruited in a memory clinic (in an academic center) construction, abstraction, and information. It was shown to
and in the community. The MoCA can be administered in be as insensitive (less then 80% for both sensitivity and
10 minutes. specificity) as the MMSE for prevalent MCI and modest in
Clock Drawing Test (CDT) was tested in 89 Japanese- its ability to predict conversion to AD, with follow-up
speaking subjects (41 normal elderly controls and 48 MCI average of 5.6 years [117].
individuals) with 75% sensitivity and 76% specificity to The following screening instruments have shown prom-
detect MCI and separate it from NC [113]. ising results with well-designed studies based on CDR 0.5
The AB Cognitive Screen (ABCS) evaluates delayed criteria (questionable dementia) [21,22]:
recall, orientation, registration, clock drawing, and word The CERAD 10-word list (CWL) [118] evaluates imme-
fluency. It was assessed in 235 English-speaking subjects diate and delayed verbal memory of a list of 10-words. It
(124 MCI, 111 NC). The MCI subjects’ diagnosis was not has shown promising results with well-designed studies
confirmed with neuropsychological testing, and demonstra- based on CDR 0.5 criteria. It was tested in English, with 471
tion of objective memory deficits was not considered as part subjects classified according to CDR score of 0, 0.5, or 1
of the MCI criteria. The area under the curve (AUC) was (NC 119, MCI 95, mild dementia 257). The test achieved a
0.7 for distinguishing normal subjects from MCI. No sensitivity of 82% and specificity of 91% to detect MCI and
sensitivity or specificity was reported. MCI subjects were separate it from NC. With correspondence analysis, a tech-
recruited in a memory clinic. The test can be adminis- nique that creates weighted scores from the individual per-
tered in 3 minutes [114]. formance profile, the sensitivity and specificity reached
The Modified Mini-Mental State Examination with Ex- 94% and 89%, respectively. PPV was 100%, and NPV was
tended Delayed Recall evaluates delayed recall of the three 97%. MCI subjects were recruited in a memory clinic and in
MMSE words at three 5-minute intervals. It was assessed in the community. Administration time is not reported.
102 subjects (52 NC, 24 MCI, 26 dementia). Sensitivity and The Mini-Cog [119] evaluates word learning, delayed
specificity for MCI were 70.8% and 84.6%, respectively. recall, and clock drawing. It was tested in English with 371
Subjects were recruited in a memory clinic [47]. multiethnic subjects (71 MCI, 112 AD, 48 other dementia,
The following screening instrument was studied by using 140 NC). The test achieved a sensitivity of 55% and spec-
a retrospective predictive accuracy design: ificity of 83% to detect MCI and separate it from NC. MCI
The Combined Mini-mental-cognitive Capacity (CMC) subjects were recruited in a memory clinic. The Mini-Cog
combines the MMSE with the Cognitive Capacity Screen- can be administered in 3 minutes.
ing Examination evaluating multiple cognitive domains and The following screening instruments have shown either
are both scored out of 30 points. The total score of the CMC low sensitivity or specificity (less than 80%) results with
is 47 points, scoring only once overlapping items in both well-designed studies, on the basis of CIND criteria rather
tests. The CMC was tested in English in 351 subjects eval- than MCI:
uated for memory impairment at a memory clinic. Eighty- The 6-Item Screener [120] evaluates delayed recall and
four (23.9%) developed dementia (47 AD) in follow-up up orientation. It was tested in English with 995 subjects (244
278 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

CIND, 43 dementia, 708 NC). The test achieved a sensitiv- earlier point in time and make the current confusion
ity of 97% and specificity of 49% to detect MCI and sepa- regarding MCI a thing of the past.
rate it from NC in the community. PPV was 41%, and NPV (4) Different clinical assessment algorithms for MCI
was 98.3. MCI subjects were recruited in a memory clinic screening by first-line physicians need to be tested.
and in the community. The 6-Item Screener can be admin- There is a need to assess and adapt simple memory
istered in 1.5 minutes in person or over the phone. clinic tools to be used by first-line physicians in the
Letter F Fluency and Animal Fluency [121] achieved community setting.
sensitivities of 48% and 44% and specificities of 57% and (5) We should also assess the predictive value of screen-
96%, respectively, to detect MCI and separate it from NC. ing measures in assessing risk of conversion to AD.
In conclusion, there is no accepted, agreed on, or recom- (Table 1).
mended way to diagnose or screen for MCI at the present
time according to the literature. No studies exist that com- Author Disclosures
pare MCI screening tests validated according to different
definitions for mild memory loss, namely CDR 0.5, CIND, Howard Chertkow has received support from Pfizer Can-
and MCI as defined by Petersen [30 –32]. It appears that no ada (Advisory Board, Speaker, Grant Recipient), Neuro-
clear consensus exists in the literature for a specific ap- chem Inc. (Advisory Board), and Lundbeck Canada (Advi-
proach for diagnosing MCI. sory Board, Speaker).
One can formulate the slightly different question as to Ziad Nasreddine has received support from Pfizer (Con-
what approach to diagnosis makes sense for general prac- sultant, Adviser, Program Organizer, Lecturer), Novartis
titioners and specialists? The CWL was validated in CDR (Lecturer, Drug Trials, Advisor), Janssen Ortho Inc. (Lec-
0.5, and its administration time is not clear. Only three other turer, drug trials, advisor), Neurochem (Drug Trial), Myriad
cognitive screening tests have shown high (greater than (Drug Trial), and Sanofi-Aventis (Drug Trial).
80%) sensitivity and specificity for diagnosis of MCI and Fadi Massoud has received support from Janssen Ortho
therefore are the most promising tests currently available. Inc. (Advisory Board, CME Speaker, Research Funds),
Furthermore, they are all easy to use in a clinical setting and Lundbeck Canada (Advisory Board, CME Speaker, Re-
can be administered in less than 15 minutes. The three tests search Funds), Novartis (Advisory Board, CME Speaker,
are the MoCA, the DemTec, and the CMC. Research Funds), and Pfizer (Advisory Board, CME
Speaker, Research Funds).
Howard Bergman has received support from Pfizer
9. Recommendations approved at the CCCDTD3
(Drug Trial) and Novartis (Drug Trial).
meeting regarding diagnostic maneuvers

1. In cases in which there is suspicion of cognitive Acknowledgments


impairment or concern about the patient’s cognitive This work was supported by grants from a number of
status and the MMSE score is in the normal range (24 agencies. Howard Chertkow is supported by operating
to 30), tests such as the MoCA, DemTect, or CMC grants from the CIHR (Canadian Institutes for Health Re-
could be administered. These would help to demon- search), the Alzheimer Society of Canada, and the Fonds de
strate objective cognitive loss (Grade B, Level 2). la recherche en santé du Québec (FRSQ).
2. There is good evidence that the addition of in-depth Howard Chertkow and Sylvie Belleville receive “cher-
neuropsychological testing can be recommended to cheur national” awards from the FRSQ (Fonds de la Re-
aid in the confirmation of the diagnosis (Grade A, cherche en Santé du Québec.
Level 1). Howard Bergman is supported through the Dr Joseph
Kaufmann Chair in Geriatric Medicine at McGill Univer-
10. Areas for future research sity.
Yves Joanette was supported by a grant from the Cana-
(1) There is a need for long-term studies of the natural dian Institutes of Health Research (MOP-15006).
history of MCI in the general physician or memory Sylvie Belleville is supported by research grants from the
clinic setting, as opposed to population studies. Canadian Institutes of Health Research (CIHR), Heart and
(2) There is a need for formal comparison of the Stroke Foundation of Canada, Alzheimer’s Society of Can-
different diagnostic approaches and labels to com- ada, CAREC, and NSERC.
pare their natural history and prognosis in a clin-
ical setting. References
(3) There is a strong need for development of better AD
biomarkers. Ultimately, establishment of such biomar- [1] Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Ga-
kers will make it possible to diagnose AD at a much vaghan DJ, et al. Assessing the quality of reports of randomized
H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 279

clinical trials: is blinding necessary? Control Clin Trials 1996; [20] Folstein M, Anthony JC, Parhad I, Duffy B, Gruenberg EM. The
17:1–12. meaning of cognitive impairment in the elderly. J Am Geriatr Soc
[2] Woolf SH, Battista RN, Anerson G, Logan A, Wang, E. Asssessing 1985;33:228 –35.
the clinical effectiveness of preventive manoeuvers: analytical prin- [21] Hughes C, Berg L, Danziger W, Coben L, Martin R. A new clinical
ciples and systematic methods in reviewing evidence and develop- scale for the staging of dementia. Br J Psychiatry 1982;140:566 –72.
ing clinical practice recommendations. J Clin Epidemiol 1990;43: [22] Morris JC, McKeel DW Jr, Storandt M, Rubin EH, Price JL, Grant
891–905. EA, et al. Very mild Alzheimer’s disease: informant-based clinical,
[3] CSHA. The incidence of dementia in Canada: the Canadian Study of psychometric, and pathologic distinction from normal aging. Neu-
Health and Aging Working Group. Neurology 2000;55:66 –73. rology 1991;41:469 –78.
[4] Drachman D. If we live long enough, will we all be demented? [23] Reisberg B, Ferris SH, de Leon MJ, Crook T. Global Deterioration
Neurology 1994;44:1563–5. Scale (GDS). Psychopharmacol Bull 1988;24:661–3.
[5] McKann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan [24] Kluger A, Ferris SH, Golomb J, Mittelman MS, Reisberg B. Neu-
EM. Clinical diagnosis of Alzheimer’s disease: report of the ropsychological prediction of decline to dementia in nondemented
NINCDS-ADRDA work group under the auspices of the Depart- elderly. J Geriatr Psychiatry Neurol 1999;12:168 –79.
ment of Health Services Task Force on Alzheimer’s disease. Neu- [25] Jonker C, Geerlings MI, Schmand B. Are memory complaints pre-
rology 1984;34:939 – 44. dictive for dementia? a review of clinical and population-based
[6] Crook T, Bartus RT, Ferris SH, Whitehouse P, Cohen GD, Gershon studies. Int J Geriatr Psychiatry 2000;15:983–91.
SG. Age-associated memory impairment: proposed diagnostic cri- [26] O’Brien J, Beats B, Hill K, Howard R, Sahakian B, Levy R. Do
teria and measures of clinical change: report of a National Institute subjective memory complaints precede dementia? a three-year
of Mental Helath Work Group. Dev Neuropsychol 1986;2:261–76. follow-up of patients with supposed “benign senescent forgetful-
[7] Blackford RC, LaRue A. Criteria for diagnosing age-associated ness”. Int J Geriatr Psychiatry 1992;7:481– 6.
memory impairment: proposed revisions from the field. Dev Neu- [27] Flicker C, Ferris SH, Reisberg B. A longitudinal study of cognitive
ropsychol 1989;5:295–306. function in elderly persons with subjective memory complaints.
[8] Hanninen T, Hallikainen M, Koivisto K, Helkala EL, Reinikainen J Am Geriatr Soc 1993;41:1029 –32.
KJ, Soininen H, et al. A follow-up study of age-associated memory [28] Levy R. Report of the working party of the International Psychoge-
riatric Association in collaboratino with the World Health Organi-
impairment: neuropsychological predictors of dementia. J Am Geri-
zation: aging-associated cognitive decline [published erratum ap-
atr Soc 1995;43:1007–15.
pears in Int Psychogeriatr 1994;6:133]. Int Psychogeriatr 1994;
[9] Larrabee GJ. Age-associated memory impairment: definition and
6:63– 8.
psychometric characteristics. Aging Neuropsychology & Cognition
[29] Smith AD, Jobst K. Use of structural imaging to study the progres-
1996;3:118 –31.
sion of Alzheimer’s disease. Br Med Bull 1996;52:3.
[10] Salthouse TA. Theoretical perspectives on cognitive aging. Hills-
[30] Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG,
dale, NJ. Lawrence Erlbaum Associates, Inc; 1991. p. 438.
Kokmen E. Mild cognitive impairment: clinical characterization
[11] Collie A, Maruff P, Shafiq-Antonacci R, Smith M, Hallup M,
and outcome. Arch Neurol 1999;56:303– 8.
Schofield PR, et al. Memory decline in healthy older people: impli-
[31] Petersen RC, Stevens JC, Ganguli M, Tangalos EG, Cummings JL,
cations for identifying mild cognitive impairment. Neurology 2001;
DeKosky ST. Practice parameter: early detection of dementia: mild
56:1533– 8.
cognitive impairment (an evidence-based review)—report of the
[12] Sliwinski M, Lipton RB, Buschke H, Stewart W. The effects of
Quality Standards Subcommittee of the American Academy of Neu-
preclinical dementia on estimates of normal cognitive functioning in
rology. Neurology 2001;56:1133– 42.
aging. J Gerontol B Psychol Sci Soc Sci 1996;51:217–25.
[32] Petersen RC, Smith GE, Ivnik RJ, Tangalos EG, Schaid DJ, Thi-
[13] Bennett DA, Schneider JA, Arvanitakis Z, Kelly JF, Aggarwal NF, bodeau SN, et al. Apolipoprotein E status as a predictor of the
Shah RC, et al. Neuropathology of older persons without cognitive development of Alzheimer’s disease in memory-impaired individu-
impairment from two community-based studies. Neurology 2006; als [published erratum appears in JAMA 1995;274:538]. JAMA
66:1837– 44. 1995;273:1274 – 8.
[14] Tuokko H, Frerichs RJ. Cognitive impairment with no dementia [33] Petersen RC. Mild cognitive impairment as a diagnostic entity.
(CIND): longitudinal studies, the findings and the issues. Clin Neu- J Intern Med 2004;256:183–94.
ropsychol 2000;14:504 –25. [34] Ganguli M, Dodge HH, Shen C, DeKosky ST. Mild cognitive
[15] Graham JE, Rockwood K, Beattie BL, Eastwood R, Gauthier S, impairment, amnestic type: an epidemiologic study. Neurology
Tuokko H, et al. Prevalence and severity of cognitive impairment 2004;63:115–21.
with and without dementia in an elderly population. Lancet 1997; [35] Lopez O, Jagust W, DeKosky S, Becker J. Risk factors for mild
349:1793– 6. cognitive impairment in a population study. Neurology 2002;58:
[16] Ebly EM, Hogan DB, Parhad IM. Cognitive impairment in the A238.
nondemented elderly: results from the Canadian Study of Health and [36] Chertkow H. Mild memory loss in the elderly: can we predict
Aging. Arch Neurol 1995;52:612–9. development in dementia? Can J Neurol Sci 1998;25:1–S4.
[17] Tuokko H, Frerichs R, Graham J, Rockwood K, Kristjansson B, [37] Royall DR, Chiodo LK, Polk MJ. Misclassification is likely in the
Fisk J, et al. Five-year follow-up of cognitive impairment with no assessment of mild cognitive impairment. Neuroepidemiology 2004;
dementia. Arch Neurol 2003;60:577– 82. 23:185–91.
[18] Feldman H, Levy AR, Hsiung GY, Peters KR, Donald A, Black SE, [38] Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund
et al. A Canadian cohort study of cognitive impairment and related LO, et al. Mild cognitive impairment: beyond controversies, towards
dementias (ACCORD): study methods and baseline results. Neuro- a consensus—report of the International Working Group on Mild
epidemiology 2003;22:265–74. Cognitive Impairment. J Intern Med 2004;256:240 – 6.
[19] Palmer K, Wang H-X, Backman L, Winblad B, Fratiglioni L. Dif- [39] Chertkow H. What are the characteristics of patients with mild
ferential evolution in non-demented older persons: results from the cognitive impairment who do not progress to dementia? The Amer-
Kungsholmen Project. Am J.Psychiatry 2002;159:436 – 42. ican Neurological Association, San Francisco, CA, Oct 21, 2003.
280 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

[40] Chertkow HM, Duong A, Philips N, Bergman H, Babins L, Kelner N. [59] Bennett DA, Schneider JA, Bienias JL, Evans DA, Wilson RS. Mild
Mild cognitive impairment. the danger of basing the diagnosis on cognitive impairment is related to Alzheimer disease pathology and
published normative groups (P3-072). Paper presented at ICAD- cerebral infarctions. Neurology 2005;64:83441.
Alzheimer’s Association 10th International Conference on Alzheimer’s [60] Davis KL, Mohs RC, Marin D, Purohit DP, Perl DP, Lantz M, et al.
Disease and Related Disorders, 2006, Madrid, Spain. Cholinergic markers in elderly patients with early signs of Alzhei-
[41] Portet F, Ousset PJ, Visser PJ, Frisoni GB, Nobili F, Scheltens P, et mer disease. JAMA 1999;281:1401– 6.
al. Mild cognitive impairment (MCI) in medical practice: a critical [61] DeKosky ST, Ikonomovic M, Cochran EJ, Kordower JH, Mufson
review of the concept and new diagnostic procedure—report of the EJ. Cholinergic upregulation in hippocampus in mild cognitive
MCI Working Group of the European Consortium on Alzheimer’s impairment. relation to Alzheimer Neuropathology. Paper presented
Disease. J Neurol Neurosurg Psychiatry 2006;77:714 – 8. at 54th Annual Meeting of the American Academy of Neurology,
[42] Grundman M, Petersen RC, Morris JC. ADSC Cooperative Study: Denver, Colorado, April 13-20, 2002.
rate of dementia of the Alzheimer type (DAT) in subjects with mild [62] Daly E, Zaitchik D, Copeland M, Schmahmann J, Gunther J, Albert
cognitive impairment. Neurology 1996;46:A403. M. Predicting conversion to Alzheimer disease using standardized
[43] Visser PJ, Kester A, Jolles J, Verhey F. Ten-year risk of dementia clinical information. Arch Neurol 2000;57:675– 80.
in subjects with mild cognitive impairment. Neurology 2006;67: [63] Fisk JD, Merry HR, Rockwood K. Variations in case definition
1201–7. affect prevalence but not outcomes of mild cognitive impairment.
[44] Chertkow H, Verret L, Bergman H, Wolfson C, McKelvey R. Neurology 2003;61:1179 – 84.
Predicting progression to dementia in elderly subjects with mild [64] Hsiung G-Y, Johnston D, Donald A, Feldman H. An analysis of
cognitive impairment. a multidisciplinary approach. Paper presented vascular risk factors and ApoE genotype in subtypes of dementia in
at 2001 Contemporary Clinical Issues Plenary Session, 53rd Annual the ACCORD study. Neurology 2002;58(Suppl 3):A107.
Meeting American Academy of Neurology, 2001, Philadelphia, PA. [65] Jonker C, Schmand B, Lindeboom J, Havekes LM, Launer LJ.
[45] Ritchie K, Leibovici D, Ledesert B, Touchon J. A typology of Association between apolipoprotein E epsilon4 and the rate of cog-
sub-clinical senescent cognitive disorder. B J Psychiatry 1996;168: nitive decline in community-dwelling elderly individuals with and
470 – 6. without dementia. Arch Neurol 1998;55:1065–9.
[46] Morris JC, Storandt M, Miller JP, McKeel DW, Price JL, Rubin EH, [66] Dik MG, Jonker C, Bouter LM, Geerlings MI, van Kamp GJ, Deeg
DJ. APOE-epsilon4 is associated with memory decline in cogni-
et al. Mild cognitive impairment represents early-stage Alzheimer
tively impaired elderly. Neurology 2000;54:1492–7.
disease. Arch Neurol 2001;58:397– 405.
[67] Small BJ, Basun H, Backman L. Three-year changes in cognitive
[47] Luis CA, Loewenstein DA, Acevedo A, Barker WW, Duara R. Mild
performance as a function of apolipoprotein E genotype: evidence
cognitive impairment: directions for future research. Neurology
from very old adults without dementia. Psychology & Aging 1998;
2003;61:438 – 44.
13:80 –7.
[48] Loewenstein DA, Barker WW, Harwood DG, Luis C, Acevedo A,
[68] Lehmann M, Gottfries CG, Regland B. Identification of cognitive
Rodriguez I, et al. Utility of a modified Mini-Mental State Exami-
impairment in the elderly: homocysteine is an early marker. Dement
nation with extended delayed recall in screening for mild cognitive
Geriatr Cogn Disord 1999;10:12–20.
impairment and dementia among community dwelling elders. Int J
[69] Corrada M, Kawas C. Plasma total cholesterol, LDL, HDL levels
Geriatr Psychiatry 2000;15:434 – 40.
and the risk of Alzheimer disease: the Baltimore Longitudinal Study
[49] Ritchie K, Artero S, Touchon J. Classification criteria for mild cog-
of Aging. Neurology 2002;58:A106.
nitive impairment: a population-based validation study. Neurology
[70] Yaffe K, Simonsick E, Launer L, Lindquist K, Penninx BW, Pahor
2001;56:37– 42.
M, et al. Inflammatory markers and cognitive decline in well-
[50] Ritchie K, Touchon J. Mild cognitive impairment: conceptual basis functioning elders: the Health ABC Study. Neurology 2002;58:
and current nosological status. Lancet 2000;355:225– 8. A106.
[51] Chertkow HM, Bergman H, Wolfson C, Babins L, Kelner N. Stan- [71] Jack CR, Petersen RC, Xu Y, O’Brien PC, Smith GE, Ivnik RJ, et
dard neuropsychological tests do not predict development of Alz- al. Rate of medial temporal lobe atrophy in typical aging and
heimer’s Disease in indivduals with “Age-associated Cognitive De- Alzheimers disease. Neurology 1998;51:993–9.
cline”. Can J Neurol Sci 1998;25(Suppl 1):S27. [72] Fox NC. Increased rates of atrophy in early and preclinical AD:
[52] Craik F. Memory changes in normal aging. Curr Dir Psychol Sci studies with registration of serial MRI. Neurobiol Aging 2000;21:
1994;3:1558. S74.
[53] Artero S, Touchon J, Ritchie K. Disability and mild cognitive [73] Jack CR Jr, Petersen RC, Xu Y, O’Brien PC, Smith GE, Ivnik RJ,
impairment: a longitudinal population-based study. Int J Geriatr et al. Rates of hippocampal atrophy correlate with change in clinical
Psychiatry 2001;16:1092–7. status in aging and AD. Neurology 2000;55:484–9.
[54] Visser P, Verhey F. Distinction between preclinical Alzheimer’s [74] de Leon MJ, Convit A. Longitudinal measures of hippocampal
disease and depression. J Am Geriatr Soc 2000;48:479 – 84. formation pathology and CSF measures of phospho-tau and amyloid
[55] Lautenschlager NT, Riemenschneider M, Drzezga A, Kurz AF. beta: early markers for Alzheimer disease. Paper presented at Sev-
Primary degenerative mild cognitive impairment: study population, enth International Geneva/Springfield symposium on advances in
clinical, brain imaging and biochemical findings. Dement Geriatr Alzheimer Therapy, 2002; Geneva, Switzerland.
Cogn Disord 2001;12:379 – 86. [75] de Leon MJ, Convit A, Wolf OT, Tarshish CY, DeSanti S, Rusinek
[56] Bouras C. Evolution of AD pathology in the aging brain. Paper H, et al. Prediction of cognitive decline in normal elderly subjects
presented at American Academy of Neurology, 2002, Geneva, with 2-[(18)F]fluoro-2-deoxy-D-glucose/poitron-emission tomogra-
Switzerland. phy (FDG/PET). Proc Natl Acad Sci U S A 2001;98:10966 –71.
[57] Markesbery W, Schmitt FA, Kryscio RH, Davis D, Smith CD, [76] Reiman EM, Caselli RJ, Yun LS, Chen K, Bandy D, Minoshima S,
Wekstein D. Neuropatholic substrate of mild cognitive impairment. et al. Preclinical evidence of Alzheimer’s disease in persons ho-
Arch Neurol 2006;63:38 – 46. mozygous for the epsilon 4 allele for apolipoprotein E. N Engl
[58] Knopman DS, Parisi JE, Salviati A, Floriach-Robert M, Boeve J Med 1996;334:752– 8.
BF, Ivnik RJ, et al. Neuropathology of cognitively normal el- [77] McKelvey R, Bergman H, Stern J, Rush C, Zahirney G, Chertkow
derly. J Neuropathol Exp Neurol 2003;62:1087–95. H. Lack of prognostic significance of SPECT abnormalities in non-
H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282 281

demented elderly subjects with memory loss. Can J Neurol Sci [99] Larrieu S, Letenneur L, Orgogozo JM, Fabrigoule C, Amieva H, Le
1999;26:23– 8. Carret N, et al. Incidence and outcome of mild cognitive impairment
[78] Johnson KA, Jones K, Holman BL, Becker JA, Spiers PA, Satlin A, in a population-based prospective cohort. Neurology 2002;59:
et al. Preclinical prediction of Alzheimer’s disease using SPECT. 1594 –9.
Neurology 1998;50:1563–71. [100] Wolf H, Grunwald M, Ecke GM, Zedlick D, Bettin S, Dannenberg
[79] Kantarci K, Jack CR Jr, Xu YC, Campeau NJ, O’Brien PC, Smith C, et al. The prognosis of mild cognitive impairment in the elderly.
GE, et al. Regional metabolic patterns in mild cognitive impairment J Neural Transm Suppl 1998;54:31–50.
and Alzheimer’s disease: a 1H MRS study. Neurology 2000;55: [101] Chertkow H. Five year follow-up of mild cognitive impairment.
210 –7. predictors of progression—the 8th International Conference on Alz-
[80] Nordberg A. PET activation in MCI and AD patients. Paper pre- heimer’s Disease and Related Disorders. Stockholm, Sweden, July
sented at Seventh Annual Springfield/Geneva symposium on ad- 2002.
vances in Alzheimer Therapy, 2002, Geneva, Switzerland. [102] Davis HS, Rockwood K. Conceptualization of mild cognitive im-
[81] Bookheimer SY, Strojwas M, Cohen MS, Saunders AM, Pericak- pairment: a review. Int J Geriatr Psychiatry 2004;19:313–9.
Vance MA, Mazziotta JC, et al. Patterns of brain activation in people [103] Petersen RC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive
at risk for Alzheimer’s disease. N Engl J Med 2000;343:450 – 6. impairment: clinical characterization and outcome— correction.
[82] Backman L. The cognitive transition from normal aging to AD. Arch Neurol 1999;56:6.
Paper presented at the Seventh Springfield/Geneva symposium on [104] Collie A, Maruff P. An analysis of systems of classifying mild
advances in Alzheimer Therapy, 2002, Geneva, Switzerland. cognitive impairment in older people. Aust N Z J Psychiatry 2002;
[83] Bozoki A, Giordani B, Heidebrink J, Berent S, Foster N. Mild 36:133– 40.
cognitive impairments predict dementia in non-demented elderly [105] Feldman HH, Jacova C. Mild cognitive impairment. Am J Geriatr
patients with memory loss. Arch Neurol 2001;58:411– 6. Psychiatry 2005;13:645–55.
[84] Tierney MC, Szalai JP, Snow WG, Fisher RH. The prediction of [106] DeCarli C. Mild cognitive impairment: prevalence, prognosis, aeti-
Alzheimer disease: the role of patient and informant perceptions of ology, and treatment. Lancet Neurol 2003;2:15–21.
cognitive deficits. Arch Neurol 1996;53:423–7. [107] Salloway S, Ferris S, Kluger A, Goldman R, Griesing T, Kumar D,
[85] Devanand DP, Folz M, Gorlyn M, Moeller JR, Stern Y. Question- et al. Efficacy of donepezil in mild cognitive impairment: a random-
ized placebo-controlled trial. Neurology 2004;63:651–7.
able dementia: clinical course and predictors of outcome. J Am
[108] Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins RV,
Geriatr Soc 1997;45:321– 8.
et al. Current concepts in mild cognitive impairment. Arch Neurol
[86] Devanand DP, Michaels-Marston KS, Liu X, Pelton GH, Padilla M,
2001;58:1985–92.
Marder K, et al. Olfactory deficits in patients with mild cognitive
[109] Jelic V, Winblad B. Treatment of mild cognitive impairment: ratio-
impairment predict Alzheimer’s disease at follow-up. Am J Psychi-
nale, present and future strategies. Acta Neurol Scand Suppl 2003;
atry 2000;157:1399 – 405.
179:83–93.
[87] Albert MS, Moss MB, Tanzi R, Jones K. Preclinical prediction of
[110] Kalbe E, Kessler J, Calabrese P, Smith R, Passmore AP, Brand M,
AD using neuropsychological tests. J Int Neuropsychol Soc 2001;
et al. DemTect: a new, sensitive cognitive screening test to support
7:631–9.
the diagnosis of mild cognitive impairment and early dementia. Int
[88] Graves AB, Bowen JD, Rajaram L, McCormick WC, McCurry SM,
J Geriatr Psychiatry 2004;19:136 – 43.
Schellenberg GD, et al. Impaired olfaction as a marker for cognitive
[111] Ivanoiu A, Adam S, Van der Linden M, Salmon E, Juillerat AC,
decline: interaction with apolipoprotein E epsilon4 status. Neurol-
Mulligan R, et al. Memory evaluation with a new cued recall test in
ogy 1999;53:1480 –7.
patients with mild cognitive impairment and Alzheimer’s disease.
[89] Schipper HM, Chertkow H, Mehindate K, Frankel D, Melmed C, J Neurol 2005;252:47–55.
Bergman H. Evaluation of heme oxygenase-1 as a systemic biolog- [112] Nasreddine ZS, Phillips NA, Bedirian V, Charbonneau S, White-
ical marker of sporadic AD. Neurology 2000;54:1297–304. head V, Collin V, et al. The Montreal Cognitive Assessment,
[90] Andreasen N, Minthon L, Vanmechelen E, Vanderstichele H, Dav- MoCA: a brief screening tool for mild cognitive impairment. J Am
idsson P, Winblad P, et al. Cerebrospinal fluid tau and Abeta42 as Geriatr Soc 2005;53:695–9.
predictors of development of Alzheimer’s disease in patients with [113] Yamamoto S, Mogi N, Umegaki H, Suzuki H, Ando F, Shimokata
mild cognitive impairment. Neurosci Lett 1999;273:5– 8. H, et al. The clock drawing test as a valid screening method for mild
[91] Saxton J, Lopex O, Ratcliff G, Dulberg C, Fried L, Carlson MC, cognitive impairment. Dement Geriatr Cogn Disord 2004;18:172–9.
Kuller L. Preclinical Alzheimer disease: neuropsychological test [114] Molloy DW. Screening for mild cognitive impairment: comparing
performance 1.5 to 8 years prior to onset. Neurology 2004;63: the SMMSE and the ABCS. Can J Psychiatry 2005;50:52– 8.
2341–7. [115] Xu G, Meyer JS, Huang Y, Chen G, Chowdhury M, Quach M.
[92] Frisoni GB, Padovani A, Wahlund LO. The predementia diagnosis Cross-cultural comparison of mild cognitive impairment between
of Alzheimer disease. Alzheimer Dis Assoc Disord 2004;18:51–3. China and USA. Curr Alzheimer Res 2004;1:55– 61.
[93] Bennett DA. Mild cognitive impairment. Clin Geriatr Med 2004;20: [116] Artero S, Ritchie K. The detection of mild cognitive impairment in
15–25. the general practice setting. Aging Ment Health 2003;7:251– 8.
[94] Bennett DA, Wilson RS, Schneider JA, Evans DA, Beckett LA, [117] Tang-Wai DF, Knopman DS, Geda YE, Edland SD, Smith GE,
Aggarwal NT, et al. Natural history of mild cognitive impairment in Ivnik RJ, et al. Comparison of the short test of mental status and the
older persons. Neurology 2002;59:198 –205. mini-mental state examination in mild cognitive impairment. Arch
[95] Dubois B, Albert ML. Amnestic MCI or prodromal Alzheimer’s Neurol 2003;60:1777– 81.
disease? Lancet Neurol 2004;3:246 – 8. [118] Shankle WR, Romney AK, Hara J, Fortier D, Dick MB, Chen JM,
[96] Morris J. Mild cognitive impairment is early-stage Alzheimer dis- et al. Methods to improve the detection of mild cognitive impair-
ease. Arch Neurol 2006;63:15– 6. ment. Proc Natl Acad Sci U S A 2005;102:4919 –24.
[97] Bennett DA. Update on mild cognitive impairment. Curr Neurol [119] Borson S, Scanlan JM, Watanabe J, Tu SP, Lessig M. Simplifying
Neurosci Rep 2003;3:379 – 84. detection of cognitive impairment: comparison of the Mini-Cog and
[98] Ritchie K. Mild cognitive impairment: an epidemiological perspec- Mini-Mental State Examination in a multiethnic sample. J Am
tive. Dialogues in Clinical Neuroscience 2004;6:401– 8. Geriatr Soc 2005;53:871– 4.
282 H. Chertkow et al. / Alzheimer’s & Dementia 3 (2007) 266 –282

[120] Callahan CM, Unverzagt FW, Hui SL, Perkins AJ, Hendrie HC. [130] Grundman M, Petersen RC, Ferris SH, Thomas RG, Aisen PS,
Six-item screener to identify cognitive impairment among Bennett DA, et al. Mild cognitive impairment can be distinguished
potential subjects for clinical research. Med Care 2002;40:771– from Alzheimer disease and normal aging for clinical trials. Arch
81. Neurol 2004;61:59 – 66.
[121] Canning SJ, Leach L, Stuss D, Ngo L, Black SE. Diagnostic utility [131] Solfrizzi V, Panza F, Colacicco AM, D’Introno A, Capurso C,
of abbreviated fluency measures in Alzheimer disease and vascular Torres F, et al. Vascular risk factors, incidence of MCI, and rates of
dementia. Neurology 2004;62:556 – 62. progression to dementia. Neurology 2004;63:1882–91.
[122] Darby D, Maruff P, Collie A, McStephen M. Mild cognitive im- [132] de Jager CA, Budge MM. Stability and predictability of the classi-
pairment can be detected by multiple assessments in a single day. fication of mild cognitive impairment as assessed by episodic mem-
Neurology. Oct 8 2002;59:1042– 6. ory test performance over time. Neurocase 2005;11:72–9.
[123] Hanninen T, Hallikainen M, Tuomainen S, Vanhanen M, Soininen [133] Devanand DP, Pelton GH, Zamora D, Liu X, Tabert MH, Goodkind
H. Prevalence of mild cognitive impairment: a population-based M, et al. Predictive utility of apolipoprotein E genotype for Alzhei-
study in elderly subjects. Acta Neurol Scand 2002;106:148 –54. mer disease in outpatients with mild cognitive impairment. Arch
[124] Kabani NJ, Sled JG, Chertkow H. Magnetization transfer ratio in Neurol 2005;62:975– 80.
mild cognitive impairment and dementia of Alzheimer’s type. Neu- [134] Geslani DM, Tierney MC, Herrmann N, Szalai JP. Mild cognitive
roimage 2002;15:604 –10. impairment: an operational definition and its conversion rate to
[125] Busse A, Bischkopf J, Riedel-Heller SG, Angermeyer MC. Subclas- Alzheimer’s disease. Dement Geriatr Cogn Disord 2005;19(5-6):
sifications for mild cognitive impairment: prevalence and predictive 383–389.
validity. Psychol Med 2003;33:1029 –38. [135] Kumar R, Dear KB, Christensen H, Ilschner S, Jorm AF, Meslin
[126] Lambon Ralph MA, Patterson K, Graham N, Dawson K, Hodges JR. C, et al. Prevalence of mild cognitive impairment in 60- to
Homogeneity and heterogeneity in mild cognitive impairment and 64-year-old community-dwelling individuals: The Personality
Alzheimer’s disease: a cross-sectional and longitudinal study of 55 and Total Health through Life 60⫹ Study. Dement Geriatr Cogn
cases. Brain 2003;126:2350 – 62. Disord 2005;19:67–74.
[127] Lopez OL, Jagust WJ, DeKosky ST, Becker JT, Fitzpatrick A, [136] Purser JL, Fillenbaum GG, Pieper CF, Wallace RB. Mild cognitive
Dulberg C, et al. Prevalence and classification of mild cognitive impairment and 10-year trajectories of disability in the Iowa Estab-
impairment in the Cardiovascular Health Study Cognition Study: lished Populations for Epidemiologic Studies of the Elderly cohort.
part 1. Arch Neurol 2003;60:1385–9. J Am Geriatr So 2005;53:1966 –72.
[128] Farlow MR, He Y, Tekin S, Xu J, Lane R, Charles HC. Impact of [137] Visser PJ, Scheltens P, Verhey FR. Do MCI criteria in drug trials
APOE in mild cognitive impairment. Neurology. Nov 23 2004; accurately identify subjects with predementia Alzheimer’s disease?
63(10):1898 –1901. J Neurol Neurosurg Psychiatry 2005;76:1348 –54.
[129] Feldman H, Scheltens P, Scarpini E, Hermann N, Mesenbrink P, [138] Verghese J, Levalley A, Derby C, Kuslansky G, Katz M, Hall C,
Mancione L, et al. Behavioral symptoms in mild cognitive impair- et al. Leisure activities and the risk of amnestic mild cognitive
ment. Neurology 2004;62:1199 –201. impairment in the elderly. Neurology 2006;66:821.
Alzheimer’s & Dementia 3 (2007) 283–291

Mild cognitive impairment and cognitive impairment, no dementia:


Part B, therapy
Fadi Massouda,**, Sylvie Bellevilleb,c, John Kirkd, Howard Chertkowb,d,e,f,*,
Ziad Nasreddineg, Yves Joanetteb,h, Howard Bergmand,e, Morris Freedmani
a
Service de Gériatrie, Centre Hospitalier de l’Université de Montréal, and Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
b
Centre de recherche, Institut Universitaire de Gériatrie de Montréal, Montreal, Quebec, Canada
c
Department de Psychologie, Université de Montréal, Montreal, Quebec, Canada
d
Division of Geriatric Medicine, Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada
e
Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Sir Mortimer B. Davis-Jewish General Hospital, McGill
University, Montreal, Quebec, Canada
f
Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
g
Département de médecine, service de neurologie, Hôpital Charles LeMoyne and Université de Sherbrooke, Montreal, Quebec, Canada
h
Faculté de Médicine, Université de Montréal, Montreal, Quebec, Canada
i
Department of Medicine (Neurology), University of Toronto, and Behavioural Neurology Program, Baycrest Centre for Geriatric Care, Toronto, Ontario, Canada

Abstract Mild cognitive impairment (MCI) and cognitive impairment, no dementia (CIND) might be the
optimum stage at which to intervene with preventative therapies. This article reviews recent work
on the possible treatment and presents evidence-based recommendations approved at the meeting of
the Third Consensus Conference on the Diagnosis and Treatment of Dementia held in Montreal in
March 2006. A number of promising nonpharmacologic interventions have been examined. Asso-
ciations exist with both cognitive and physical activity that suggest that both of these, together or
separately, can delay progression to dementia. Similarly, case control studies as well as prospective
long-term studies suggest a number of low toxicity interventions and supplements that might
significantly impact on MCI progression; folate, B6, and B12 to lower homocysteine levels,
omega-fatty acids, and anti-oxidants (fruit juices or red wine) are good examples. In selected
genotypes such as individuals with APOE e4, therapy with donepezil might slow progression. The
concern, however, is that none of these therapies (including cholinesterase inhibitors) have dem-
onstrated a clinically meaningful effect with randomized, placebo-controlled studies. Just as ran-
domized controlled studies have failed to support primary prevention of dementia by using estrogen
or nonsteroidal anti-inflammatory drugs (NSAIDs), there exists the possibility that well-designed
randomized controlled trials might fail to definitively demonstrate putative or promising mild
cognitive impairment interventions. Pharmacologic interventions and nonpharmacologic therapies,
while tantalizing, are currently for the most part insufficiently proven to allow serious consideration
by physicians. Recommendation were supported for a general “healthy lifestyle” including physical
exercise, healthy nutrition, smoking cessation, and mental stimulation. Close monitoring and
treatment of vascular risk factors are justified and were also supported.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: MCI; CIND; Cognitive impairment; Psychosocial intervention; Cognitive intervention; Exercise

*Corresponding author. Tel.: (514) 340-8260; Fax: (514) 340-8925.


E-mail address: howard.chertkow@mcgill.ca
**Additional Corresponding author. Tel.: (514) 890-8000 26769; Fax: (514) 412-7506.
E-mail address: fadimassoud@videotron.ca

1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.002
284 F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291

1. Treatment for mild cognitive impairment 2-year follow-up, and the effect sizes were moderate to
large. Thus there is good evidence that cognitive training
The preceding article reviewed the concepts and diagno-
increases cognitive efficacy on target measures in healthy
sis of mild cognitive impairment (MCI) and cognitive im-
older adults.
pairment, no dementia (CIND), reviewed our approach to an
Two nonrandomized studies and two randomized con-
evidence-based review of the literature, and presented the
trolled trials (RCTs) have been reported on the effect of
recommendations that received consensus at the Third Con-
cognitive training in MCI. With an RCT design, Olazaran
sensus Conference on the Diagnosis and Treatment of De-
et al [8] reported decreased depression and improved cog-
mentia (CCCDTD3) held in Montreal in March 2006.This
nition (cognitive subscale of the Alzheimer’s Disease As-
second part will use the same approach to review therapy
sessment Scale, cognitive portion) in a mixed group com-
for MCI and CIND. The treatment of MCI has been the
prising 72 AD and 12 MCI patients after a 1-year program
subject of a number of recent chapters and reviews [1,2].
of cognitive-motor stimulation plus psychosocial compared
There have been relatively few randomized controlled trials
with psychosocial support. In a small-scale (n ⫽ 18) RCT
of any therapy sufficient to rank as Level 1 evidence. Nev-
trial, Rapp et al [9] compared cognitive intervention with no
ertheless, there are a number of potential interventions both
treatment. They reported improved subjective memory and
pharmacologic and nonpharmacologic that deserve to be
long-term maintenance during a 6-month period but no
addressed.
effect on objective tests of memory. Gunther et al [10]
reported long-term improvement in cognitive performance
2. Nonpharmacologic treatment for MCI (working memory and verbal episodic memory) in a pre-
2.1. Cognitive intervention in MCI post comparison study of computer-assisted cognitive train-
ing in persons with MCI. Finally, Belleville et al [11]
There have been several relevant studies in this area. We compared the effect of a multifactorial memory training
will first review those carried out in normal elderly and then program with a no-training condition (28 MCI participants)
in MCI. A full listing of these studies is seen in Table 1. and reported larger memory improvement on post-test in the
Longitudinal cohort studies of healthy elderly persons show trained MCI participants compared with the untrained ones.
that engagement in stimulating cognitive activities (engaged Moderate to large effect sizes were obtained for the training
lifestyle; novel and intellectually challenging activities) is effect on target episodic memory measures. Thus, studies
associated with better memory and verbal abilities [3]. In a investigating the effect of cognitive intervention in MCI
case-control study, participation in intellectually stimulating provide encouraging findings. However, the effort required
and social activities in midlife has been associated with to implement such therapeutic measures is not trivial, and
reduced risk of developing Alzheimer’s disease (AD) [4]. In large scale cognitive intervention for MCI would require
a longitudinal cohort study of healthy elderly persons (av- considerable resources. Before widespread recommendation
erage follow-up, 4.5 years), a participant’s frequency of of this therapy can occur, more replication studies are re-
participation in common cognitive activities at baseline was quired with properly controlled RCT designs, larger sample
associated with reduced risk of clinical diagnosis of AD and sizes, and analyses that control for type 1 error.
reduced cognitive decline (annualized change on global In conclusion, longitudinal cohort studies of healthy
cognition, working memory, and perceptual speed) during older adults indicate that engagement in intellectually stim-
the follow-up period [5]. However, the methodology in ulating activities is associated with decreased risk of AD
these studies is based on association; therefore, the direction and decreased cognitive decline. However, the evidence at
of causality remains to be clarified. For example, it is the present time is insufficient to conclude that organized
unclear whether cognitive activities have a protective effect cognitive intervention is beneficial to preventing progres-
on the development of cognitive deficits in aging, or sion in MCI or warrants prescription. On the other hand,
whether reduced engagement in cognitive activities is an given that there is little or no “downside” to cognitive
early sign of AD. activity, it is not unreasonable for physicians and therapists
Verhaeghen et al [6] conducted a quantitative review of to promote engagement in cognitive activity as part of an
studies measuring the efficacy of memory intervention stud- overall healthy lifestyle formulation for elderly individuals
ies in healthy aging. They reported that memory training with and without memory loss.
improved performance on targeted memory tasks and that
the effect sizes for the training effect were in the moderate 2.2. Physical training in MCI
range. One large scale randomized controlled trial on cog-
nitive interventions (memory, speed, or reasoning vs no Several longitudinal cohort studies carried out in nor-
training) was completed in a sample of 2,832 healthy older mal elderly individuals indicated that physical exercise is
adults [7]. The results indicated improved performance after associated with reduced cognitive decline and reduced
training on the cognitive domains that were targeted by the risk of dementia. These studies looked at outcomes such
interventions. The positive effects were sustained during a as a change score on the Mini-Mental State Examination
Table 1
Studies of non-pharmacological treatment of MCI
Authors Year Type Population Outcome Measures Results Notes

Physical training
Colcombe and 2003 Meta-analysis (18 RCTs) Healthy and clinical aged Cognitive tests ES, .478; largest on executive Not MCI; controlled
Kramer [15] intervention: ??
Heyn et al [16] 2004 Meta-analysis (30 RCTs) Demented Physical, functional, behavioral, ES, .62; large effect on most Heterogeneous group; 9 with
and cognitive measures components mild impairment; controlled
intervention: ??
Kramer et al [13] 2004 Literature review of Healthy aged Cognitive tests Improves executive functions
RCT and prospective
studies
Lytle et al [12] 2004 Prospective study Community sample Decline on MMSE over a Protects against severe
2-year time decline

F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291


Laurin et al [13] 2001 Prospective Community sample Cognitive impairment Physical activity reduces risk
of cognitive impairment,
AD, and dementia 5 y later
Psychosocial
intervention
Ishizaki et al [45] 2002 Prospective study Mild AD (CDR .05) and Improvement on cognition Small N; a specific intervention
controls and affective measures (eg, reminiscence); no placebo
Cognitive
intervention
Kramer et al [3] 2004 Literature review of Healthy aging Cognitive tests Improves targeted cognitive
RCTs and prospective functions (cognition)
studies; cognitive
intervention and effect
of leisure and
environment
Wilson et al [5] 2002 Prospective study; Longitudinal cohort Diagnosis of AD after a mean Reduces the risk of AD and
cognitively stimulating follow-up of 4.5 yrs the decline on global
activities cognition, WM and speed.
Lindstrom et al [4] 2005 Retrospective study of AD and controls Diagnosis of AD TV viewing increases risk of Limitations: association direction
leisure activity (TV AD; intellectual and social unclear; retrospective; based
viewing in midlife) activities reduce it on questionnaire in surrogate
respondents
Verhagen et al [6] 1992 Literature review of Healthy aging Memory Improved performance (ES, Negative effect of lower mental
intervention studies 0.7; 0.2 in controls) status
Ball et al [7] 2002 RCT Healthy aging Cognition Improves targeted cognitive High quality study; follow-up to
functions; long-standing appear soon
effect (2 years); no
generalisation.
Poon et al [46] 2005 RCT of videoconference MCI and early AD Cognition Equivalent effect of VC and Small N; no placebo
of cognitive and Face to face training
psychosocial
intervention
Olazaran et al [8] 2005 RCT of cognitive MCI and AD Cognition, depression, Positive effect on cognitive No distinction between MCI and
stimulation (1yr functional impact and depression AD; no control of type 1 error
program)

285
286 F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291

(MMSE) 2 years later [12] and risk of dementia 5 years later

Not randomized; no treatment as


No-treatment group as control;
[13]. However, there are also studies that failed to find a

control; small N (26 MCI)


protective effect of physical exercise on cognitive decline

No group of untrained
and on incident dementia [14]. Two recent meta-analyses

small N (9 MCI)
have been published regarding the impact of physical exer-
cise programs on the cognitive function of older adults

participants
[15,16]. Colcombe and Kramer [15] included 18 RCT stud-
ies in their meta-analysis, and Heyn et al [16] included 30
Notes

studies. Both meta-analyses reported moderate effect sizes


for the exercise training effect on global cognitive scores,
with a larger effect on tasks measuring executive control
maintenance but no effect
Positive effect on subjective

Positive effect on cognitive

reported by Colcombe and Kramer. There are immense


measure and long-term

on objective measures

implications of such research in terms of potential public


Pre-post training effect

health measures to prevent dementia and cognitive decline.


For fitness: more studies are needed to assess modality, safety, intensity. Problems: blinding, small N; long-term; appropriate control; to target MCI.
and well-being

However, more studies, particularly RCTs, are needed to


assess the optimal exercise training modalities in older
adults, particularly in terms of intensity and duration. Safety
Results

issues will also need to be better addressed. Furthermore,


there were methodologic limitations in past studies regard-
ing appropriate blinding procedures and sample size. Fi-
nally, no studies have been carried out specifically with
MCI persons to assess the effect of physical training on their
Cognition and well-being

cognitive capacities and cognitive decline.


In conclusion, longitudinal cohort studies of healthy
Outcome Measures

For psychosocial intervention: more studies are needed in MCI to assess potential efficacy, feasibility, safety.

older adults indicate that physical training might be protec-


tive against cognitive decline. RCTs have provided compel-
For cognitive intervention: encouraging results but more RCT studies are needed in MCI with larger N.
Cognition

Cognition

ling evidence for a beneficial effect of physical training on


the cognitive performance of healthy older adults. However,
the evidence is insufficient to conclude that physical train-
ing is beneficial to preventing progression in MCI or war-
rants prescription. Furthermore, safety issues will need to be
better addressed. Keeping this in mind, physicians and ther-
MCI and controls

apists might promote physical activity at an intensity level


that is adapted to the person’s overall physical capacities as
Population

part of a healthy lifestyle for older individuals with and


without memory loss.
MCI

MCI

2.3. Psychosocial intervention


Clinical trial of cognitive

cognitive intervention

There are few data in this area, and no suggestions will


be reviewed at this time.
Pre-post study of
RCT of memory
intervention

intervention

2.4. Cognitive intervention in MCI


Type

2.4.1. Recommendation 7
The evidence at the present time is insufficient to con-
clude that organized cognitive intervention is beneficial to
2002

2006

2003
Year

preventing progression in MCI or warrants prescription


(Grade C, Level 1).
Gunther et al [10]

2.4.2. Recommendation 8
Rapp et al [9]

et al [11]

There is fair evidence that physicians and therapists


Belleville
Continued

should promote engagement in cognitive activity as part of


Authors
Table 1

an overall healthy lifestyle formulation for elderly individ-


uals with and without memory loss (Grade B, Level 2).
F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291 287

Table 2 suggest early cholinergic “functional” modifications such as


Drugs under investigation to prevent progression to AD choline transport and acetylcholine release at this stage [18].
CHEIs All three cholinesterase inhibitors (ChEIs) currently avail-
Anti-inflammatories able in Canada have been used in clinical trials of MCI.
Estrogen
Statins
Salloway et al [19] conducted a 24-week, multicenter,
Cholesterol-lowering drugs randomized, double-blind placebo-controlled study in 270
Anti-amyloid drugs (beta-secretase inhibitors, gamma-secretase inhibitors, individuals with MCI, comparing donepezil 10 mg daily
GAG mimetics, amyloid immunotherapy) with placebo. There were no significant beneficial effects on
Various anti-oxidants including vitamin E the two primary outcome measures used, the New York
AMPAkines
University Paragraph Delayed Recall test and the Alzheimer
Disease Cooperative Study Clinician’s Global Impression
of Change for MCI (ADCS-CGIC-MCI). Subjects on done-
2.5. Physical training as therapy in MCI pezil exhibited statistically significant benefit on two a pri-
2.5.1. Recommendation 9 ori secondary outcome measures, the Alzheimer’s Disease
There is fair evidence that physicians and therapists Assessment Scale– cognitive subscale (ADAS-cog) and the
should promote physical activity at an intensity level that is Patient Global Assessment (PGA). More individuals treated
adapted to the person’s overall physical capacities as part of with donepezil also had adverse events.
a healthy lifestyle for older individuals with and without The Alzheimer’s Disease Cooperative Study Group
memory loss (Grade B, Level 2). (ADCS) led by Petersen conducted a 3-year double-blind
study randomizing individuals with amnestic MCI to vita-
2.5.2. Recommendation 10 min E 2000 IU daily, donepezil 10 mg daily, and placebo
Current evidence is insufficient to conclude that a spe- [20]. In this study, the primary outcome was conversion to
cific program of physical training warrants prescription in possible or probable AD. At the end of the study, there were
MCI patients to prevent progression to dementia (Grade C, no differences in the progression to AD between the three
Level 3). groups. However, on preplanned interim analyses, donepezil-
treated individuals had a slightly reduced likelihood of pro-
3. Pharmacologic approaches to MCI gression to AD during the first year of the study. On a priori
pharmacogenetic analyses, the study confirmed that posses-
Several pharmacologic treatment approaches have been sion of the APOE e4 allele is a major predictor of progres-
attempted in MCI (Table 2). The formal review of the sion to AD and indicated that most of the treatment effect
evidence is presented in Table 3. occurred among the APOE e4 carriers. On secondary anal-
ysis confined to APOE e4 carriers, the effect of donepezil
3.1. Cholinesterase inhibitors
was significant at 12, 24, and 36 months. “In fact, a closer
Although evidence suggests the absence of significant look at their data reveals no convincing evidence of a
cholinergic deficits in MCI [17], several lines of evidence difference in treatment effect according to APOE e4 carrier

Table 3
Standard evidence table for pharmacologic therapy in MCI
Agent Design Author, Year N Statistical Significance Quality Magnitude Absolute Numbers Comments
of of Benefit Risk Needed
Study* Reduction to Treat

Donepezil RCT Salloway et al [19], 2004 270 On secondary outcome 4/5 NA NA NA


measures
Donepezil RCT Petersen et al [20], 2005 769 None in primary outcome; 4/5 NA NA NA
some positive
secondary analyses
Rivastigmine RCT Feldman et al [23], 2007 1018 No 5/5 NA NA NA
(InDDEX)
Galantamine RCT Scheltens et al [24], 2004
Rofecoxib RCT Thal et al [26], 2005 1457 No 5/5 NA NA NA
Estrogen RCT Shumaker et al [28], 2003 4532 Increased risk of dementia 5/5 NA NA NA
(hazard ratio, 2.1)in
treatment group
Ginkgo biloba RCT van Dongen et al [30], 2003 214 No 5/5 NA NA NA

* Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Gavaghan DJ, et al. The quality of reports of randomized clinical trials: is blinding
necessary? Control Clin Trials 1996;1:1–12.
288 F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291

status; the effect of donepezil was similar among APOE e4 3.3. Hormone replacement therapy
carriers and noncarriers (hazard ratio for progression of 0.66
Estrogen has been shown to have neuroprotective effects
as compared with 0.80 for the entire cohort, with overlap-
in several studies. It is considered a neurotrophic factor for
ping confidence intervals). These numbers suggest that the
cholinergic neurons; it stimulates cerebral cellular growth,
observed difference in significance might have been due to
contributes to synaptic reorganization, and decreases beta-
analogous differences in statistical power, because AD de-
amyloid production. It is also associated with antioxidant
veloped in about twice as many APOE e4 carriers (who are
properties and beneficial vascular effects [27]. In a meta-
more likely to be on a course toward AD) as noncarriers analysis of 12 observational studies, estrogen is associated
within the 3 years after enrolment” [21]. Also, the authors with a decreased risk of developing AD, with an odds ratio
indicate that “ . . . there are insufficient data to warrant rec- of 0.69 [27]. In the Women’s Health Initiative Memory
ommending APOE e4 genotyping in persons with mild Study, 4,532 women were randomized to estrogen 0.625 mg
cognitive impairment, and our results cannot be used to daily with medroxyprogesterone 2.5 mg daily and placebo
make this recommendation, since the study was not statis- and were followed up between 3 and 4 years [28]. Not only
tically powered to determine the effects of treatment in did this study fail to demonstrate any benefit on the risk of
separate groups of APOE e4 carriers and noncarriers.” developing MCI and dementia, it actually showed a dou-
Results from a 4-year double-blind, placebo-controlled, bling (hazard ratio, 2.05) of the risk of dementia in the
clinical trial of rivastigmine in MCI individuals (InDDEX) hormone-treated group.
showed no difference in the likelihood of progression to AD In the Multiple Outcomes of Raloxifene Evaluation
between the treatment group and those on placebo [22]. At (MORE) study, postmenopausal women were randomized
the time of the CCCDTD3, preliminary data had been pub- to a selective estrogen receptor modulator, raloxifene, at
lished in abstract format only. Although the final published doses of 60 mg and 120 mg daily and placebo [29]. At
form of the study came out after the CCCDTD3 voting [23], 3-year follow-up, women taking 120 mg of raloxifene had a
there was no meaningful change in the recommendations 33% decreased risk of developing MCI. This benefit was not
based on this study. The overall rate of progression from observed in the group taking raloxifene 60 mg daily.
MCI to AD in this randomized clinical trial was much lower
3.3.1. Recommendation 13
than predicted.
There is currently fair evidence to recommend against
Two clinical trials with galantamine in MCI similarly
the use of estrogen replacement therapy in MCI (Grade D,
showed no difference in the probability of conversion to
Level 1).
AD. However, there was a reduced rate of whole-brain
atrophy in patients treated with galantamine [24]. Therapy 3.4. Ginkgo biloba
with galantamine was associated with an increased all-cause
Ginkgo biloba is commonly used for memory impair-
mortality risk that might be explained, at least in part, by the
ment because of its presumably beneficial antioxidant, anti-
very low mortality rate in the placebo groups. These results
platelet, and neuroprotective properties. The only relevant
are published as abstracts only; therefore, there is little
randomized, placebo-controlled clinical trial of ginkgo bi-
information concerning the methodology.
loba was conducted by van Dongen et al [30] in patients
with dementia and Age-Associated Memory Impairment
3.1.1. Recommendation 11 (AAMI). This category includes individuals with memory
There is currently insufficient evidence to recommend impairment when compared with young controls. At the end
for the use of ChEIs in MCI (Grade C, Level 1). of the 36-week-long trial, there were no statistically signif-
icant differences between the ginkgo biloba and placebo
3.2. Anti-inflammatory agents groups. A Cochrane Database systematic review of ginkgo
Several large well-conducted epidemiologic studies have biloba for cognitive impairment concludes that the three
shown a negative association between the use of NSAIDs most recent clinical trials, which are methodologically more
sound, show inconsistent results [31]. They add that “there
and the development of AD [25]. In the only clinical trial of
is a need for a large trial using modern methodology . . .”.
NSAIDs in MCI, Thal et al [26] randomized patients to rofe-
coxib 25 mg and placebo daily for up to 4 years. Individuals on 3.4.1. Recommendation 14
rofecoxib did not exhibit significant benefit in terms of clinical There is currently fair evidence to recommend against
conversion to clinical AD, which was the primary outcome the use of ginkgo biloba in MCI (Grade D, Level 1).
measure, or on secondary outcome measures.
3.5. Vitamin E
3.2.1. Recommendation 12 Epidemiologic data suggest a negative association be-
There is currently fair evidence to recommend against tween ingestion of vitamin E from natural [32] and artificial
the use of NSAIDs in MCI (Grade D, Level 1). sources [33] and developing AD. The potential neurocog-
F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291 289

nitive benefit of vitamin E was further supported by a should be screened for and treated optimally in MCI (Grade
randomized clinical trial showing it delayed reaching sig- B, Level 2).
nificant clinical milestones in patients with AD [34]. The Other pharmacologic agents that have been considered in
only trial of vitamin E in MCI, discussed in the ChEI MCI have shown either marginal or inconsistent benefit.
section, was negative [20]. Concerns have been raised about These include neurotrophic medications (phosphatidylser-
the safety of vitamin E since the publication of a meta- ine, acetyl-l-carnitine, piracetam), memory stimulants (am-
analysis of 19 clinical trials that suggested an increased pakines, N-methyl-D-aspartate modulators, CREB modula-
all-cause mortality in individuals ingesting 400 or more IU tors), antioxidants, vitamin supplements, and omega fatty
daily [35]. acids.

3.5.1. Recommendation 15
There is currently fair evidence to recommend against
the use of vitamin E in MCI (Grade D, Level 1). 4. Pharmacologic treatment recommendations for
MCI (summary)
3.6. Treatment of vascular risk factors
4.1. Recommendation 11
Several epidemiologic studies have shown the associa-
There is currently insufficient evidence to recommend
tion between vascular risk factors and cognitive impairment
for the use of ChEIs in MCI (Grade C, Level 1).
[36 –39]. Clinical trials have evaluated the role of primary
prevention of vascular risk factors and the risk of cognitive
deterioration. In the Systolic Hypertension in EURope Trial 4.2. Recommendation 12
(SYST-EUR), 2,418 subjects were randomized to active There is currently fair evidence to recommend against
treatment with nitrendipine with the possible addition of the use of NSAIDs in MCI (Grade D, Level 1).
enalapril and hydrochlorothiazide or placebo [40]. In the
active treatment group, the incidence of dementia was re- 4.3. Recommendation 13
duced by 50% at 2 years. In the Perindopril Protection
Against Recurrent Stroke Study (PROGRESS) 6,015 pa- There is currently fair evidence to recommend against
tients with previous stroke or transient ischemic attack were the use of estrogen replacement therapy in MCI (Grade D,
randomized to receive perindopril with or without indapam- Level 1).
ide or placebo [41]. At the end of the 3.9-year follow-up, the
risk of dementia and cognitive decline was significantly 4.4. Recommendation 14
reduced in the active treatment group. The risk was reduced
There is currently fair evidence to recommend against
even further in subjects with recurring stroke. The two large
the use of ginkgo biloba in MCI (Grade D, Level 1).
primary prevention trials with statins have reached some-
what different conclusions; statins do reduce cardiovascular
4.5. Recommendation 15
and cerebrovascular events in primary prevention, but they
do not provide any benefit on cognitive function [42,43]. There is currently fair evidence to recommend against
These seemingly counterintuitive results can be explained the use of vitamin E in MCI (Grade D, Level 1).
by some of the methodologic limitations of these trials,
namely the relatively short duration of follow-up and the 4.6. Recommendation 16
limited, and probably incomplete, evaluation of cognitive
function. One clinical trial evaluated the role of hyperten- Because vascular risk factors and comorbidities impact
sion treatment in individuals with mild cognitive deficits on the development and expression of dementia, they
broadly defined as a MMSE score between 20 and 28 [44]. should be screened for and treated optimally in MCI (Grade
In this study, captopril was compared with bendrofluazide B, Level 2).
in 81 subjects with systolodiastolic hypertension for 24 In conclusion, an evidence-based review of the literature
weeks. There were no statistically significant differences be- provides evidence that leisure activities, cognitive stimula-
tween the two drugs, but patients with the best response to tion, and physical activity should be promoted as part of a
treatment in terms of reduction of their diastolic blood pressure healthy lifestyle in elderly individuals and those with MCI.
significantly improved on two cognitive tests. Interestingly, Vascular risk factors should be treated optimally in these
this study suggests that reduction of blood pressure in elderly individuals as well. No other specific therapies can yet be
hypertensive patients with MCI is not hazardous. recommended or supported as having adequate demonstra-
tion of efficacy. There is certainly a need for larger scale
3.6.1. Recommendation 16 trials to demonstrate efficacy of some of the pharmacologic
Because vascular risk factors and comorbidities impact interventions, and our recommendations might have to be
on the development and expression of dementia, they altered in the future as such trials are completed.
290 F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291

Author Disclosures [4] Lindstrom HA, Fritsch T, Petot G, Smyth KA, Chen CH, Debanne
SM, et al. The relationships between television viewing in midlife and
Fadi Massoud has had associations with Janssen Ortho the development of Alzheimer’s disease in a case-control study. Brain
Inc. (Advisory Board, CME Speaker, Research Funds), Cogn 2005;58:157– 65.
Lundbeck Canada (Advisory Board, CME Speaker, Re- [5] Wilson RS, Barnes LL, Mendes de Leon CF, Aggarwal NT, Schnei-
der JS, Bach J, et al. Participation in cognitively stimulating activities
search Funds), Novartis (Advisory Board, CME Speaker,
and risk of incident Alzheimer disease. JAMA 2002;287:742– 8.
Research Funds), and Pfizer (Advisory Board, CME [6] Verhaeghen P, Marcoen A, Goossens L. Improving memory perfor-
Speaker, Research Funds). mance in the aged through mnemonic training: a meta-analytic study.
Howard Chertkow has had associations with Pfizer Can- Psychol Aging 1992;7:242–51.
ada (Advisory Board, Speaker, Grant Recipient), Neuro- [7] Ball K, Berch DB, Helmers KF, Jobe JB, Leveck MD, Marsike M, et
chem Inc. (Advisory Board), and Lundbeck Canada (Advi- al. Effects of cognitive training interventions with older adults: a
randomized controlled trial. JAMA 2002;288:2271– 81.
sory Board, Speaker).
[8] Olazarán J, Muniz R, Resiberg B, Pena-Casanova J, del Ser T,
Ziad Nasreddine has had associations with Pfizer (Con- Cruz-Jentoft AJ, et al. Benefits of cognitive-motor intervention in
sultant, Adviser, Program Organizer, Lecturer), Novartis MCI and mild to moderate Alzheimer disease. Neurology 2004;63:
(Lecturer, Drug Trials, Advisor), Janssen Ortho Inc. (Lec- 2348 –53.
turer, Drug Trials, Advisor), Neurochem (Drug Trial), Myr- [9] Rapp S, Brenes G, Marsh AP. Memory enhancement training for
iad (Drug Trial), and Sanofi-Aventis (Drug Trial). older adults with mild cognitive impairment: a preliminary study.
Aging Ment Health 2002;6:5–11.
Howard Bergman has had associations with Pfizer (Drug
[10] Gunther VK, Schafer P, Holzner BJ, Kemmler GW. Long-term im-
Trial) and Novartis (Drug Trial). provements in cognitive performance through computer-assisted cog-
Morris Freedman has had associations with Pfizer (Ad- nitive training: a pilot study in a residential home for older people.
visory Board, CME Speaker, Research Funds), Janssen- Aging Ment Health 2003;7:200 – 6.
Ortho Inc. (Advisory Board, CME Speaker), Novartis (Ad- [11] Belleville S, Gilbert B, Fontaine F, Gagnon L, Menard E, Gauthier S.
visory Board, CME Speaker), Lundbeck (CME Speaker), Improvement of episodic memory in persons with mild cognitive
impairment and healthy older adults: evidence from a cognitive in-
Eli Lilly (Research Funds), and Orphan Medical (Research
tervention program. Dement Geriatr Cogn Disord 2006;22:486 –99.
Funds). [12] Lytle ME, Vander Bilt J, Pandav RS, Dodge HH, Ganguli M. Exer-
cise level and cognitive decline: the MoVIES project. Alzheimer Dis
Acknowledgments Assoc Disord 2004;18:57– 64.
[13] Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K.
This work was supported by grants from a number of Physical activity and risk of cognitive impairment and dementia in
agencies. Howard Chertkow is supported by operating elderly persons. Arch Neurol 2001;58:498 –504.
grants from the CIHR (Canadian Institutes for Health Re- [14] Wilson RS, Beckett LA, Barnes LL, Schneider JA, Bach J, Evans
DA, et al. Individual differences in rates of change in cognitive
search), the Alzheimer Society of Canada, and the Fonds de abilities of older persons. Psychol Aging 2002;17:179 –93.
la recherche en santé du Québec (FRSQ). [15] Colcombe S, Kramer AF. Fitness effects on the cognitive function of
Howard Chertkow and Sylvie Belleville receive “cher- older adults: a meta-analytic study. Psychol Sci 2003;14:125–30.
cheur national” awards from the FRSQ (Fonds de la Re- [16] Heyn P, Abreu BC, Ottenbacher KJ. The effects of exercise training
cherche en Santé du Québec). on elderly persons with cognitive impairment and dementia: a meta-
Howard Bergman is supported through the Dr Joseph Kauf- analysis. Arch Phys Med Rehabil 2004;85:1694 –704.
[17] DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski S,
mann Chair in Geriatric Medicine at McGill University.
Bennett DA, et al. Upregulation of choline acetyltransferase activity
Yves Joanette was supported by a grant from the Cana- in hippocampus and frontal cortex of elderly subjects with mild
dian Institutes of Health Research (MOP-15006). cognitive impairment. Ann Neurol 2002;51:145–55.
Sylvie Belleville is supported by research grants from the [18] Terry AV Jr, Buccafusco JJ. The cholinergic hypothesis of age and
Canadian Institutes of Health Research (CIHR), Heart and Alzheimer’s disease-related cognitive deficits: recent challenges and
Stroke Foundation of Canada, Alzheimer’s Society of Can- their implications for novel drug development. J Pharmacol Exp Ther
2003;306:821–7.
ada, CAREC, and NSERC.
[19] Salloway S, Ferris S, Kluger A, Goldman R, Griesing T, Kumar D, et
al. Efficacy of donepezil in mild cognitive impairment: a randomized
References placebo-controlled trial. Neurology 2004;63:651–7.
[20] Petersen RC, Thomas RG, Grundman M, Bennet D, Doody R, Ferris
[1] Chertkow H. Mild cognitive Impairment and very early stage Alz- S, et al. Vitamin E and donepezil for the treatment of mild cognitive
heimer’s disease. In: Gauthier S, ed. Clinical diagnosis and manage- impairment. N Engl J Med 2005;352:2379 – 88.
ment of Alzheimer’s disease, third edition. London: Informa UK; [21] Blacker D. Mild cognitive impairment: no benefit from vitamin E,
2006. p. 205–21. little from donepezil. N Engl J Med 2005;352:2439 – 41.
[2] Chertkow H. Emerging pharmacological therapies for mild cognitive [22] Ferris S, Feldman H, Sfikas N, Mancione L, Lane R. Time to clinical
impairment. In: Hultsch IHTaD, ed. Mild cognitive impairment: inter- diagnosis of dementia in MCI subjects receiving rivastigmine: a
national perspectives. New York: Taylor & Francis; 2006. p. 217– 43. randomized, double-blind, placebo-controlled study (abstract). Int
[3] Kramer AF, Bherer L, Colcombe SJ, Dong W, Greenough WT. Psychogeriatr 2005;17:242.
Environmental influences on cognitive and brain plasticity during [23] Feldman HH, Ferris S, Winblad B, Sfikas N, Mancione L, He Y, et al.
aging. J Gerontol A Biol Sci Med Sci 2004;59:M940 –57. Effect of rivastigmine on delay to diagnosis of Alzheimer’s disease
F. Massoud et al. / Alzheimer’s & Dementia 3 (2007) 283–291 291

from mild cognitive impairment: the InDDEx study. Lancet Neurol [35] Miller ER 3rd, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ,
2007;6:501–12. Guallar E. Meta-analysis: high-dosage vitamin E supplementation
[24] Scheltens P, Fox NC, Barkhof F, Gold M. Effect of galantamine may increase all-cause mortality. Ann Intern Med 2005;142:37– 46.
treatment on brain atrophy as assessed by MRI in patients with mild [36] Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM.
cognitive impairment. Neurobiol Aging 2004;25(Suppl 2):S270 –1. Diabetes mellitus and the risk of dementia: the Rotterdam Study.
[25] Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflammatory Neurology 1999;53:1937– 42.
drugs on risk of Alzheimer’s disease: systematic review and meta- [37] Yaffe K, Blackwell T, Kanaya AM, Davidowitz N, Barrett-Connor E,
analysis of observational studies. BMJ 2003;327:128 –31. Krueger K. Diabetes, impaired fasting glucose, and development of
cognitive impairment in older women. Neurology 2004;63:658 – 63.
[26] Thal LJ, Ferris SH, Kirby L, Block GA, Lines CR, Yuen E, et al. A
[38] Yaffe K, Kanaya A, Lindquist K, Simonsick EM, Harris T, Shorr
randomized, double-blind, study of rofecoxib in patients with mild
RI, et al. The metabolic syndrome, inflammation, and risk of
cognitive impairment. Neuropsychopharmacology 2005;30:1204 –15.
cognitive decline. JAMA 2004;292:2237– 42.
[27] Massoud F, Yaffe K, Sano M. Estrogen. In: Qizillbash N, ed. Evidence-
[39] Elias MF, Elias PK, Sullivan LM, Wolf PA, D’Agostino RB.
based dementia practice. Oxford, UK: Blackwell Science; 2002. p. Lower cognitive function in the presence of obesity and hyperten-
523– 6. sion: the Framingham heart study. Int J Obes Relat Metab Disord
[28] Shumaker SA, Legault C, Rapp SR, Thal L, Wallace RB, Ockene 2003;27:260 – 8.
JK, et al. Estrogen plus progestin and the incidence of dementia [40] Forette F, Seux ML, Staessen JA, Thijs L, Birkenhäger WH, Babar-
and mild cognitive impairment in postmenopausal women: the skiene MR, et al. Prevention of dementia in randomised double-blind
Women’s Health Initiative Memory Study—a randomized con- placebo-controlled Systolic Hypertension in Europe (Syst-Eur) trial.
trolled trial. JAMA 2003;289:2651– 62. Lancet 1998;352:1347–51.
[29] Yaffe K, Krueger K, Cummings SR, Blackwell T, Henderson VW, [41] Tzourio C, Anderson C, Chapman N, Woodward N, Neal B, Mac-
Sarkar S, et al. Effect of raloxifene on prevention of dementia and Mahon S, et al. Effects of blood pressure lowering with perindopril
cognitive impairment in older women: the Multiple Outcomes of and indapamide therapy on dementia and cognitive decline in patients
Raloxifene Evaluation (MORE) randomized trial. Am J Psychiatry with cerebrovascular disease. Arch Intern Med 2003;163:1069 –75.
2005;162:683–90. [42] Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe
[30] van Dongen M, van Rossum E, Kessels A, Sielhorst H, Knipschild P. SM, et al. Pravastatin in elderly individuals at risk of vascular
Ginkgo for elderly people with dementia and age-associated memory disease (PROSPER): a randomised controlled trial. Lancet 2002;360:
impairment: a randomized clinical trial. J Clin Epidemiol 2003;56: 1623–30.
367–76. [43] Collins R, Armitage J, Parish S, Sleight P, Peto R. Effects of cholesterol-
lowering with simvastatin on stroke and other major vascular events in
[31] Birks J, Grimley EV, Van Dongen M. Ginkgo biloba for cogni-
20536 people with cerebrovascular disease or other high-risk conditions.
tive impairment and dementia. Cochrane Database Syst Rev 2002.
Lancet 2004;363:757– 67.
CD003120.
[44] Starr JM, Whalley LJ, Deary IJ. The effects of antihypertensive
[32] Morris MC, Evans DA, Bienias JL, Tangney CC, Wilson RS.
treatment on cognitive function: results from the HOPE study. J Am
Vitamin E and cognitive decline in older persons. Arch Neurol Geriatr Soc 1996;44:411–5.
2002;59:1125–32. [45] Ishizaki J, Meguro K, Ohe K, Kimura E, Tsuchiya E, Ishii H, et al.
[33] Engelhart MJ, Gerlings MI, Ruitenberg A, van Swieten JC, Hofman Therapeutic psychosocial intervention for elderly subjects with very
A, Witteman JC, et al. Dietary intake of antioxidants and risk of mild Alzheimer disease in a community: the tajiri project. Alzheimer
Alzheimer disease. JAMA 2002;287:3223–9. Dis Assoc Disord 2002;16:261–9.
[34] Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grund- [46] Poon P, Hui E, Dai D, Kwok T, Woo J. Cognitive intervention for
man M, et al. A controlled trial of selegiline, alpha-tocopherol, or community-dwelling older persons with memory problems: telemedi-
both as treatment for Alzheimer’s disease: the Alzheimer’s Dis- cine versus face-to-face treatment. Int J Geriatr Psychiatry 2005;20:
ease Cooperative Study. New Engl J Med 1997;336:1216 –22. 285– 6.
Alzheimer’s & Dementia 3 (2007) 292–298

Clinical diagnosis of dementia


Alain Robillard
Division of Neurology, Department of Medicine, Hôpital Maisonneuve-Rosemont & Université de Montréal, Montréal, Quebec, Canada

Abstract This paper presents recommendations deriving from the Third Canadian Consensus Conference on
the Diagnosis and Treatment of Dementia, concerning the clinical diagnosis of dementia. There are
currently no universally accepted biological or radiological markers of dementia. In the absence of
these, the diagnosis of dementia remains a clinical exercise aiming to integrate all available clinical
and laboratory information. It is proposed that the currently used National Institute of Neurological
and Communicative Disorders and Stroke/Alzheimer’s Disease and Related Disorders Association
(NINCDS/ADRA) criteria for diagnosis of Alzheimer’s disease (AD) be retained. The currently
available vascular dementia (VaD) diagnostic criteria have variable accuracy. An integrative
approach to VaD diagnosis based on all the available evidence (history, vascular risk factors,
physical exam, clinical course, neuroimaging, cognitive impairment pattern) is recommended. The
separation of Lewy body dementia (DLB) from Parkinson’s disease dementia (PDD) is based on the
dominant clinical presenting feature of each syndrome, and relies on the duration of this feature:
long duration of parkinsonian “motor” syndrome preceding dementia for PDD versus early/initial
dementia accompanied by extrapyramidal symptoms for DLB. It is recognized that it impossible
clinically to characterize DLB with (pathologically) coexisting AD changes. The Frontotemporal
group of dementia syndromes are discussed in regards to their typical clinical pictures, recognizing
that their neuropathological substrate are not predictable from their mode of presentation. Finally,
the particular rapid time sequence of evolution of the dementias due to prior disease is recognized
as the clinically most useful distinguishing feature of these syndromes.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Diagnosis; Alzheimer; Lewy body; Frontotemporal; Vascular; Behavior

1. Introduction paper in this volume) by Rockwood et al, which addresses


the question of clinical criteria from a different perspective.
This paper presents recommendations deriving from the
That paper critically reviews the literature and proposes
Third Canadian Consensus Conference on the Diagnosis
changes in the current criteria. The recommendations of this
and Treatment of Dementia, concerning the clinical diag-
paper pertain to how physicians should apply the current
nosis of dementia. These recommendations are the result of
criteria as presently understood. The purpose of the Rock-
a standardized literature review (see series Introduction for
wood et al paper is to propose changes which might lead in
methods) and all of the recommendations were supported by
at least 80% of the participants at the national meeting held the future to more cogent or coherent conceptualization of
in Montreal in 2006. Our focus was on whether there were the dementias. These are complementary goals.
clearly agreed upon clinical diagnostic criteria for different The diagnosis of dementia and of its various etiologies
dementia entities, and if so, what those recommended cri- remain at its core a clinical exercise based on the presenting
teria were. Note that there is a later paper (in fact the final and dominant symptoms, their order of appearance and the
evolution of symptoms and signs over time. A number of
classification criteria based on this procedure have been
Corresponding author: Alain Robillard Tel.: (514) 252-3528; Fax:
proposed through the years for Alzheimer’s disease (AD)
(514) 252-3529. and vascular dementia (VaD), with various precision when
E-mail address: robillal@videotron.ca compared with diagnosis based on pathological outcome.
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.08.002
A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298 293

The same applies to the Frontotemporal group of dementias, (89%, compared with pathology) (Class II, one trial). The
where pathological diagnosis is more difficult to predict on diagnostic accuracy is enhanced in this group of patients by
clinical grounds alone. The problem is similar in the Lewy the use of a more comprehensive neuropsychological test
Body dementias (Parkinson’s disease dementia and Lewy battery [24].
Body dementia) where there exist a considerable overlap in Patients with mild cognitive impairment (MCI), as de-
the presenting symptoms, and where again neuropathology fined by Petersen et al [25], have a memory impairment
cannot always be relied on to “confirm” a clinical diagnosis. beyond that expected for age and education, yet they are not
Here the sequence of appearance of the cardinal symptoms demented. These subjects have become the focus of many
in both syndromes is central to a clinical diagnosis. In all of prediction studies and are diagnosed according to clinical
these dementias, a descriptive and “integrative” approach to criteria. Debate is still ongoing regarding the specific sub-
diagnosis is proposed in the following discussions. types of MCI (amnestic and non-amnestic, single or multi-
The essential symptoms of dementia are “. . . an acquired ple domains), with definitions used on the basis of a pre-
impairment in short and long-term memory, associated with sumed etiology. With the criteria of Petersen et al for
impairment in abstract thinking, impaired judgment, other amnestic MCI, it has been shown that a high proportion of
disturbances of higher cortical function, or personality these patients go on to AD [25–28]. The definition of MCI
changes. The disturbance is severe enough to interfere with is difficult to apply in a clinical setting, and the available
work or usual social activities or relationships with others. diagnostic instruments used for diagnosing dementia lack
The diagnosis of dementia is not made if these symptoms sensitivity to MCI. A new tool, the Montreal Cognitive
occur in the presence of Delirium.” This definition is the Assessment (MoCA), has been proposed and validated in
most widely used in practice and is based on the Diagnostic one trial [29], demonstrating a higher sensitivity for MCI
and Statistical Manual [of Mental Disorders], Third Edition, (100% when judged against the MMSE, with a cutoff score
Revised (DSM-IIIR), DSM-IV, and the National Institute of of 26), with good specificity (87%). A separate paper by
Neurological and Communicative Disorders and Stroke/ Chertkow et al in this issue further pursues the concept of
Alzheimer’s Disease and Related Disorders Association MCI and derives evidence-based recommendations for MCI
(NINCDS/ADRDA) [1–3]. diagnosis.
This DSM III-R definition of dementia has good reliabil-
ity particularly with respect to Alzheimer’s disease. The
3. VaD
DSM-IV version has not been validated, although it is
identical to the former definition [2,4,5]. VaD is a dementia syndrome resulting from cerebrovas-
After identifying the presence of a dementia syndrome, cular damage. This syndrome can occur as the result of a
the specific causes can be recognized by using clinical single strategically placed brain infarct or as the result of
criteria for the various clinical profiles. numerous discrete smaller (and most often subcortical) le-
sions. The progression is classically in a stepwise fashion,
2. AD but increasingly a slow insidious progress is recognized.
The clinical hallmark of VaD is dysexecutive syndrome.
AD is characterized by gradual onset and continued Four consensus criteria for VaD are in use: the State of
decline of memory and at least one additional cognitive California AD Diagnostic and Treatment Centers criteria
domain that is not explained by another systemic or neuro- (the California criteria), the National Institute of Neurologic
logic disorder [3]. Disorders and Stroke and the Association Internationale
Both the reliability of the clinical diagnosis and inter- pour la Recherche et l’Enseignement en Neurosciences
rater reliability have been subjected to review, with more (NINDS-AIREN) criteria, the Hachinski Ischemic Score
than 14 studies addressing these concerns [6 –23]. The gen- (HIS) modified by Rosen, and those found in the DSM-IV.
eral consensus is that on the basis of Class I and Class II They all have poor sensitivity at the cost of high specificity,
studies and with neuropathologic data as confirming evi- when judged against the best clinical judgment (including
dence of clinical diagnosis, there is very good sensitivity of neuroimaging) of trained clinicians [30] (Class II evidence).
clinical criteria (average, 81%; range, 49% to 100%), at the In studies that compared clinical diagnoses and neuropatho-
expense of specificity (average, 70%; range, 47% to 100%), logic findings, the NINDS-AIREN and the California crite-
with the NINCDS/ADRDA definition of “probable” AD. ria (as well as DSM-IIIR) had very low sensitivity but
“Possible” AD, as a diagnostic category, achieves higher higher specificity. One Class I study [31] reported the sen-
sensitivity (average, 93%) but much lower specificity (av- sitivity (43%) and specificity (95%) of the NINDS-AIREN
erage, 48%); this reflects the fact that there are many com- criteria for VaD. Five Class II studies [32–36] reported
mon features between different types of dementia. sensitivity and specificity of the diagnosis of VaD with any
“Mild” AD (as defined by a Mini Mental State Exami- criteria. The results showed low sensitivity (average of 5
nation [MMSE] score of ⱖ24) can also be diagnosed ac- studies, 50%; range, 20% to 89%) but high specificity (av-
cording to clinical criteria, with a high degree of certainty erage across 5 studies, 87%; range, 64% to 98%) for the
294 A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298

HIS, DSM-IIIR, NINDS-AIREN, or California clinical cri- The recognition that AD and DLB neuropathology can
teria. Only one retrospective (Class II) study [35] showed coexist to various degrees in patients presenting initially
better sensitivity and specificity (both 89%) with the HIS. with either AD or DLB symptomatology has lead to at-
This lack of sensitivity in the available diagnostic criteria tempts at clinical differentiation. In a Class II study, Lopez
is confounded by the presence on neuropathologic exami- et al [49] reported that in patients with pathologically diag-
nation of considerable overlap between vascular and degen- nosed AD and DLB, sensitivity of the clinical diagnostic
erative (AD) pathology (some vascular pathology exists in criteria improved as dementia worsened, at the expense of
29% to 41% of dementia cases coming to autopsy), whereas specificity. There were no distinguishing features between
pure vascular pathology accounts for dementia in only 9% AD and AD with DLB in mild dementia. In moderate
to 10% [12,14], leading some to propose that pure VaD or dementia, the presence of extrapyramidal signs (EPS) and
AD is rare [12–17]. major depression were associated with AD ⫹ DLB, and in
A descriptive approach to the diagnosis of VaD is pro- severe dementia, EPS and diurnal hypersomnia were indi-
posed, which would take into account the neuropsycholog- cators of AD ⫹ DLB.
ical profile of the dementia syndrome, neuroimaging, and Del Ser et al [50] noted in a Class II study that the clinical
vascular risk factors [37,38]. features of AD ⫹ DLB patients were similar to those of AD
patients except for more frequent acute-subacute onset and a
4. Lewy body disease fluctuating evolution. Differentiation between three groups
of patients (pure DLB, AD ⫹ LB, and AD) or between both
The third report of the Dementia with Lewy Bodies groups with LB (DLB) from AD could only be attained in
(DLB) consortium was published in December 2005 [39]. 70% of cases. Accuracy was excellent for the diagnosis of
The central feature of DLB remains the presence of a pure DLB but only mediocre for separating AD ⫹ LB as
dementia syndrome of sufficient intensity to interfere with well as the pure DLB and DLB ⫹ AD groups from AD.
social or occupational function, although memory impair- In a Class II retrospective study of 90 patients with
ment might not be prominent at diagnosis, whereas deficits autopsy-proven DLB, Merdes et al [51] noted that only 27%
on tests of attention, visuospatial ability, and executive of these demonstrated both visual hallucinations and spon-
function might be more prominent. The core features in- taneous EPS. Subjects with lower tangle burden on Braak
clude spontaneous parkinsonian symptoms, fluctuating cog- staging had a higher frequency of visual hallucinations
nition with pronounced variations in attention and alertness, (65%) than did subjects with DLB with higher Braak stages
and recurrent well-formed and detailed visual hallucina- (33%) and showed a slightly greater but not significant
tions. The consensus report also details suggestive as well as degree of EPS. Although clinical diagnostic accuracy for
supportive features that add a degree of probability to the DLB was relatively low (49%), it was higher for subjects
diagnosis. Clinically the most important supportive symp- with low (75%) compared with high (39%) Braak stages.
toms remain rapid eye movement sleep behavior disorder The degree of concomitant AD tangle pathology had an
and severe neuroleptic sensitivity. important influence on the clinical characteristics and the
The time sequence in which symptoms of DLB appear is accuracy of clinical diagnosis of DLB.
key to differentiation from Parkinson’s disease dementia
(PDD); typically dementia appears after a number of years 5. Frontotemporal dementia
in PD (10 years usually), whereas parkinsonian symptoms
are central to the initial diagnosis of DLB. There are no The clinical presentation in frontotemporal dementia
distinguishing clinical features that can discriminate be- (FTD) reflects the distribution of the pathologic changes
tween PDD and DLB at any given point in time [40]. rather than the exact histologic subtype of the disease. Three
The sensitivity of the DLB diagnosis based on pathology clinical syndromes are recognized, all of insidious onset.
has been subject to controversy [41] due in large part to the 5.1. Frontal variant FTD
original pathologic reports stating that the presence of any
LBs in whatever distribution was sufficient for diagnosis Frontal variant FTD (dementia of frontal type), in which
[42,43]. One Class I study [12] investigated the diagnostic changes in social behavior and personality predominate,
accuracy of DLB criteria against neuropathologic findings; reflects the orbitobasal frontal lobe focus of the pathology.
sensitivity was low at 22%, but specificity was high (100%). 5.2. Semantic dementia
Six Class II studies also showed low sensitivities (average
across 5 studies, 58%; range, 30% to 75%) but higher In semantic dementia (progressive fluent aphasia) there
specificities (average across 5 studies, 87%; range, 71% to is a breakdown in the conceptual database that underlies
100%) for the Consortium diagnostic criteria of DLB language production and comprehension. Patients with se-
[10,44 – 48]. New pathologic criteria have been proposed by mantic dementia have asymmetric anterolateral temporal
the DLB Consortium that will take into account the con- atrophy with relative sparing of the hippocampal formation,
comitant presence of AD pathology in the future [39]. typically worse on the left side.
A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298 295

5.3. Progressive nonfluent aphasia sive dementia, periodic sharp waves in the EEG, and at least
two of the following four findings: myoclonus, visual and/or
In progressive nonfluent aphasia the phonologic and syn-
cerebellar symptoms, pyramidal and/or extrapyramidal
tactic components of language are affected in association
signs, and akinetic mutism. Cases that fulfill these criteria
with left perisylvian atrophy [52].
but do not have a typical EEG are classified as possible
The original Lund-Manchester criteria [53] and the Con-
CJD, according to the criteria of Masters et al [65]. Other
sensus revision [54] have been tested against pathology,
criteria have been proposed (French [66] and European
with low specificity; an autopsy-based Class II study (based
criteria [67]). In a prospective Class I study Poser et al [68]
on retrospective clinical diagnoses) showed that most pa-
tested the diagnostic criteria for CJD. Diagnosis was con-
tients with FTD fulfill diagnostic criteria for AD [55]. In
firmed in 188 autopsy-confirmed cases (97%) of 193 cases
contrast, a Class II study without autopsy confirmation [56]
diagnosed with probable CJD. In this cohort, among 54
found that the Lund-Manchester criteria differentiated
cases diagnosed with possible CJD, the diagnosis was con-
100% of FTD and AD patients. A recent Class II retrospec-
firmed in 44 (81%). Only two pathologically diagnosed CJD
tive study concluded that FTD, corticobasal degeneration,
cases were found among 111 patients who had been given
and progressive supranuclear palsy have overlapping clini-
other clinical diagnoses. Thus the criteria achieved high
cal features, with FTD being an unpredictable disease in
sensitivity and specificity.
terms of its tau-positive biochemistry [57].
In a Class II retrospective trial Brandel et al [69] com-
pared three sets of clinical diagnostic criteria in a cohort of
6. Normal pressure hydrocephalus 428 patients, showing that the three sets of criteria had a
high specificity and low sensitivity as judged against neu-
The classic clinical presentation of normal pressure hy- ropathology; all three criteria had poor negative predictive
drocephalus (NPH) is one of progressive gait apraxia, uri- value (average, 44%; range, 29% to 57%).
nary incontinence, and ultimately dementia [58 – 60]. There An immunoassay for the detection of the 14-3-3 protein
have been a number of reports of amelioration of the clinical in CSF was described in 1996 [70] that had a reported
picture through ventriculoperitoneal shunting but no posi- specificity of 99% and a sensitivity of 96% for the diagnosis
tive predictive test to differentiate those patients who might of CJD among patients with dementia who had not had a
benefit from the procedure from those who will not. One stroke within 1 month of testing. During the ensuing years
trial of temporary external lumbar cerebrospinal fluid (CSF) it has been shown that the 14-3-3 assay is not valid to
drainage proved negative because of a high rate of false- discriminate between unselected rapidly progressing de-
negative results [61] (Class II); although the predictive mentia syndromes. Burkhard et al [71] did a Class I study
value of a positive external lumbar drainage was high, the but validated against clinical diagnosis, showing 14 of 100
costs and invasiveness of the test and the possibility of positive patients, of whom only two had a final diagnosis of
serious test-related complications limit its usefulness in CJD. There is also variation in the sensitivity of the 14-3-3
managing patients with presumed NPH. assay among CJD subtypes. In a Class II study Castellani
Lately there has been interest for low-flow CSF drainage et al [72] reported sensitivity between 77% and 94%, ac-
as a treatment for AD. Advancing age is the key risk factor cording to subtypes of CJD; the lower sensitivity was re-
for developing AD. With aging, abnormal metabolism and ported in the less frequent atypical subtypes.
clearance of amyloid beta proteins (A␤) can lead to accu- Magnetic resonance imaging (MRI), especially with
mulation of these proteins in brain parenchyma, resulting in diffusion-weighted imaging, in a Class II study but with a
plaque formation. It has been postulated that age-associated small number of subjects [73], showed evidence of a com-
impairment in the CSF circulation and cerebral vasculature parable sensitivity to 14-3-3 assay. In a prospective study of
could play a role, through diminished clearance of A␤, in 219 patients suspected of having CJD, the presence of basal
the high incidence of AD in the elderly [62,63]. The result ganglia high signal on T2-weighted MRI showed a sensi-
of one pilot trial of low-flow CSF drainage has been re- tivity of 63% and a specificity of 88% in definite/probable
ported, indicating a small stabilizing effect on dementia CJD [74].
severity rating (Class III) [64]. There is one ongoing trial for
which no results have been published.
8. Recommendations approved at the Third Canadian
7. Prion diseases Consensus Conference on Diagnosis and Treatment
of Dementia
The clinical diagnosis of Creutzfeldt-Jakob disease (CJD)
rests on the occurrence of a rapidly progressing dementia
syndrome, associated with myoclonus and a characteristic (1) The diagnosis of dementia remains clinical. There
electroencephalogram (EEG) pattern of periodic sharp waves. is good evidence to retain the diagnostic criteria
Probable CJD is diagnosed if patients have rapidly progres- currently in use (Grade A, Level 2).
296 A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298

(2) The sensitivity of clinical diagnosis for possible or (10) CJD is a distinctively rapidly progressive dementia
probable AD based on the NINCDS-ADRDA cri- that in Canada requires reporting to the CJD Sur-
teria remains high. The specificity is lower. The veillance Network and requires special infection
continued use of the NINCDS-ADRDA criteria is control procedures. Its diagnosis is supported by a
recommended (Grade A, Level 1). positive 14-3-3 test on CSF and by an abnormal
(3) Mild AD can be diagnosed with a high degree of MRI scan, with either basal ganglia high signal on
specificity, when the presenting clinical picture is T2 or abnormalities on diffusion-weighted imaging,
one of memory impairment (Grade B, Level 1). and by a progressively worsening EEG with peri-
(4) The currently available VaD diagnostic criteria odic complexes. Specialist referral is recommended
have variable accuracy. An integrative approach to (Grade B, Level 2).
VaD diagnosis based on all the available evidence
(history, vascular risk factors, physical exam, clin-
ical course, neuroimaging, cognitive impairment Author Disclosures
pattern) is recommended (Grade B, Level 2).
(5) The clinical features of DLB and PDD overlap con- Alain Robillard has received support from Pfizer Canada
siderably. At present, DLB should be diagnosed when (Speaker, Administrative Board Member, Investigator), No-
this pattern of dementia occurs before or concurrently vartis (Speaker, Administrative Board Member, Investiga-
with parkinsonism. PDD can be diagnosed when de- tor), Janssen Ortho Inc. (Speaker, Administrative Board
mentia occurs in the context of well-established PD, Member, Investigator), Lundbeck Canada (Speaker, Ad-
generally after many years (Grade B, Level 3). ministrative Board Member, Investigator), Sanofi-Aventis
(6) There is frequent coexistence of AD and LB (Clinical Investigator).
neuropathology in subjects presenting with the
initial clinical picture of either pathology. At
present, it is impossible to propose clinical References
guidelines that would permit separation of the
two diagnoses and AD ⫹ DLB with a high spec- [1] American Psychiatric Association. Diagnostic and statistical manual
ificity (Grade A, Level 2). of mental disorders. 4th ed. Washington, DC: American Psychiatric
Association; 1994.
(7) In patients presenting primarily with progressive [2] American Psychiatric Association. Diagnostic and statistical manual
decline in language or praxis or with prominent of mental disorders. 3rd ed–revised. Washington, DC: American
changes in behavior or personality, Pick complex dis- Psychiatric Association; 1987.
ease (also known as FTD or frontotemporal lobar [3] McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan
degeneration) should be considered. These disorders EM. Clinical diagnosis of Alzheimer’s disease: report of the
NINCDS-ADRDA Work Group under the auspices of Department of
of Pick complex include semantic dementia, FTD Health and Human Services Task Force on Alzheimer’s Disease.
behavioral variant, primary progressive aphasia, Neurology 1984;34:939 – 44.
corticobasal degeneration, progressive supranuclear [4] O’Connor DW, Blessed G, Cooper B, Jonker C, Morris JC, Presnell
palsy, and FTD with motor neuron disease. These IB, et al. Cross-national interrater reliability of dementia diagnosis in
disorders have clinical features that are distinctive the elderly and factors associated with disagreement. Neurology
1996;47:1194 –9.
and are best referred for specialist care (Grade A, [5] Graham JE, Rockwood K, Beattie BL, McDowell I, Eastwood R,
Level 2). Gauthier S. Standardization of the diagnosis of dementia in the
(8) When gait apraxia or urinary incontinence occurs Canadian Study of Health and Aging. Neuroepidemiology 1996;15:
early in the course of dementia, NPH should be 246 –56.
considered and should be supported by computed [6] Larson EB, McCurry SM, Graves AB, Bowen JD, Rice MM, Mc-
Cormick WC, et al. Standardization of the clinical diagnosis of the
tomography or MRI. Specialist referral is advised dementia syndrome and its subtypes in a cross-sectional study: the
for further assessment. The diagnostic work-up Ni-Hon-Sea experience. J Gerontol A Biol Sci Med Sci 1998;53A:
might include the removal of a large CSF volume M313–9.
with documentation of clinical response, if surgical [7] Fratiglioni L, Grut M, Forsell Y, Viitanen M, Winblad B. Clinical
intervention is considered an appropriate option diagnosis of Alzheimer’s disease and other dementias in a population
survey: agreement and causes of disagreement in applying Diagnostic
(Grade B, Level 2). and Statistical Manual of Mental Disorders, Revised Third Edition,
(9) Rapidly progressing dementia associated with my- criteria. Arch Neurol 1992;49:927–2.
oclonus and an EEG with the presence of periodic [8] Farrer LA, Cupples LA, Blackburn S, Kiely DK, Auerbach S, Grow-
sharp waves is typical of CJD. There are three don JH, et al. Interrater agreement for diagnosis of Alzheimer’s
clinical criteria sets in use to diagnose CJD, all with disease: the MIRAGE study. Neurology 1994;44:652– 6.
[9] Blacker D, Albert MS, Bassett SS, Go RC, Harrell LE, Folstein MF.
some specificity. The recognition of rapid progres- Reliability and validity of NINCDS-ADRDA criteria for Alzheimer’s
sion in a dementia syndrome should by itself sug- disease: the National Institute of Mental Health Genetics Initiative.
gest the possibility of CJD (Grade A, Level 2). Arch Neurol 1994;51:1198 –204.
A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298 297

[10] Lopez OL, Litvan I, Catt KE, Stowe R, Klunk W, Kaufer DI, et al. [29] Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R, Ferris
Accuracy of four clinical diagnostic criteria for the diagnosis of S, et al. Vitamin E and donepezil in the treatment of mild cognitive
neurodegenerative dementias. Neurology 1999;53:1292–9. impairment. N Engl J Med. 2005;352:2379 – 88.
[11] Lopez OL, Becker JT, Klunk W, Saxton J, Hamilton RL, Kaufer DI, [30] Nasreddine ZS, Phillips NA, Bedirian V, Charbonneau S, Whitehead
et al. Research evaluation and diagnosis of probable Alzheimer’s V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief
disease over the last two decades: I. Neurology 2000;55:1854 – 62. screening tool for mild cognitive impairment. J Am Geriatr Soc
[12] Forette F, Henry JF, Orgogozo JM, et al. Reliability of clinical criteria 2005;53:695–9.
for the diagnosis of dementia: a longitudinal multicenter study. Arch [31] Rockwood K, Davis H, MacKnight C, Vandorpe R, Gauthier S,
Neurol 1989;46:646 – 8. Guzman A, et al. The Consortium to Investigate Vascular Impairment
[13] Holmes C, Cairns N, Lantos P, Mann A. Validity of current clinical of Cognition: methods and first findings. Can J Neurol Sci 2003;30:
criteria for Alzheimer’s disease, vascular dementia and dementia with 237– 43.
Lewy bodies. Br J Psychiatry 1999;174:45–50. [32] Gold G, Giannakopoulos P, Montes-Paixao Junior C, et al. Sensitivity
[14] Jobst KA, Barnetson LP, Shepstone BJ. Accurate prediction of and specificity of newly proposed clinical criteria for possible vas-
histologically confirmed Alzheimer’s disease and the differential cular dementia. Neurology 1997;49:690 – 4.
diagnosis of dementia: the use of NINCDS-ADRDA and DSM- [33] Gold G, Bouras C, Canuto A, Bergallo Herrmenn FR, Hof P, Mayor
III-R criteria, spect, X-ray CT, and apo E4 in medial temporal lobe PA, et al. Clinicopathological validation study of four sets of clinical
dementias—Oxford Project to Investigate Memory and Aging. Int criteria for vascular dementia. Am J Psychiatry 2002;159:82–7.
Psychogeriatr 1998;10:271–302. [34] Pohjasvaara T, Mantyla R, Ylikoski R, Kaste M, Erkinjuntti T.
[15] Lim A, Tsuang D, Kukull W, Nochlin D, Leverenz J, McCormick W, Comparison of different clinical criteria (DSM-III, ADDTC, ICD-
et al. Clinico-neuropathological correlation of Alzheimer’s disease in 10,NINDS-AIREN, DSM-IV) for the diagnosis of vascular dementia.
a community-based case series. J Am Geriatr Soc 1999;47:564 –9. Stroke 2000;31:2952–7.
[16] Wade JP, Mirsen TR, Hachinski VC, Fisman M, Lau C, Merskey H. [35] Cosentino SA, Jefferson AL, Carey M, Price CC, Davis-Garrett K,
The clinical diagnosis of Alzheimer’s disease. Arch Neurol 1987;44: Swenson R, et al. The clinical diagnosis of vascular dementia: a
24 –9. comparison among four classification systems and a proposal for a
[17] Victoroff J, Mack WJ, Lyness SA, Chui HC. Multicenter clinicopath- new paradigm. Clin Neuropsychol 2004;18:6 –21.
[36] Chui HC, Mack W, Jackson JE, Mungas D, Reed BR, Tinklenberg J,
ological correlation in dementia. Am J Psychiatry 1995;152:1476 –
et al. Clinical criteria for the diagnosis of vascular dementia: a
84.
multicenter study of comparability and interrater reliability. Arch
[18] Galasko D, Hansen LA, Katzman R, Wiederholt W, Masliah E, Terry
Neurol 2000;57:191– 6.
R, et al. Clinical-neuropathological correlations in Alzheimer’s dis-
[37] Graham NL, Emery T, Hodges JR. Distinctive cognitive profiles in
ease and related dementias. Arch Neurol 1994;51:888 –95.
Alzheimer’s disease and subcortical vascular dementia. J Neurol
[19] Mayeux R, Saunders AM, Shea S, Mirra S, Evans D, Roses AD, et al.
Neurosurg Psychiatry 2004;75:61–71.
Utility of the apolipoprotein E genotype in the diagnosis of Alzhei-
[38] Tierney MC, Black SE, Szalai JP, Snow WG, Fisher RH, Nadon G,
mer’s disease: Alzheimer’s Disease Centers Consortium on Apoli-
et al. Recognition memory and verbal fluency differentiate probable
poprotein E and Alzheimer’s Disease. N Engl J Med 1998;338:506 –
Alzheimer disease from subcortical ischemic vascular dementia. Arch
11.
Neurol 2001;58:1654 –9.
[20] Klatka LA, Schiffer RB, Powers JM, Kazee AM. Incorrect diagnosis
[39] McKeith G, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H,
of Alzheimer’s disease: a clinicopathologic study. Arch Neurol 1997;
et al. Diagnosis and management of dementia with Lewy bodies: third
53:35– 42.
report of the DLB consortium Neurology 2005;65:1863–72.
[21] Gearing M, Mirra SS, Hedreen JC, Sumi SM, Hansen LA, Heyman [40] Noe E, Marder K, Bell KL, Jacobs DM, Manly JJ, Stern Y. Com-
A. The Consortium to Establish a Registry for Alzheimer’s Disease parison of dementia with Lewy bodies to Alzheimer’s disease and
(CERAD): part X—neuropathology confirmation of the clinical diag- Parkinson’s disease with dementia. Mov Disord 2004;19:60 –7.
nosis of Alzheimer’s disease. Neurology 1995;45:461– 6. [41] Litvan I, Bhatia KP, Burn DJ, Goetz CG, Lang AE, McKeith I, et al.
[22] Berg L, McKeel DW Jr., Miller JP, Storandt M, Rubin EH, Morris JC, SIC Task Force Appraisal of clinical diagnostic criteria for parkin-
et al. Clinicopathologic studies in cognitively healthy aging and sonian disorders. Mov Disord 2003;18:467– 86.
Alzheimer’s disease: relation of histologic markers to dementia se- [42] Kosaka K. Lewy bodies in cerebral cortex: report of three cases. Acta
verity, age, sex, and apolipoprotein E genotype. Arch Neurol 1998; Neuropathol 1978;42:127–34.
55:326 –35. [43] Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of Lewy
[23] Massoud F, Devi G, Stern Y, Lawton A, Goldman JE, Liu Y, et al. A body disease: progressive dementia with abundant cortical Lewy
clinicopathological comparison of community-based and clinic-based bodies and senile changes of varying degree: a new disease? Clin
cohorts of patients with dementia. Arch Neurol 1999;56:1368 –73. Neuropathol 1984;3:185–92.
[24] Salmon DP, Thomas RG, Pay MM, Booth A, Hofstetter CR, Thal LJ, [44] Litvan I, MacIntyre A, Goetz CG, Wenning GK, Jellinger K, Verny
et al. Alzheimer’s disease can be accurately diagnosed in very mildly M, et al. Accuracy of the clinical diagnoses of Lewy body disease,
impaired individuals. Neurology 2002;59:1022– 8. Parkinson disease, and dementia with Lewy bodies: a clinicopatho-
[25] Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen logic study. Arch Neurol 1998;55:969 –78.
E. Mild cognitive impairment: clinical characterization and outcome. [45] Luis CA, Barker WW, Gajaraj K, Harwood D, Petersen R, Kashuba
Arch Neurol 1999;56:303– 8. A, et al. Sensitivity and specificity of three clinical criteria for de-
[26] Petersen RC, Morris JC. Mild cognitive impairment as a clinical mentia with Lewy bodies in an autopsy-verified sample. Int J Geriatr
entity and treatment target. Arch Neurol 2005;62:1160 –3. Psychiatry 1999;14:526 –33.
[27] Petersen RC. Mild cognitive impairment clinical trials. Nat Rev Drug [46] Mega MS, Masterman DL, Benson DF, Vinters HV. Dementia with
Discov 2003;2:646 –53. Lewy bodies: reliability and validity of clinical and pathologic crite-
[28] Grundman M, Petersen RC, Ferris S, Thomas RG, Aisen PS, Bennett ria. Neurology 1996;47:1403–9.
DA, et al. Mild cognitive impairment can be distinguished from [47] Verghese J, Crystal HA, Dickson DW, Lipton RB. Validity of clinical
Alzheimer disease and normal aging for clinical trials. Arch Neurol criteria for the diagnosis of dementia with Lewy bodies. Neurology
2004;61:59 – 66. 1999;53:1974 – 82.
298 A. Robillard / Alzheimer’s & Dementia 3 (2007) 292–298

[48] Lopez OL, Becker JT, Kaufer D, Hamilton R, Sweet R, Klunk W, [62] Silverberg GD, Mayo M, Saul T, Rubenstein E, McGuire D. Alzhei-
et al. Research evaluation and prospective diagnosis of dementia with mer’s disease, normal□pressure hydrocephalus, and senescent
Lewy bodies. Arch Neurol 2002;59:43– 6. changes in CSF circulatory physiology: a hypothesis. Lancet Neurol-
[49] Lopez OL, Hamilton RL, Becker JT, Wisniewski S, Kaufer DI, 2003;2:506 –11.
DeKosky ST. Severity of cognitive impairment and the clinical di- [63] Silverberg GD, Heit G, Huhn S, Jaffe RA, Chang SD, Bronte-Stewart
agnosis of AD with Lewy bodies. Neurology 2000;54:1780 –7. H, et al. CSF production is reduced in dementia of the Alzheimer’s
[50] Del Ser T, Hachinski V, Merskey H, Munoz DG. Clinical and patho- type. Neurology 2001;57:1763– 6.
logic features of two groups of patients with dementia with Lewy [64] Silverberg GD, Levinthal E, Sullivan EV, Bloch DA, Chang SD,
bodies: effect of coexisting Alzheimer-type lesion load. Alzheimer Leverenz J, et al. Assessment of low-flow CSF drainage as a treat-
Dis Assoc Disord 2001;15:31– 44. ment for AD: results of a randomized pilot study. Neurology 2002;
[51] Merdes AR, Hansen LA, Jeste DV, Galasko D, Hofstetter CR, Ho GJ, 59:1139 – 45.
et al. Influence of Alzheimer pathology on clinical diagnostic accu- [65] Masters CL, Harris JO, Gajdusek DC, Gibbs CJ Jr, Bernoulli C,
racy in dementia with Lewy bodies. Neurology 2003;60:1586 –90. Asher DM. Creutzfeldt-Jakob disease: patterns of worldwide occur-
[52] Hodges JR. Frontotemporal dementia (Pick’s disease): clinical fea- rence and the significance of familial and sporadic clustering. Ann
tures and assessment. Neurology 2001;56(Suppl 4):S6 –10. Neurol 1979;5:177– 88.
[53] Clinical and neuropathological criteria for frontotemporal dementia. [66] Cathala F, Brown P, Castaigne P, Gajdusek DC. La maladie de
J Neurol Neurosurg Psychiatry 1994;57:416 – 8.
Creutzfeldt-Jakob en France continentale: etude rétrospective de
[54] Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, et
1968 –1977. Rev Neurol 1979;5:439 –54.
al. Frontotemporal lobar degeneration: a consensus on clinical diag-
[67] Concerted action of the EU. Surveillance of Creutzfeldt-Jakob disease
nostic criteria. Neurology 1998;51:1546 –54.
in the European community: minutes of the second meeting in Rome,
[55] Varma AR, Snowden JS, Lloyd JJ, Talbot PR, Mann DM, Neary D.
July 3, 1993, and the third meeting in Paris, April 29 –30, 1994.
Evaluation of the NINCDS-ADRDA criteria in the differentiation of
[68] Poser S, Mollenhauer B, Kraubeta A, Zerr I, Steinhoff BJ, Schroeter
Alzheimer’s disease and frontotemporal dementia. J Neurol Neuro-
A, et al. How to improve the clinical diagnosis of Creutzfeldt–Jakob
surg Psychiatry 1999;66:184 – 8.
disease. Brain 1999;122:2345–51.
[56] Miller BL, Ikonte C, Ponton M, Levy M, Boone K, Darby A, et al. A
[69] Brandel JP, Delasnerie-Laupretre N, Laplanche JL, Hauw JJ, Alpero-
study of the Lund-Manchester research criteria for frontotemporal
dementia: clinical and single-photon emission CT correlations. Neu- vitch A. Diagnosis of Creutzfeldt-Jakob disease: effect of clinical
rology 1997;48:937– 42. criteria on incidence estimates. Neurology 2000;54:1095–9.
[57] Josephs KA, Petersen RC, Knopman DS, Boeve BF, Whitwell JL, [70] Hsich G, Kenney K, Gibbs CJ, Lee KH, Harrington MG. The 14-3-3
Duffy JR, et al. Clinicopathologic analysis of frontotemporal and brain protein in cerebrospinal fluid as a marker for transmissible
corticobasal degenerations and PSP. Neurology 2006;66:41– 8. spongiform encephalopathies. N Engl J Med 1996;335:924 –30.
[58] Hakim S, Adams RD. The special clinical problem of symptomatic [71] Burkhard PR, Sanchez JC, Landis T, Hochstrasser D. CSF detection
hydrocephalus with normal cerebrospinal fluid pressure: observations of the 14-3-3 protein in unselected patients with dementia. Neurology
on cerebrospinal fluid hydrodynamics. J Neurol Sci. 1965;2:307–27. 1997;56:1528 –33.
[59] Vanneste JA. Three decades of normal pressure hydrocephalus: are [72] Castellani RJ, Colucci M, Xie Z, Zou W, Li C, Parchi P, et al.
we wiser now? J Neurol Neurosurg Psychiatry 1994;57:1021–5. Sensitivity of 14-3-3 protein test varies in subtypes of sporadic
[60] Vanneste J, Augustijn P, Dirven C, Tan WF, Goedhart ZD. Shunting Creutzfeldt-Jakob disease. Neurology 2004;63:436 – 42.
normal-pressure hydrocephalus: do the benefits outweigh the risks? [73] Shiga Y, Miyazawa K, Sato S, Fukushima R, Shibuya S, Sato Y, et
Neurology 1992;42:54 – 8. al. Diffusion-weighted MRI abnormalities as an early diagnostic
[61] Josephs KA, Petersen RC, Knopman DS, Boeve BF, Whitwell JL, marker for Creutzfeldt-Jakob disease. Neurology 2004;63:443–9.
Duffy JR, et al. The value of temporary external lumbar CSF drainage [74] Meissner B, Köhler K, Körtner K, Bartl M, Jastrow U, Mollenhauer
in predicting the outcome of shunting on normal pressure hydroceph- B, et al. Sporadic Creutzfeldt-Jakob disease: magnetic resonance
alus. J Neurol Neurosurg Psychiatry 2002;72:503– 6. imaging and clinical findings. Neurology 2004;63:450 – 6.
Alzheimer’s & Dementia 3 (2007) 299 –317

Neuropsychological testing and assessment for dementia


Claudia Jacovaa, Andrew Kerteszb, Mervin Blairb, John Fiskc, Howard H. Feldmana,*
a
Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
b
Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada
c
QE II Health Sciences Centre, Departments of Psychiatry, Medicine and Psychology, Dalhousie University, Halifax, Nova Scotia, Canada

Abstract This evidence-based review examines the utility of brief cognitive tests and neuropsychological
testing (NPT) in the detection and diagnosis of Mild Cognitive Impairment (MCI) and dementia. All
patients presenting with cognitive complaints are recommended to a brief screening test adminis-
tered to document the presence and severity of memory/cognitive deficits. There is fair evidence to
support the use of a range of new screening tests that can detect MCI and mild dementia with higher
sensitivity (ⱖ80%) than the Mini-Mental State Exam (MMSE). NPT should be part of a clinically
integrative approach to the diagnosis and differential diagnosis of dementia. It should be applied
selectively to address specific clinical and diagnostic issues including: 1) The distinction between
normal cognitive functioning in the aged, MCI and early dementia: there is fair evidence that NPT
can document the presence of specific diagnostic criteria and provide additional useful information
on the pattern of memory/cognitive impairment. 2) The evaluation of risk for Alzheimer Disease
(AD) or other types of dementia in persons with MCI: there is fair evidence that NPT measures or
profiles can predict progression to dementia (predictive accuracy ranges from ⬃80 to 100%,
sensitivities from 53 to 80%, and specificities from 67 to 99%). 3) Differential diagnosis: There is
fair evidence that NPT can complement clinical history and neuroimaging in determining the
dementia etiology. Different dementia types have distinguishable NPT profiles though these may be
stage-dependent, and increased sensitivity may be at the expense of specificity. 4) When NPT is part
of a comprehensive assessment, which also entails clinical interviews and consideration of other
clinical data, there is good evidence that it can contribute to management decisions in MCI and
dementia, including the determination of retained and impaired cognitive abilities, their functional
and vocational impact, and opportunities for cognitive rehabilitation.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: ●●●

1. Introduction cognitive domains and that yield scores indexing the func-
tioning of these domains. In the professional practice of
The neuropsychological evaluation of suspected cogni-
neuropsychology, such testing is used to answer a variety of
tive impairment and dementia is conducted at different
clinical questions including, but not limited to, differential
levels of comprehensiveness and professional specializa-
diagnosis. At an advanced level, this testing is an integrative
tion. At a basic level, brief cognitive tests are administered
to screen for the presence of cognitive impairment and to part of a clinical neuropsychological assessment, which also
obtain a global index of cognitive functioning. At an inter- entails clinical interviews and the interpretation of data
mediate level, neuropsychological testing is performed with obtained from other sources to render a diagnostic opinion
batteries of selected standardized tests that probe a range of and recommendations for management [1].
The primary aim of this article is to determine the utility
of brief cognitive screening tests and neuropsychological
*Corresponding author. Tel.: 604-822-7979; Fax: (604) 822-7703. testing in the detection and diagnosis of dementia, whereas
E-mail address: hfeldman@interchange.ubc.ca a secondary aim is to define the role of neuropsychological
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.011
300 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

Table 1
Studies of brief cognitive tests for the detection of MCI
Study Reference Test Time to Sample Setting Findings Evidence Methodologic
Administer Description Level Observations

Nasreddine et al [15], MoCA ⱕ10 min 94 aMCI, 93 AD, Two memory clinics SE/SP, 90%/87% II-2 Samples from different
2005 90 normal (MMSE, 18%/ clinics; reliability
100%) estimate; education
correction
Kalbe et al [16], 2004 DemTect ⱕ10 min 97 aMCI, 121 Memory assessment SE/SP, 80%/92% II-2 Age & education
AD, 145 center (MMSE, 69%/ correction;
normal 77%) transformation
algorithm into
MMSE
Loewenstein et al [17], 3-trial delayed ⱕ5 min with 24 MCI, 26 Community memory SE/SP, 83%/90% II-2 Inclusion limited to
2000 recall of 3 5-min dementia, 52 screening program (MMSE, 71%/ subjects for whom
MMSE intervals normal 85%) diagnostic consensus
words existed between
neuropsychologist
and neurologist
Tang-Wai et al [18], STMS ⱕ10 min 129 aMCI, 788 Community sample & AUC, 0.74 III Retrospective series;
2003 normal, 235 referrals to Mayo (MMSE, 0.70) test scores available
AD Clinic ADRC to clinicians
Eibenstein et al [19], SSST ⱕ10 min 29 aMCI, 29 Alzheimer Evaluation AUC, 0.84; SE/SP, II-2 Test awkward to
2005 normal Unit 84%/72% administer (patient
needs to fast); small
series

Abbreviations: AUC, area under the curve; SE, sensitivity; SP, specificity.

assessment in the management of mild cognitive impair- (evidence level III); and (3) reported on sensitivity and
ment (MCI) and dementia. Brief cognitive tests and neuro- specificity, odds ratios, or relative risks. Where needed,
psychological testing can be appropriately applied across a older studies, studies reporting group differences, and clin-
range of clinical/diagnostic issues. They might be called on ical qualitative studies were also consulted. For studies on
to discriminate MCI from normal aging, to estimate the risk neuropsychological testing a further selection was applied
of progression from MCI to dementia, and to identify the by including only those that evaluated a battery of tests
etiology, severity, and prognosis of dementia. The current covering different domains, as is done in the standard clin-
article aims to provide evidence-based recommendations on ical application of neuropsychological testing, not studies of
the optimal use of brief cognitive screening tests and neu- single neuropsychological tests or domains. Detailed infor-
ropsychological testing. mation on all included empirical studies is presented in
summary Tables 1 to 6. Meta-analyses, review articles, and
position papers were also considered.
2. Methods
This evidence-based review included studies that as- 3. Brief cognitive tests
sessed the discriminative validity of brief cognitive tests and
neuropsychological tests in the normal aging to dementia Brief cognitive tests are designed to address memory/
spectrum. Studies were identified by searching the PubMed cognitive complaints and to objectively identify cognitive
and Embase databases with the following search strings: impairment that warrants diagnostic work-up and treatment.
(1) “Questionable dementia” OR “Cognitive Impairment” The sensitivity and specificity of these tests are not fixed
OR “Cognitive-Impairment-Not-Dementia” OR “Mild Cog- properties but vary with the setting. It is often the case that
nitive Impairment” AND “Diagnosis” AND “Cognitive the sensitivity and specificity of a test are estimated in the
tests” OR “Screening tests” OR “Neuropsychological tests”; specialty clinic setting, with samples of convenience.
(2) “Dementia” OR “Senile dementia” OR “Alzheimer’s These estimates might be inflated, particularly with re-
Disease” AND “Diagnosis” AND “Cognitive tests” OR gard to sensitivity, because cases are preselected and
“Screening tests” OR “Neuropsychological tests”. Prefer- potentially more severe [2]. The test might perform less
ence was given to empirical studies that (1) were published well in other settings including general practice. It has
between 1996 and 2005; (2) were cross-sectional cohort been proposed that population-based studies are better
studies (evidence level II-2), prospective cohort studies able to estimate a test’s sensitivity and specificity in
(evidence level II-2), or retrospective case-control studies general practice than clinic-based studies [3].
Table 2
Studies of brief cognitive tests for the detection of dementia and its subtypes
Study Reference Test Time to Sample Description Setting Findings Evidence Methodologic Observations
Administer Level

Tombaugh et al [6], 3MS ⱕ20 min 119 AD, 406 not Community sample 3MS overall comparable to II-2 Large population-based sample, good
1996 cognitively impaired MMSE; AUC, 0.93 (MMSE, reliability; age- and education-specific
0.91); 3MS possibly better accuracy estimates and norms
for low education
Powlishta et al [28], CDT ⱕ5 min 60 AD, 15 normal Study of healthy aging Very mild dementia (CDR 0.5) III Retrospective; examines CDR ⫽ 0.5, 1, ⱖ2
2002 and AD vs no dementia; SE/SP, separately; estimates for six scoring
20%–60%/60%–93% methods
De Lepeleire 7 tests (brief to ⱕ10 min 50 Dementia, 101 no GP offices AUC, 0.92 for functional index II-2 Assesses diagnostic gain of instruments
et al [29], 2005 NPT computer dementia followed by CDT ranked from least to most burdensome
battery)

C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317


Nasreddine et al [15], MoCA ⱕ10 min 93 AD, 94 aMCI, 90 Two memory clinics AD vs normal: SE/SP, 100%/ II-2 Samples from different clinics; good
2005 normal 87% (MMSE 78%/100%) reliability; education correction
Tang-Wai et al [18], STMS ⱕ10 min 235 AD, 129 aMCI, Community sample & AD vs normal: AUC, 0.96 III Retrospective series; test scores available to
2003 788 normal referrals to Mayo (MMSE, 0.94) AD vs MCI: clinicians; discrimination of AD from
Clinic ADRC AUC, 0.86 (MMSE, 0.85) MCI
Kalbe et al [16], 2004 DemTect ⱕ10 min 121 AD, 97 aMCI, 145 Memory assessment AD vs normal: SE/SP, 100%/ II-2 Age/education correction; transformation
normal center 92% (MMSE, 92%/86%) algorithm into MMSE; discrimination of
AD vs MCI: SE/SP, AD from MCI
85%/81%
Kuslansky et al [33], MIS ⱕ5 min 28 AD, 212 no Community participants AD vs no dementia (includes II-2 Replication study in community sample;
2002 dementia in Einstein Aging CDR 0 & 0.5): SE/SP, strong theoretical foundation for testing
Study 86%/97% paradigm
Solomon et al [31], 7-Minute Screen ⱕ10 min 60 AD, 60 normal Memory clinic SE/SP, 92%/96% II-2 1000⫻ randomly resampled runs; reliability
1998 community estimates; but polarized groups (clinic-
referred AD vs community normal)
Loewenstein et al [17], 3-trial delayed ⱕ5 min with 26 Dementia, 24 MCI, Community memory SE/SP, 92%/94% (MMSE, II-2 Inclusion limited to subjects for whom
2000 recall of MMSE 3 5-min 52 normal screening program 100%/85%) diagnostic consensus existed between
words intervals neuropsychologist & neurologist
Brodaty et al [34], GPCOG ⱕ5 min 82 Dementia, 50 no GP offices Dementia vs no dementia: SE/ II-2 Test designed/validated in GP setting in
2002 dementia but meeting SP, 82%–85%/83%–86% groups with graded severity; requires
some DSM criteria, (MMSE, 81%/76%) informant in ⬃75% of cases; good
20 normal reliability
Kilada et al [35], 2005 Blessed Memory ⱕ5 min with 378 Dementia, 378 BLSA, Washington Dementia vs normal/ III Retrospective study of samples from two
Test & animal brief delay normal University ADRC nondemented: SE/SP, 71%/ large cohort studies; dementia with CDR
fluency 98% (MMSE, 44%/98%) ⬎1 excluded; follow-up of false
negatives
Storey et al [36], 2004 RUDAS ⱕ10 min 45 Dementia, 45 normal Community SE/SP, 89%/98% II-2 Sound test development for cross-cultural
(culturally purpose, good reliability; insensitive to
heterogeneous sample demographic factors
w/ESL subjects)
Darvesh et al [37], BNA Short Form ⱕ20 min 29 Dementia, 29 Behavioural Neurology SE/SP, 93%/93% (MMSE, II-2 Polarized groups; validation of short form
2005 matched normal Clinic (patients), 79%/97%) within long form; good reliability
community (normal)

301
Abbreviations: AUC, area under the curve; BLSA, Baltimore Longitudinal Study of Aging; ESL, English as a second language; SE, sensitivity; SP, specificity.
302
Table 3
Neuropsychological studies of discrimination between normal aging, MCI, and early dementia
Study Reference Sample Description Setting Design Findings Evidence Methodologic Observations
Level

Peters et al [43], 205 CIND, 68 NCI Dementia clinic Cross-sectional Measures of learning, memory, visuoconstruction, II-2 NCI subjects were clinic-referred, hence
2004 (ACCORD) & cognitive flexibility discriminate between not normal controls: discriminative
CIND and NCI with 75%–78% accuracy; inter- ability of NPT might be
test scatter and impairment count indicate underestimated and not generalizable
substantial heterogeneity to population at large
Peters et al [44], 461 CIND (CSHA), Community (CSHA), Cross-sectional Five subgroups characterized by distinct II-2 Use of base (CSHA) and replication
2005 166 CIND dementia clinic neuropsychological impairment patterns (ACCORD) sample for identification
(ACCORD) (ACCORD) of subgroups
Lopez et al [45], 38 MCI, 374 Community (CHS) Cross-sectional MCI multiple cognitive deficits type (74%) more II-2 Neuropsychological criteria for
2006 normal frequent than MCI amnestic type (26%) classification into MCI subtypes not

C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317


independently validated (eg,
clinically, by neuroimaging)
Petersen et al [10], 76 aMCI, 48 mild Mayo AD Center, Cross-sectional aMCI & mild AD impaired on memory measures II-2 aMCI classified clinically,
1999 AD, 234 normal community (⬃1.5 SD below normal subjects); aMCI subtly independently of NPT; comparison of
impaired on non-memory measures (⬃0.5 SD), aMCI is with very mild AD (CDR ⫽
mild AD more substantially impaired (ⱖ1 SD) 0.5 for both groups)
Grundman et al [46], 769 aMCI, 107 ADCS centers Cross-sectional aMCI had prominent memory impairment (⬎1 SD); II-2 Very large and carefully characterized
2004 normal (aMCI from more subtle impairments (⬍1 SD) on language, cohort of aMCI
ADCS-MIS trial) attention, and executive functioning tasks
Kramer et al [47], 22 aMCI, 33 mild UCSF Memory and Cross-sectional ⬎50% of aMCI scored ⱖ1 SD below controls on II-2 aMCI classified according to Petersen
2006 AD, 35 normal Aging Center ⱖ4 non-memory measures including executive criteria plus CDR impairment only on
functioning and fluency tasks memory and orientation domains
Steenhuis et al [53], 450 CIND, 219 Community (CSHA) Cross-sectional Memory, reasoning, and fluency tasks differentiated II-2 Final diagnoses not independent of
1995 dementia, 591 CIND from normal (77% accuracy); memory, NPT, although same pattern found for
normal fluency, and visuospatial tasks differentiated preliminary clinical diagnoses
dementia from CIND (82% accuracy)
Lambon Ralph et al 18 MCI, 38 AD Memory Clinic Longitudinal (2 y) Stage pattern of NP deficits in MCI⬍mild II-2 Cross-sequential analysis of natural
[54], 2003 AD⬍moderate AD: episodic memory history combining cross-sectional and
deficits⬎semantic memory deficits⬎language longitudinal data
deficits; longitudinal change in MCI and AD
identical, albeit starting at different points
Rubin et al [57], 82 cognitively Community Longitudinal (12 y) Change in general cognitive factor regression-fitted; II-2 Serial assessments; only those before
1998 healthy CDR ⫽ intercept but not slope significantly different CDR change were included in
0, 27 w/CDR ⬎0 between stable group and group that developed regression; visual representation of
at f/u CDR ⬎0 cognitive/CDR change
Amieva et al [58], 1,265 nondemented Population-based Longitudinal (9 y) Lower initial scores and accelerated decline ⬃3 y II-2 Serial assessments entered into linear
2005 subjects, 215 AD study (PAQUID) before diagnosis in episodic memory, abstract mixed model with modification to
at f/u reasoning & semantic fluency; decline was allow for non-linear effects; separate
steeper for highly educated subjects in analyses for low and high education
acceleration phase
Chen et al [59], 551 nondemented Community Longitudinal (3.5 y) Rate ratios (cases/controls who declined ⱖ1 SD on II-2 No comparison between predictive
2001 subjects, 68 AD test): executive function, 3.7; verbal learning & utility of baseline and change scores
at f/u recall, 2.6–3.6; praxis, 2.5; naming, 2.1

Abbreviations: f/u, follow-up; NCI, not cognitively impaired.


Table 4
Neuropsychological studies of prediction of dementia or AD
Study Reference Sample Description Setting Follow-up Follow-up Findings Evidence Methodologic Observations
Duration (y) Diagnosis Level

Artero & Ritchie [66], 308 Subjects w/cognitive GP referred to study 2 AD Delayed story recall, semantic II-2 Heterogeneous (MCI, normal) and
2003 complaints, 19 AD at f/u (Eugeria Study) fluency, copying abstract figure: insufficiently characterized
SE/SP, 73%/99% cohort at baseline
Chen P et al [59], 603 nondemented subjects, Population-based study 1.5 AD Delayed recall ⫹ Trails B: AUC, 0.83 II-2 Heterogeneous (MCI, normal) and
2001 120 AD at f/u (MoVIES) insufficiently characterized
cohort at baseline; test scores
available for diagnosis
Blackwell et al [69], 43 Subjects with questionable Memory clinic 2.5 AD Visuospatial associative learning ⫹ II-2 Battery covers only episodic and
2003 dementia, 11 AD at f/u confrontation naming: 100% semantic memory
accuracy
Delayed verbal recall ⫹ semantic

C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317


Tierney et al [63], 551 nondemented subjects, 77 Population-based study 5 or 10 AD II-2 Heterogeneous cohort composed
2005 AD at f/u [5-y study]; 263 (CHSA) fluency ⫹ information (5-y f/u): of NCI and CIND; predictive
nondemented subjects, 47 SE/SP, 74%/83% Delayed verbal parameters validated by
AD at f/u [10-y study] recall (10-y f/u): SE/SP, 73%/70% bootstrapping
Tierney et al [64], 123 subjects with memory GP referred to study 2 AD Delayed verbal recall ⫹ mental II-2 Model validated on subsample
1996 impairment, 29 AD at f/u control: SE/SP, 76%/94% w/matched MMSE; research
NPT battery different from
clinical NPT battery
Elias et al [67], 2000 1043 nondemented subjects Population-based study ⱖ5 or ⱖ10 AD (y of AD after ⱖ5 y: logical memory II-2 Very long surveillance period
with 106 AD at f/u (Framingham Study) diagnosis) retention. OR 1.53; similarities, OR (22 y); Cox regression for
1.32; paired associate learning, OR prediction of time to diagnosis;
1.29 AD after ⱖ10 y: logical NPT battery used for later
memory retention, OR 1.17; diagnosis
imilarities, OR 1.35
Howieson et al [68], 47 normal subjects, 16 AD at Population-based study ⱖ2 AD/questionable Logical memory ⫹ naming: accuracy, II-2 Cohort of very old subjects (ⱖ80
1997 f/u (Oregon Brain dementia 83% y of age); diagnosis CDR-based
Aging Study) and inclusive of questionable
dementia (CDR 0.5)
Palmer et al [70], 121 subjects with memory Population-based study 3 Any dementia Episodic memory impairment: SE/SP, II-2 Investigates the added value of
2003 complaints, 38 dementia at (Kungsholmen) 50%/89%; verbal fluency NPT tests over MMSE score
f/u impairment: SE/SP, 48%/92%; but very limited NPT battery
visuospatial impairment: SE/SP,
26%/46%
Bowen et al [62], 21 subjects with isolated HMO registry 4 Any dementia No NPT test predictive of diagnosis II-2 Very small sample; NPT results
1997 memory loss, 10 dementia available for clinical diagnosis
at f/u
Devanand et al [71], 62 subjects w/questionable Memory Disorders ⱕ7 Dementia (87% SRT long-term retrieval, digit symbol, II-2 NPT results available for clinical
1997 dementia, 31 dementia at Clinic AD) picture arrangement, block design, diagnosis; high rate of
f/u semantic fluency: SE/SP, 67/67% reversion (41%)
for demented; SE/SP, 61%/78%
for AD
Fabrigoule et al [76], 1159 nondemented subjects, Population-based study 2 y AD and General cognitive factor reflecting II-2 Principal component analysis of
1998 16 incipient AD, 9 other (PAQUID) dementia executive control processes: OR ⫽ NPT scores to address
dementia 2.3 for dementia, OR ⫽ 3.3 for AD colinearity problem; very low
number of incident cases

303
304
Table 4
Continued
Study Reference Sample Description Setting Follow-up Follow-up Findings Evidence Methodologic Observations
Duration (y) Diagnosis Level

Arnaiz et al [65], 303 subjects with MCI Mayo Clinic Max 5–10 y AD Number of impaired domains (⫺1.5 II-2 Cross-national study; outcome is
2004 (number of incident AD SD): SE/SP, 80%/75%; delayed time to diagnosis with Cox

C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317


not stated) recall best predictor in both regression; model validated in
samples each sample separately
Tian et al [75], 2003 129 subjects w/questionable Memory Disorders 2 Any dementia Predictive models unstable; better w/ II-2 Clinically well characterized
dementia, 37 dementia Clinic categorical than continuous NP cohort; Cox regression for
at f/u variables; total immediate recall prediction with different
most consistent predictor SE/SP, variants to establish stability
⬃53%–64%/67%–83%
Bozoki et al [72], 48 subjects with isolated Michigan ADRC 2 AD Subjects classified into M⫺ (memory III Retrospective database review;
2001 memory impairment, 16 database impairment only) and M⫹ repeated NP test results
AD at f/u (additional non-memory available to treating clinician
impairment), relative risk 4.6 for
M⫹; no single test predictive
Peters et al [44], 2005 CIND subjects Community (CSHA), 2–5 y Any dementia Membership in episodic memory II-2 Use of two independent samples
dementia clinic dysfunction subgroup: OR 2.27 to for cluster solution and
(ACCORD) 3.48 prediction
Ritchie et al [77], 283 subjects with recent GP research network 3 Any dementia Membership in general cognitive II-2 Preliminary report with few cases
1996 cognitive decline, 19 (Eugeria) impairment with language and non-completion of
dementia at f/u dysfunction (OR 4.4) or memory surveillance period; no
impairment (OR 3.9) cluster independent sample for cluster
validation
Herlitz et al [74], 285 nondemented subjects, 70 Population-based study 3 Any dementia & Episodic memory measures & Trails- II-2 Analysis of discordant diagnostic
1997 dementia at f/u (Kungsholmen) AD based dementia diagnoses w/out classifications based on NPT
DSM confirmation had 65% higher and DSM
DSM dementia at f/u than test-
based normal diagnoses

Abbreviations: AUC, area under the curve; f/u, follow-up; NCI, not cognitively impaired; OR, odds ratio; SE, sensitivity; SP, specificity; SRT, Selective Reminding Test.
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 305

Table 5
Neuropsychological differentiation of AD from depressive dementia, DLB, and PDD
Comparisons Study Reference & Sample Description Findings Conclusion (Evidence Level in Parentheses)
Design

AD vs depressive Christensen et al [153], Reviewed 154 articles Depressives ⬎ AD: paired associate Depression has widespread effects on
dementia 1997, meta-analysis with 225 learning task, anomalous sentence cognitive functioning including
comparisons repetition test, Boston naming & intelligence, problem solving, & memory;
WAIS block design; tests depressives do better on untimed tasks
requiring little cognitive capacity (II); clinical examination & depression
or effort (eg, nonspeeded tests inventories are basic for diagnosis (III)
superior to speeded tests); no
convincing difference between
the groups on recall vs
recognition task
AD vs DLB, Collerton et al [93], Reviewed 21 articles: AD ⬎ DLB on visuoperceptual/ Composite measures (eg, MMSE) might
PDD 2003, meta-analysis 312 DLB vs 380 semantic tasks regardless of underestimate severity of DLB because
on DLB AD, 48 DLB vs disease stage or diagnostic of inadequate testing of attentional,
65 PD criteria, attentional/executive/ executive, or visuoperceptual
timed motor responses performance; memory deficits of DLB
Simard et al [154], Reviewed 68 studies DLB ⬍ AD on spatial working are less severe than their visuoperceptual/
2000, review of on cognitive & memory (eg, Trails B), attentional/executive deficits (II); similar
DLB behavioral changes visuospatial function (eg, more cognitive profiles in DLB & PDD
in DLB impaired on clock copy vs free probably reflect superimposition of
drawing) subcortical deficits on those typically
Aarsland et al [83], Mild to severe Mild-moderate DLB ⬍ PDD associated with AD
2003, cross-sectional dementia patients: conceptualization subscore;
60 DLB, 35 PDD, severe DLB ⫽ PDD on all
49 PSP, 29 AD; subscores; PDD, DLB, & PSP ⬎
comparison on AD on memory subscores; AD ⬎
Mattis dementia PDD & DLB on
rating scale initiation/perseveration &
construction subscores; AD ⬎
DLB on conceptualization
subscore; PSP ⬎ PDD & DLB
on memory subscores

Abbreviations: PSP, progressive supranuclear palsy; WAIS, Weschler Adult Intelligence Scale.

By design, brief tests have a limited measurement range, ables, primarily age and education [5,6]. Typically, perfor-
and both ceiling and floor effects can threaten their validity. mance is lower in older age and lower education groups.
Ceiling effects are a particularly important threat when mild When single cutoff scores are used across different age
symptoms are being evaluated. For example, the widely and education levels, the evident risk is an increased rate
used 30-item Mini-Mental State Examination (MMSE) [4] of false-positive classifications among older subjects and
has lacked sensitivity to mild degrees of impairment [5]. those with lower education. However, test users should
With the conventional cutoff level at ⬍24 for dementia, the be mindful that a “correction” for age and education
test has only a 6-point scale for discrimination between MCI effects, through application of different cutoff scores or
or very mild dementia and normal functioning, with score adjustment, also carries risks. Both old age and low
overlap and skew as a result of the test’s ceiling. The education have consistently been associated with an in-
MMSE has become a benchmark against which newer tests creased risk for cognitive impairment and dementia [7].
are compared to establish their improved sensitivity to mild The application of adjusted cutoff scores can reduce the
degrees of impairment. sensitivity of tests, with reduced identification of true
When using brief cognitive tests, it is important to realize positives in these at-risk groups [8].
that no single test has utility along the entire spectrum of
severity. Different tests will need to be chosen depending on 3.1. Brief cognitive tests for the detection of MCI
whether MCI or dementia is suspected. Some brief cogni-
tive tests have been specifically developed to detect Alzhei- MCI is a diagnostic label that has been variously defined
mer’s disease (AD) and might not be able to detect other but that generally refers to individuals with memory and/or
dementia subtypes because of their emphasis on memory cognitive impairment not sufficiently severe to fulfill de-
abilities. It is also critical to understand that performance on mentia criteria. In the current article, we apply the term MCI
most brief cognitive tests is affected by demographic vari- in this general sense, thus covering many proposed specific
306 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

Table 6
Neuropsychological differentiation of AD from FTD and VaD
Comparisons Study Reference & Sample Description Findings Recommendation
Design

AD vs FTD Kramer et al [155], 2003, 21 FTD, 14 SD, 30 AD AD and SD ⬍ FTD on Tests of verbal memory, language,
AD vs PPA case control 52 FTD-bv, 52 AD verbal memory and and executive function
AD vs SD Kertesz et al [106], 2003, 26 FTD naming; FTD worse on discriminate AD from FTD, but a
case control executive function behavioral inventory improves the
Mathunarath et al [156], FTD ⬎ AD on memory, but distinction - Level II evidence;
2000, case control lower language scores; A language/memory ratio (ACE) and
FBI best discriminates the the FBI can be adapted to a
groups clinical community service
[verbal fluency ⫹ language]/ - Level III and II evidence
[orientation ⫹ memory] Formal language testing is helpful,
ratio best discriminates but clinical experience is
the groups necessary for interpretation.
Kertesz et al [106], 2003, 67 PPA, 99 AD Anomia at start; change in Detailed language and semantic
case control 11 NFPA, 10 SD, 11 fluency is stage testing may be necessary for
Gorno-Tempini, 2004 logopenic progressive dependent; logopenia, accurate distinctions
[157], case control aphasia aphemia, Brocha type
Hodges et al [158], 1999, 9 FTD, 9 AD, 9 SD speech in PPA; Wernicke
case control type speech in AD
Apraxia and agrammatism in
NFPA; fluent semantic
memory deficit in SD;
anomia in LP
Naming and semantic
knowledge in SD
AD vs VaD Looi et al [87], 1999, Meta-analysis of 27 Better verbal memory and AD is different from VaD on
VAD vs FTD meta-analysis articles worse executive function neuropsychological testing, but the
AD vs Huntington, Sjogren et al [159], 1996, 11 VAD, 21 FTD in VaD differences are stage-related and
PSP, CBD case control Behavior worse in FTD subtle. Mixed dementia resembles
whereas memory is more AD. Neuropsychological testing is
impaired in VAD, but supportive, to be used in specialist
there is no overlap setting only
VaD and FTD overlap, but memory
and language tests and behavioral
quantification helps - Level II
Butters et al [160], 1985, Many case control studies Subcortical dementia: frontal
evidence
case control and reviews on deficit; recognition
The concept of subcortical dementia
subcortical dementia memory better than recall;
is useful and can be supported by
CBD has apraxia and
neuropsychology - Level II
aphasia
evidence. Most of these conditions
are diagnosed by the movement
disorder

Abbreviations: ACE, Addenbrooke’s Cognitive Examination; CBD, corticobasal degeneration; FBI, frontal behavior inventory; NFPA, progressive
nonfluent aphasia; PPA, primary progressive aphasia; SD, semantic dementia.

definitions [9]. We further distinguish the entities of amnes- appears similar in population samples [12]. This MCI pro-
tic MCI (aMCI) and Cognitive Impairment Not Dementia gression rate compares with 1% to 2% among matched
(CIND). The term aMCI was developed to capture the healthy elderly [9]. An American Academy of Neurology
preclinical stage of AD, with emphasis on memory loss Practice Parameter has recommended the identification and
within the context of otherwise normal cognitive function- further clinical evaluation of MCI [14].
ing [10]. The term CIND was developed for population Recently a number of new instruments have been
epidemiologic studies to denote any state of cognitive func- developed for the detection of MCI: the Montreal Cog-
tioning that falls outside of normal range yet does not meet nitive Assessment (MoCA) [15], the DemTect [16], a
dementia criteria [11]. Depending on the definition that is three-trial delayed recall adaptation of the MMSE [17],
used, the prevalence of MCI ranges from as low as 1% [12] the Short Test of Mental Status (STMS) [18], and the
to ⬃40% in the population and 30% in clinic-referred sam- Sniffin’ Sticks Screening Test (SSST) [19]. Table 1 sum-
ples [13]. The rate of progression from MCI to dementia is marizes studies of these five tests, with clinically diag-
10% to 15% per year in clinic-referred samples [9] and nosed MCI as the standard.
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 307

The MoCA is a 30-point test that covers multiple cog- miliarity in Canada. Tombaugh et al [6] have compared the
nitive domains. The MoCA has been found to have good 3MS and MMSE in CSHA participants who were diagnosed
internal consistency and test-retest reliability and was able with AD or cognitively intact. The 3MS did not have sig-
to correctly identify 90% of a large sample of aMCI subjects nificantly improved sensitivity over the MMSE, but its
from two different clinics [15]. The DemTect, scored on an accuracy tended to be about 5% higher in subjects with low
18-point scale, has similar coverage of domains. Although education (0 to 8 years).
its reliability remains to be determined, its discriminative The Clock Drawing Test (CDT) has achieved wide-
validity was superior to the MMSE in a large clinic sample spread clinical utilization and has been studied extensively
of aMCI patients, compared with normal controls. DemTect [24]. A Canadian survey found that primary practitioners
scores can be transformed into MMSE by means of a simple considered the CDT an acceptable cognitive screening test
algorithm [16]. Three delayed recall trials of the three [25]. A review of validation studies identified consistently
MMSE items at 5-minute intervals adding to total of 9 good levels of sensitivity and specificity for the CDT in the
points compared favorably with the total MMSE score in a early detection of dementia, irrespective of the scoring sys-
community sample of subjects classified as MCI and those tems that were used [26]. Recent evidence, however, invites
deemed cognitively normal [17]. There was a low diagnos- caution because sensitivity was found to be very low for
tic agreement rate (57%), and cases in which no consensus most scoring systems when dementia was very mild, at a
diagnosis could be reached (29%) were excluded, suggest- Clinical Dementia Rating (CDR) global stage [27] of 0.5
ing problems with the reliability of the gold standard in this [28]. CDT scores added significantly to an overall 92%
study. Evaluation of this repeated recall paradigm is there- accuracy in a hierarchical classification algorithm that was
fore not possible until further studies are carried out. The developed for use in general practice and that used age, sex,
38-point STMS is an expansion of the MMSE in the do- a functional status measure, and the CDT in a stepwise
mains of learning/recall and abstract reasoning. This mea- fashion [29]. Earlier studies have also suggested that the
sure did not perform better than the MMSE in separating CDT might perform better in combination with other instru-
aMCI from normal, both achieving an area under the curve ments, for example, the MMSE [30].
of ⬃0.70 [18]. New instruments for the detection of MCI have also been
Because AD neuropathology affects the limbic system investigated either with AD or with dementia in general as
that is involved in olfactory function, deficits in olfaction the standard. Whereas the MoCA and DemTect had perfect
have been investigated as a diagnostic marker of preclinical sensitivity for AD, performing better than the MMSE
AD [20,21] and aMCI. The SSST was developed as a [15,16], the STMS was comparable to the MMSE [18]. The
bedside odor identification test for early detection of AD. 7-Minute Screen (7MS) is a multiple domain test with very
The SSST separated aMCI patients from normal controls high sensitivity and specificity for AD, which was calculated in
with good accuracy (area under the curve, 0.84), but 1000 classification iterations with random subsamples [31].
cognitive tests tend to be more accurate, with both higher These measurement properties were subsequently con-
sensitivity and specificity. In addition, the test is awk- firmed in a primary care setting [32]. The Memory Impair-
ward to administer, with the subject having to fast before ment Screen (MIS) is a theoretically grounded attempt at
the test [19]. disentangling deficits caused by AD from those caused by
normal aging. This 8-point scale had high sensitivity and
3.2. Brief cognitive tests for the detection of dementia and specificity (⬎85%) for AD in a community sample includ-
its subtypes ing nondemented controls, some with a CDR of 0.5 [33].
The three-trial delayed recall adaptation of the MMSE per-
Table 2 summarizes 13 studies that have examined brief formed similarly well in the discrimination of dementia of
cognitive tests for the detection of AD or dementia in any cause (sensitivity and specificity, ⬎90%), but caution is
general. In the development of these instruments the aim has required regarding the gold standard in this study as a result
been to increase sensitivity levels to early or mild dementia of the aforementioned problems with diagnostic agreement
above those achieved with the MMSE. The Modified Mini- and subject exclusion [17].
Mental State Examination (3MS) [22] was developed as a The General Practitioner Assessment of Cognition
test with increased sensitivity to dementia by adding tasks (GPCOG) is a 15-point test specifically developed for
known to be impaired early in the disease, including delayed primary care. A patient section and potentially an infor-
recall and a verbal fluency test for the semantic category of mant section are administered in a two-step procedure.
animals. In the 3MS both ceiling and floor of the MMSE are This instrument discriminated those who met Diagnostic
extended, and the range of possible scores is increased from and Statistical Manual of Mental Disorders, Fourth Edi-
30 to 100. The 3MS is quite widely used in clinic and tion (DSM-IV) criteria for dementia from those who did
research settings. It served as a screening test for dementia not with good levels of sensitivity and specificity
and referral for clinical assessment in the Canadian Study of (⬎85%). Nearly 40% of false positives in this study were
Health and Aging (CSHA) [23], allowing it particular fa- subjects who met some but not all DSM-IV criteria for
308 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

dementia [34]. A brief paradigm consisting of the Blessed well with heterogeneous dementia samples that include
Memory test plus animal fluency has also been found to non-AD subtypes. In this latter context, tests sampling mul-
have better sensitivity than the MMSE at equal specificity in tiple domains are likely to perform better than those
retrospective studies of two large cohorts, with exclusion of weighted toward or sampling only episodic memory (eg,
cases with CDR ⬎1 [35]. MIS, three-trial delayed recall). At present, no brief cogni-
The Rowland Universal Dementia Assessment Scale tive test has been developed to differentiate between sub-
(RUDAS) was designed to detect dementia in cross-cultural types of dementia, and because of the complexities of these
settings. The RUDAS items were developed with endorse- distinctions, it seems unlikely that one will emerge [38].
ment of cross-cultural and cross-discipline advisory panels.
The test performed well (sensitivity and specificity, 89% 4. Neuropsychological testing
and 98%, respectively) in a culturally heterogeneous
community sample in Australia, with more than 50% There is no unitary view on the role of neuropsycholog-
having English as a second language and/or with educa- ical testing (NPT) in the diagnostic work-up of dementia.
tion ⬍6 years [36]. The potential of detection bias caused NPT is typically performed after administration of a brief
by Western culture idiosyncrasies has been recognized cognitive test and other clinical work-up to probe the func-
for some MMSE items, and the RUDAS might offer a tioning of memory and other cognitive domains with stan-
valid alternative in the assessment of subjects with non- dardized tasks. A systematic study of the usefulness of the
Western background. 1994 American Academy of Neurology practice parameters
The Behavioural Neurology Assessment (BNA) Short for the evaluation of dementia found evidence that NPT has
Form is a 114-point scale that assesses multiple domains but utility in the assessment of MCI and in cases in which
requires greater administration time than most other brief memory impairment is not a predominant feature [39]. A
cognitive tests. The BNA has been found more sensitive to background article to the Second Canadian Consensus Con-
dementia than the MMSE [37] and raises the question of ference on Dementia concluded that although NPT might
whether longer assessments generally have added utility that have utility in borderline cases, it is not required in the
justifies their extra time expenditure. This specific question routine diagnostic work-up of suspected dementia [40]. The
was addressed in a comparative study of three brief tests National Institute of Neurological and Communicative Dis-
(MMSE, Orientation-Memory-Concentration [OMC] test, orders and Stroke—Alzheimer Disease and Related Dis-
Mental Status Questionnaire [MSQ]) and two abbreviated orders (NINCDS-ADRDA) criteria require confirmatory
test batteries (Dementia Rating Scale [DRS], Ottawa Mental evidence of impairment on neuropsychological tests for the
Status Examination [OMSE]). The longer tests did not offer diagnosis of probable AD [41]. At a practical level, NPT
advantages over the brief tests in terms of their reliability involves often long administration times and is frequently a
and sensitivity to dementia of any type. Of note, no test or limited resource, not widely accessible to the clinician.
mini-battery was able to reliably differentiate AD from In this article we propose on the basis of current evidence
vascular dementia (VaD) [38]. that the diagnosis and differential diagnosis of dementia
The discrimination between MCI and dementia is critical must be considered a clinically integrative process. NPT
for monitoring and treatment decisions, yet this issue is alone should not be used for these purposes. Instead, NPT
rarely addressed in the evaluation of brief cognitive tests. should be applied selectively to aid in (1) addressing the
Only the DemTect and the STMS have been examined for distinction between normal cognitive functioning in the
this purpose. Although both were fairly accurate in separat- aged, MCI, and early dementia; (2) addressing the risk of
ing AD from aMCI, with ⬃85% accuracy and sensitivity/ progression to dementia in persons with MCI diagnoses;
specificity [16,18], the ability of all other instruments to and (3) the differential diagnosis of dementia and other
make similar distinctions remains to be determined. On the syndromes of cognitive impairment. We evaluate the
basis of the available evidence it is not possible to determine evidence in support of each of these applications in the
whether this discrimination can in fact be reliably achieved following paragraphs. In our evaluation of studies that
by brief cognitive tests, or whether it is an issue that might support the utility of NPT in the differential diagnosis of
have to be deferred to a neuropsychological evaluation. dementia we have chosen to take a clinically based qual-
In summary, most brief cognitive tests can detect mild itative approach aimed at delineating differential NPT
dementia accurately, with the possible exception of the profiles for the dementias, rather than examining general
CDT. Where newer tests have been directly compared with accuracy parameters.
the MMSE, the MoCA, the DemTect, the BNA, and the
4.1. The distinction between normal aging, MCI, and
GPCOG but not the 3MS appear to provide better accuracy
early dementia
as a result of their higher sensitivity. Among these the
GPCOG is the only instrument that has been validated In the cognitive spectrum from normal aging to dementia,
directly in primary care settings. Tests developed and vali- NPT might be useful in the differentiation of the bordering
dated solely with AD samples might not perform equally states of MCI/normal functioning and early dementia/MCI.
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 309

In each case, the dividing line is in part definable by criteria 4.1.2. Distinguishing mild dementia from MCI
based on neuropsychological test performance. MCI defini- NPT can discriminate quite accurately (sensitivity,
tions might require objective impairment in memory [33] or ⱖ80%, specificity, ⱖ90%) between patients with AD or
in any of the cognitive domains of learning, memory, atten- dementia, even at a very mild stage, and cognitively normal
tion, thinking, language, or visuospatial function [42] for elderly [49 –51]. The utility of NPT in the discrimination
diagnosis. The NINCDS-ADRDA clinical criteria for prob- between MCI and mild dementia has been less systemati-
able AD require abnormal performance in two or more areas cally investigated, in part perhaps because the focus in this
of cognition including orientation, memory, language, discrimination is on the requirement that cognitive deficits
praxis, attention, visual perception, and problem solving have an adverse impact on social and occupational func-
[41]. There is in consequence a degree of interdependence tioning, which defines the threshold for dementia according
between the clinical differentiation of MCI or dementia and to DSM criteria [52]. MCI groups generally score at levels
NPT. Beyond its role in the fulfillment of specific diagnostic intermediate between those of normal controls and those of
criteria, however, NPT can provide additional information patients with mild AD or dementia [10,47,53]. Episodic
that might be useful in the discrimination of MCI and early memory impairment in aMCI has been reported to be sim-
dementia. The reviewed studies are summarized in Table 3. ilar in severity to that found in mild AD, whereas perfor-
mance in other domains, particularly language, is closer to
4.1.1. Distinguishing MCI that of controls. This gradient of severity, with non-memory
In broadly defined MCI groups there is considerable measures best differentiating between aMCI and AD, is
heterogeneity with respect to neuropsychological test per- evident also in cross-sequential analyses of neuropsycho-
formance. In the Canadian Cohort Study of Cognitive Im- logical test performance in groups of aMCI and mild and
moderate AD patients. These analyses identified a staged
pairment and Related Dementias (ACCORD), subjects clas-
pattern of neuropsychological deficits, starting in the epi-
sified as CIND differed from those deemed not cognitively
sodic memory domain, spreading to the semantic memory
impaired on a range of memory and non-memory measures,
and language domains with AD onset, and subsequently, as
with measures of learning and recall, visuoconstruction, and
patients progress to moderate AD, to the perception and
cognitive flexibility providing the best discrimination be-
attention domain [54].
tween the groups [43]. However, performance patterns in
Evidence of progressive memory and cognitive loss is a
CIND were not uniform within and between individuals,
requirement for AD diagnosis in the NINCDS-ADRDA
and cluster analysis identified five distinct subgroups, with
criteria [41], and there is agreement among a number of
only two having memory impairment [44]. In the Cardio-
studies that rates of decline on serial NPT accelerate in the
vascular Health Study (CHS) subjects were generally clas- 3- to 5-year time window before AD diagnosis, with abrupt
sified as MCI if they had impairment in any domain (mem- decline shortly before a diagnosis can be rendered [55]. The
ory and/or non-memory). Within this broad grouping, 74% change point in preclinical AD has been defined as the time
of subjects met the criteria for MCI-multiple cognitive def- before diagnosis, where on average the rate of decline
icits type and had neuropsychological impairments in at among those who will develop AD and those who will not,
least two domains, whereas the remainder met criteria for will begin to diverge. On the basis of longitudinal data on
aMCI, with memory impairment only and no other domains episodic memory test performance, this point was estimated
impaired [45]. to occur 5 years before diagnosis [56]. A 12-year study
Studies comparing groups of aMCI patients with normal found that subjects whose CDR increased above 0 during
controls on comprehensive NPT batteries have consistently the surveillance period did not have steeper cognitive de-
found that in addition to the expected episodic memory im- cline before the CDR increase when compared with those
pairment on learning and delayed recall measures, even in this whose CDR remained 0. Abrupt and general deterioration of
narrowly defined MCI group subtle difficulties are manifest on performance was observed only at the time of CDR change
tests of language (naming and fluency), attention, and execu- [57]. A 9-year study reported parallel cognitive change for
tive functioning [10,46,47]. Whereas differences between those who later developed AD at follow-up and those who
aMCI and controls were generally ⬍1 standard deviation (SD) did not develop dementia until 3 years before diagnosis
on non-memory measures [10,46], one study reported that when cognitive decline accelerated in the incident AD sub-
more than half of their aMCI subjects scored at least 1 SD jects, particularly in those subjects with high education [58].
below control means on ⱖ50% of tests [47]. The ratio of incipient AD cases to controls who declined by
There is no generally accepted NPT battery for MCI. ⬎1 SD during a 2-year period was 4 AD subjects for every
However, the evidence underscores that regardless of the control subject on executive function tests, 2.5 to 3.5 on
specific MCI criteria, NPT should extensively cover a range episodic memory tests, and 2 on language tests [59]. Al-
of cognitive domains. In addition, serial testing has been though none of these longitudinal studies have specifically
recommended to establish the consistency over time of addressed the prognostic/diagnostic utility of rates of de-
memory and/or other cognitive impairments [48]. cline in MCI, evidence of significant loss on serial NPT in
310 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

these subjects, particularly in the memory, language, and Studies involving longer times to diagnosis typically
executive functioning domains, should be considered sup- identify fewer neuropsychological predictors than those in-
portive of development of AD. volving shorter times [63,67]. In the CSHA, the only pre-
There is some evidence to suggest that the detection of dictor of AD emerging from the 10-year study was delayed
preclinical change during short time periods (1 to 2 years) is verbal recall, whereas the 5-year study revealed semantic
better when computer-administered tasks are used rather memory measures as additional predictors [63]. These find-
than standard examiner-administered NPTs. For example, ings suggest that the earliest warning sign of dementia or
1-year decline in aMCI subjects was detected by a comput- AD might be an episodic memory deficit, and that as the
erized continuous learning task but not by the standard preclinical stage evolves toward diagnosable dementia, def-
neuropsychological Consortium to Establish a Registry for icits in other domains also begin to have prognostic signif-
Alzheimer’s Disease battery [60]. Similarly the Cambridge icance. A study with 2-year follow-up of memory-impaired
Neuropsychological Test Automated Battery paired associ- subjects in fact identified an almost five-fold risk of pro-
ate learning task but not standard neuropsychological mea- gression to AD for those who had impairments in non-
sures including the revised Wechsler Adult Intelligence memory domains [72].
Scale and Wechsler Memory Scale–revised subtests, verbal A meta-analysis of 47 studies calculated pooled effect
and category fluency, Rey Complex Figure, Austin Maze, sizes as well as percent score overlap (O/L) between the
and Rey Auditory Verbal Learning test detected a linear incident AD and control groups. These were Cohen’s d ⫽
2-year decline in subjects with questionable dementia who 1.0, O/L 42% for episodic memory; d ⫽ 0.8, O/L 57% for
were retrospectively classified as deteriorating [61]. How- verbal abilities; d ⫽ 0.6, O/L 62% for visuospatial abilities;
ever, because neither study provided diagnostic validation d ⫽ 0.6, O/L 62% for attention; and d ⫽ 1.1, O/L 42% for
that the declining subjects had dementia or AD at the end of executive functioning [73]. The overlap statistic suggests
the observation period, these findings must be considered that prediction, although useful, is not likely to be perfect or
preliminary. even highly accurate in most cases. Because effect sizes
were larger for shorter (⬍3 years) than longer follow-up, it
4.2. The prediction of progression to dementia can be assumed that increasing predictive accuracy is
achieved as time to diagnosis approaches.
The neuropsychological features that predict progression This meta-analytic study identifies episodic memory and
to dementia or AD in cohorts of nondemented elderly or executive functioning deficits as the best predictors of de-
patients with MCI have been investigated extensively. The mentia. Corroboration of this point can also be found from
typical research design has been to follow population-based a follow-up study of discordant neuropsychological diag-
or clinic-referred cohorts for a number of years and to noses of dementia in the population-based Kungsholmen
compare the baseline neuropsychological performance of Project. Subjects classified as demented on the basis of NPT
those who progress to dementia or AD at some point in the but not diagnosed as demented clinically had an increased
surveillance period with the performance of those who re- likelihood of receiving a dementia diagnosis at the next
main nondemented. The present review has identified 17 evaluation, particularly if the initial cognitive classification
studies, summarized in Table 3. The majority are popula- was based on episodic memory and executive functioning
tion-based studies, and the length of follow-up ranges from deficits [74].
2 to 22 years. It would currently be premature to extrapolate from the
The majority of studies have found that initial neuropsy- existing evidence that specific neuropsychological tests can
chological scores predict progression to dementia and AD, be used for prognostic purposes. A problem that is often
with one exception, likely as a result of the very small overlooked in predictive studies that analyze multiple neu-
sample size of the study [62]. Predictive accuracy has ropsychological scores is colinearity, that is, the tendency
ranged from ⬃80% to 100%, sensitivities from 53% to for test scores to vary systematically with each other as a
80%, and specificities from 67% to 99%. Episodic memory function of disease severity. This can give rise to overin-
impairment is the most consistently identified predictor, clusion of interrelated scores and to predictor sets that are
whether measured by delayed word recall [59,63– 65), de- too large to be practically useful [71]. There might also be
layed story recall [66 – 68], or associative learning [67,69]. significant instability of the predictive models. Tian et al
This is true for samples of clinic-referred subjects with [75] used the same set of neuropsychological predictor
MCI, questionable dementia, or memory complaints but variables in three variants of Cox regressions (Enter, For-
also for population-based samples of elderly who were wards Stepwise, and Backwards Stepwise), with time to
cognitively normal at study entry. Many studies have also diagnosis of dementia as the dependent variable. Each
identified predictive measures in other domains, again re- model was found to retain different variables, although each
gardless of setting and sample: semantic memory/language consistently included a measure of episodic memory. A
[63,66,68 –70], attention and executive functioning [59,64], strategy that is capable of explicitly addressing colinearity,
abstract reasoning [67], and visuospatial abilities [71]. although not very user friendly, is principal component
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 311

analysis, which extracts the latent cognitive factor(s) that the progression of the illness, remote memory becomes
might be measured by multiple tests. Most neuropsycholog- involved, along with semantic memory. Decreased sensitiv-
ical test scores in the Personnes Agées Quid (PAQUID) ity to the semantic properties of information in AD inter-
study loaded highly on the first factor, interpreted as reflect- feres with encoding of new material [88 –90]. Orientation
ing general executive control processes. When other factors and episodic memory are relatively preserved in FTD. The
were generated at eigenvalues ⬎0.75, the first factor re- complaint of forgetfulness on history and variable perfor-
mained the only reliable predictor of progression to AD mance on memory testing are usually related to inattention.
among a very large cohort of elderly subjects. Low scores FTD-bv patients might have impaired test results on imme-
on this factor were associated with a two- to three-fold risk diate and delayed recall of a story, yet they are able to recall
of dementia or AD [76]. personally relevant events, which is out of keeping with
An entirely different approach to prediction of dementia their cognitive test results [91,92]. Poor recall in PNFA and
has focused on the distinction of profiles or subgroups. This semantic dementia is likely a result of the aphasic distur-
subgroup/profile approach has the advantage of using all the bance in these conditions. Recognition memory is typically
available neuropsychological information, capitalizing on better than recall in VaD and dementia arising from sub-
the relationship between test scores, and bypassing the issue cortical neurodegenerative processes.
of colinearity. In the ACCORD and CSHA cohorts, a sub- Core executive functions assessed in NPT are planning,
group of CIND subjects defined by episodic memory dys- concept formation, sequential organization, and attention.
function alone had a two- to three-fold risk of dementia at When matched in severity, AD patients have an advantage
follow-up compared with subgroups defined by other dys- over DLB patients on tests of working memory, attention,
functions [44]. Two subgroups of subjects with recent cog- initiation, and perseveration, despite having poorer episodic
nitive decline had a four-fold risk of dementia; one was memory [83,85,93]. Qualitative analysis of neuropsycho-
defined by multiple cognitive impairments and the other by logical performance shows more inattention, distractibility,
episodic memory impairment [77]. impaired set shifting, incoherence, confabulation, perse-
In summary, NPT has been shown to provide useful veration, and intrusion in DLB compared with AD groups
prognostic information in both nonselected cohorts of el- [94]. As with other disorders of the basal ganglia, PDD
derly and selected groups with evidence of MCI. Episodic patients exhibit impaired problem solving, set maintenance,
memory tests appear to have the greatest predictive accu- and shifting attention, especially to novel stimuli [86,95].
racy, possibly in combination with executive functioning PDD patients also have similar fluctuations in attention as
tests. In part, this might reflect the fact that episodic memory seen in DLB [96], which partially accounts for mixed per-
impairment is the hallmark of AD, which, in turn, is the most formances in these groups on cognitive testing. On memory
prevalent form of dementia. The utility of NPT might be tasks PDD and DLB patients, similarly to those with FTD
further enhanced when performance on multiple tests is scru- and VaD, benefit from recognition or semantic cueing par-
tinized for patterns of impairment. The diagnostic/prognostic adigms at retrieval [92,97–101], whereas these paradigms
value of NPT would be further strengthened if, in addition are largely ineffective in AD [102,103]. The improvement
to group norms, statistics such as likelihood ratios were in recognition and recall as a result of retrieval cues sug-
developed for tests that are routinely used [78]. gests underlying impairment of working memory in FTD,
PDD, and DLB. Traditional tests of frontal lobe function,
4.3. The differential diagnosis of dementia such as the Wisconsin Card Sorting Test, Trail Making Test,
and Stroop Tests, have high sensitivity to executive dys-
NPT of memory, other cognitive domains, and behavior function, but they also have low specificity to dementia
complements clinical history and neuroimaging in differen- [104 –106].
tial diagnosis and in documenting the natural cognitive Language deficits are usually considered to occur in 8%
progression of various dementing illnesses. Distinguishable to 10% of early AD patients [107,108], although aphasia is
neuropsychological profiles are seen in patients diagnosed invariably present in the advanced stages [109]. Word find-
with various dementias including AD, VaD, dementia with ing difficulties with circumlocutions and fluent, articulated,
Lewy bodies (DLB), dementia associated with Parkinson’s and syntactically preserved speech are often seen in early
disease (PDD), and frontotemporal dementia (FTD). FTD cases [108,109], whereas the later dissolution of language in
encompasses distinct clinical subtypes including a behavioral AD resembles anomic, transcortical sensory, Wernicke’s,
variant (FTD-bv), progressive nonfluent aphasia (PNFA), se- and, finally, global aphasia [108]. Although language abil-
mantic dementia (SD), as well as corticobasal degeneration ities in PDD are largely intact compared with AD [110,111],
(CBD) and progressive supranuclear palsy (PSP) [79,80]. PD patients might have decreased information content,
Episodic memory deficit is a primary feature of AD in difficulty comprehending complex sentences, anomia, and
contrast to FTD, where it is spared initially [81,82], and to reduced fluency that likely relate to the dysexecutive syn-
a lesser extent in PDD, DLB, and VaD [83– 87]. Memory drome seen in this condition [112,113]. Language distur-
for recent events or names is affected early in AD, but with bance is also an early feature in some variants of FTD.
312 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

Phonemic paraphasias, word finding and picture naming but their performance shows a range of intact and impaired
difficulty, and decreased word list generation (letter more skills. This variable cognitive profile of VaD patients is
than semantic fluency) are usually evident during language considered a reflection of the location and extent of pathol-
testing in patients with PFNA. FTD-bv patients might also ogy, making a stereotypic neuropsychological pattern less
manifest anomia and reduction in spontaneous conversa- likely [140]. Similar patchy and inconsistent performance
tion. Although fluency might remain intact in the early can be seen in FTD-bv, although this seems due to impul-
stages, semantic dementia patients manifest decreased sivity and cursory or amotivational responding. On the
speech content and impaired performance on tests that re- CDT, which calls on multiple cognitive skills [141], plan-
quire access to semantic knowledge. They have deficits in ning, visuospatial, and conceptual errors predominate in
category fluency, noun definitions and comprehension, pic- DLB, PDD, and AD groups [142–144], whereas early FTD
ture naming, reading irregular words, and impoverished patients can perform better than AD patients [119]. The
results on the Pyramids and Palm Trees Tests, which require MMSE, probably the most widely used screening test, is not
individuals to match semantically associated items [114]. sensitive in early FTD [145,146]. Along with behavioral
Cognitive tests of visuoperceptual, visuoconstructive, measures, a double dissociation with FTD patients perform-
and visuospatial ability are often problematic for patients ing poorly on tasks requiring inference of mental states,
with DLB in the relatively early stages [115], and both DLB thoughts, and feelings of others (theory of mind) and AD
and PDD patients have greater visuospatial dysfunction than performing poorly on tests of episodic memory further aids
AD patients [84,93,111,115–117]. Cormack et al [116] differential diagnosis [147], although theory-of-mind tasks
found that constructional performance was associated with have yet to be designed so as to be easily incorporated into
global cognitive impairment in PDD and AD, whereas in NPT batteries in clinical practice. Because PDD and DLB
DLB it was only linked with perceptual and praxis ability. share overlapping clinical symptomatology [83,132] and
Visuospatial difficulty in PDD has been attributed to deficits neuropathology [148], it is not surprising that both have
in planning and sequencing of responses that are analogous similar cognitive and neuropsychiatric features of executive
to their motor impairment [118]. FTD groups perform better dysfunction, visuospatial deficits, fluctuating cognition, and
on visuospatial tasks than AD groups [119], and their poor hallucinations during the course of the disease. Fluctuations
performance on construction tests frequently results from in attention and alertness can result in mixed performance in
amotivation, poor planning, and impulsivity [91,106]. tests of sustained attention such as trail making, letter can-
Behavioral and personality alterations, particularly apathy, cellations, digit span, fluency, and others.
appetite changes, disinhibition, and stereotypic behaviors, In summary, the utility of NPT in the differential diagnosis
can help distinguish FTD from AD and VaD [120 –125], of dementia is variable and stage-dependent. Increased sen-
and Kertesz et al [106] found behavioral quantification to be sitivity is often accompanied by decreased specificity. Pa-
more sensitive than cognitive testing in differentiating FTD tients cannot be easily fitted in diagnostic categories on the
and AD groups. Patients presenting with semantic dementia basis of neuropsychological performance alone. Although
or PNFA develop behavioral changes similar to those seen there is currently no consensus on the type of specific tests
in FTD-bv, although later in the course of the illness that should be used in NPT batteries, differential diagnosis
[106,126,127]. In later stages of AD, aggression and disin- requires that there be comprehensive testing of memory,
hibited behaviors can also become prominent features language, visuospatial, and executive abilities.
[97,128,129]. Whereas AD patients typically have delusions
[130], it is visual hallucinations that are primary features of
5. Neuropsychological assessment and
DLB [131] and, to a lesser extent, PDD [132–134]. Depres-
management decisions
sion is also found to a greater extent in PDD than AD [111],
whereas when matched in severity, aberrant motor behavior, Beyond diagnostic issues, comprehensive neuropsycho-
agitation, disinhibition, irritability, euphoria, and apathy are logical assessment can make important contributions to
more severe in AD [133]. The neuropsychiatric features of management decisions in MCI and dementia. Although sys-
AD and VaD are generally similar, and it has been proposed tematic studies are lacking, this role of neuropsychology is
that the increased prevalence of depression and anxiety well-recognized in clinical practice both from the standpoint
occasionally reported in VaD [135–138] might be due to of testing in general [149] and from the standpoint of as-
mixed pathology in AD and VaD clinical samples and to the sessment of dementia specifically [150]. Neuropsychologi-
heterogeneity of VaD patients studied [139]. cal assessment focuses on an individualized approach to the
Tables 5 and 6 summarize evidence in support of diag- determination of preserved as well as impaired cognitive
nostic differentiation. At comparable dementia severity, abilities that is not dependent on the use of population-based
VaD patients exhibit greater deficits in executive function- cut points [149]. The information collected in the assess-
ing than tests of verbal learning and memory, whereas AD ment might serve in estimating the functional impact of the
patients exhibit the reverse pattern [87,101,137]. VaD pa- cognitive disorder and the management of functional dis-
tients often present with memory and executive difficulty, ability [151]. For persons still in the workforce, it might
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 313

play a major role in vocational decision making. Planning (D) Determining whether there has been progression of
for cognitive rehabilitation interventions in AD and other cognitive impairment or the development of new
forms of dementia also requires a comprehensive under- impairment(s) to assist in management (Grade A,
standing of retained cognitive abilities on the part of the Level 3).
affected individual [152].

6. Recommendations from the Third Canadian Author Disclosures


Consensus Conference on Diagnosis and Treatment Andrew Kertesz has received support from Pfizer Can-
of Dementia ada (Advisory Board Member), Janssen Ortho Inc.
6.1. Recommendations for brief cognitive tests (Speaker), Novartis (Speaker, Former Advisory Board
Member), and Lundbeck Canada (Speaker).
John Fisk has received support from AstraZenca (Consul-
1. The MoCA and the DemTect are more sensitive to
tant), Bayer (Consultant), Biogen-Idec (Consultant), Bristol-
MCI than the MMSE. Their use is recommended
Myers Squibb (Consultant), Novartis (Consultant), Sanofi-
when MCI is suspected. There is insufficient evi-
Aventis (Consultant) and TEVA Neuroscience (Speaker).
dence to recommend one test over the other (Grade
Howard H. Feldman has received support from Pfizer
B, Level 2— because replication in different set-
Canada (Grant/Research Support [External], Consultant,
tings, particularly the general practice setting, is
CME Programs), Eisai (Grant/Research Support [External],
required).
Consultant, CME Programs), Janssen Ortho Inc. (Grant/Re-
2. There is insufficient evidence to recommend the de-
search Support [External], Consultant, CME Programs), As-
layed three-trial MMSE adaptation, the STMS, and
traZeneca (Grant/Research Support [External], Consultant),
the SSST for the detection of MCI (Grade C, Level 2).
Sanofi Synthelabo (Grant/Research Support [External],
3. A range of brief cognitive tests, including the
Consultant), GlaxoSmithKline (Grant/Research Support
MoCA, DemTect, BNA, GPCOG, and the 7MS,
[External], Consultant), Novartis (Consultant, CME Pro-
might be more accurate than the MMSE in discrim-
inating dementia from normal. There is insufficient grams), Eli Lilly (Grant/Research Support [External]), Ser-
evidence to recommend one test over the others vier (Consultant), Myriad (Consultant), Targacept (Consul-
(Grade B, Level 2— because replication studies are tant), Lundbeck Canada (Consultant), Axonyx (Consultant),
needed). and Forest (CME).
4. The distinction between MCI and AD is important
and is currently made on the basis of clinical
assessment of cognition and function (Grade A, Acknowledgments
Level 3).
5. Brief cognitive tests might aid in the assessment of The authors gratefully acknowledge numerous sources of
this distinction between MCI and AD. The DemTect support and funding. Claudia Jacova was funded by a post-
and the STMS can be recommended because they doctoral Canadian Institutes of Health Research/Michael
have established cut points for both MCI and AD Smith Foundation for Health Research Training Fellowship
(Grade B, Level 2). in Neurobiology and Behaviour.
Andrew Kertesz received grant support through the Med-
6.2. Recommendations for neuropsychological testing ical Research Council (MRC) of Canada and the Pharma-
and assessment ceutical Manufacturers Association of Canada (PMAC),
MRC PMAC program #14197 (A Canadian Cohort Study of
6. The diagnosis and differential diagnosis of dementia is Cognitive Impairment and Related Dementias [ACCORD]),
currently a clinically integrative one. Neuropsycholog- and through the National Institutes of Health USA (NIH),
ical assessment alone cannot be used for this purpose #1 U01-AG24904-01 (Alzheimer’s Disease Neuroimaging
and should be used selectively in clinical settings Initiative [ADNI]).
(Grade B, Level 2). Mervin Blair was funded by grant support from Janssen
7. Neuropsychological assessment may aid in: and from the Lawson Research Institute.
(A) Addressing the distinction between normal aging, Howard Feldman received grant support through the
MCI-CIND, and early dementia (Grade B, Level 2); MRC-PMAC, program #14197 (ACCORD), the National
(B) Addressing the risk of progression from MCI- Institutes of Health USA (NIH), #1 U01-AG24904-01
CIND to dementia or AD (Grade B, Level 2); (ADNI), and through the Canadian Institutes of Health
(C) Differential diagnosis of dementia and other syn- Research (CIHR) operating grant #74580 (Frontotemporal
dromes of cognitive impairment (Grade B, Level 2); Dementia with Ubiquinated Inclusions).
314 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

References [19] Eibenstein A, Fioretti AB, Simaskou MN, Sucapane P, Mearelli S,


Mina C, et al. Olfactory screening test in mild cognitive impairment.
[1] The Houston conference on specialty education and training in Neurol Sci 2005;26:156 – 60.
clinical neuropsychology. Policy statement. Archives of Clinical [20] Doty RL, Reyes PF, Gregor T. Presence of both odor identification
Neuropsychology 1998;157–250. and detection deficits in Alzheimer’s disease. Brain Res Bull 1987;
[2] Mulherin SA, Miller WC. Spectrum bias or spectrum effect? sub- 18:597– 600.
group variation in diagnostic test evaluation. Ann Intern Med 2002; [21] Morgan CD, Murphy C. Olfactory event-related potentials in Alz-
137:598 – 602. heimer’s disease. J Int Neuropsychol Soc 2002;8:753– 63.
[3] Lorentz WJ, Scanlan JM, Borson S. Brief screening tests for de- [22] Teng EL, Chui HC. The Modified Mini-Mental State (3MS) exam-
mentia. Can J Psychiatry 2002;47:723–33. ination. J Clin Psychiatry 1987;48:314 – 8.
[4] Folstein MF, Robins LN, Helzer JE. The Mini-Mental State Exam- [23] McDowell I, Hill G, Lindsay J, Helliwell B, Costa L, Beattie BL,
ination. Arch Gen Psychiatry 1983;40:812. et al. Canadian Study of Health and Aging: study methods and
[5] Tombaugh TN, McIntyre NJ. The mini-mental state examination: a prevalence of dementia. CMAJ 1994;150:899 –913.
comprehensive review. J Am Geriatr Soc 1992;40:922–35. [24] Tuokko H, Hadjistavropoulos T, Miller JA, Beattie BL. The Clock
[6] Tombaugh TN, McDowell I, Kristjansson B, Hubley AM. Mini- Test: a sensitive measure to differentiate normal elderly from those
Mental State Examination (MMSE) and the modified MMSE with Alzheimer disease. J Am Geriatr Soc 1992;40:579 – 84.
(3MS): a psychometric comparison and normative data. Psychol [25] Bush C, Kozak J, Elmslie T. Screening for cognitive impairment in
Assess 1996;8:48 –59. the elderly. Can Fam Physician 1997;43:1763– 8.
[7] Lindsay J, Laurin D, Verreault R, Hebert R, Helliwell B, Hill GB, [26] Shulman KI. Clock-drawing: is it the ideal cognitive screening test?
et al. Risk factors for Alzheimer’s disease: a prospective analysis Int J Geriatr Psychiatry 2000;15:548 – 61.
from the Canadian Study of Health and Aging. Am J Epidemiol [27] Morris JC. Clinical dementia rating: a reliable and valid diagnostic
2002;156:445–53. and staging measure for dementia of the Alzheimer type. Int Psy-
[8] Kraemer HC, Moritz DJ, Yesavage J. Adjusting Mini-Mental State chogeriatr 1997;9(Suppl 1):173– 6.
Examination scores for age and educational level to screen for [28] Powlishta KK, Von Dras DD, Stanford A, Carr DB, Tsering C,
dementia: correcting bias or reducing validity? Int Psychogeriatr Miller JP, et al. The clock drawing test is a poor screen for very mild
1998;10:43–51. dementia. Neurology 2002;59:898 –903.
[9] Feldman HH, Jacova C. Mild cognitive impairment. Am J Geriatr [29] De Lepeleire J, Heyman J, Baro F, Buntinx F. A combination of
Psychiatry 2005;13:645–55. tests for the diagnosis of dementia had a significant diagnostic value.
[10] Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, J Clin Epidemiol 2005;58:217–25.
Kokmen E. Mild cognitive impairment: clinical characterization [30] Stahelin HB, Monsch AU, Spiegel R. Early diagnosis of dementia
and outcome. Arch Neurol 1999;56:303– 8. via a two-step screening and diagnostic procedure. Int Psychogeriatr
[11] Graham JE, Rockwood K, Beattie BL, Eastwood R, Gauthier S, 1997;9(Suppl 1):123–30.
Tuokko H, et al. Prevalence and severity of cognitive impairment [31] Solomon PR, Hirschoff A, Kelly B, Relin M, Brush M, DeVeaux
with and without dementia in an elderly population. Lancet 1997; RD, et al. A 7 minute neurocognitive screening battery highly
349:1793– 6. sensitive to Alzheimer’s disease. Arch Neurol 1998;55:349 –55.
[12] Fisk JD, Merry HR, Rockwood K. Variations in case definition
[32] Solomon PR, Brush M, Calvo V, Adams F, DeVeaux RD, Pendle-
affect prevalence but not outcomes of mild cognitive impairment.
bury WW, et al. Identifying dementia in the primary care practice.
Neurology 2003;61:1179 – 84.
Int Psychogeriatr 2000;12:483–93.
[13] Feldman H, Levy AR, Hsiung GY, Peters KR, Donald A, Black SE,
[33] Kuslansky G, Buschke H, Katz M, Sliwinski M, Lipton RB. Screen-
et al. A Canadian cohort study of cognitive impairment and related
ing for Alzheimer’s disease: the memory impairment screen versus
dementias (ACCORD): study methods and baseline results. Neuro-
the conventional three-word memory test. J Am Geriatr Soc 2002;
epidemiology 2003;22:265–74.
50:1086 –91.
[14] Petersen RC, Stevens JC, Ganguli M, Tangalos EG, Cummings JL,
[34] Brodaty H, Pond D, Kemp NM, Luscombe G, Harding L, Berman
DeKosky ST. Practice parameter: early detection of dementia: mild
K, et al. The GPCOG: A new screening test for dementia designed
cognitive impairment (an evidence-based review)—report of the
Quality Standards Subcommittee of the American Academy of Neu- for general practice. J Am Geriatr Soc 2002;50:530 – 4.
rology. Neurology 2001;56:1133– 42. [35] Kilada S, Gamaldo A, Grant EA, Moghekar A, Morris JC, O’Brien
[15] Nasreddine ZS, Phillips NA, Bedirian V, Charbonneau S, White- RJ. Brief screening tests for the diagnosis of dementia: comparison
head V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: with the mini-mental state exam. Alzheimer Dis Assoc Disord 2005;
a brief screening tool for mild cognitive impairment. J Am Geriatr 19:8 –16.
Soc 2005;53:695–9. [36] Storey JE, Rowland JT, Basic D, Conforti DA, Dickson HG. The
[16] Kalbe E, Kessler J, Calabrese P, Smith R, Passmore AP, Brand M, Rowland Universal Dementia Assessment Scale (RUDAS): a mul-
et al. DemTect: a new, sensitive cognitive screening test to support ticultural cognitive assessment scale. Int Psychogeriatr 2004;16:
the diagnosis of mild cognitive impairment and early dementia. Int 13–31.
J Geriatr Psychiatry 2004;19:136 – 43. [37] Darvesh S, Leach L, Black SE, Kaplan E, Freedman M. The
[17] Loewenstein DA, Barker WW, Harwood DG, Luis C, Acevedo A, behavioural neurology assessment. Can J Neurol Sci 2005;32:
Rodriguez I, et al. Utility of a modified Mini-Mental State Exami- 167–77.
nation with extended delayed recall in screening for mild cognitive [38] Stuss DT, Meiran N, Guzman DA, Lafleche G, Willmer J. Do long
impairment and dementia among community dwelling elders. Int J tests yield a more accurate diagnosis of dementia than short tests? a
Geriatr Psychiatry 2000;15:434 – 40. comparison of 5 neuropsychological tests. Arch Neurol 1996;53:
[18] Tang-Wai DF, Knopman DS, Geda YE, Edland SD, Smith GE, 1033–9.
Ivnik RJ, et al. Comparison of the short test of mental status and the [39] Chui H, Zhang Q. Evaluation of dementia: a systematic study of the
mini-mental state examination in mild cognitive impairment. Arch usefulness of the American Academy of Neurology’s practice pa-
Neurol 2003;60:1777– 81. rameters. Neurology 1997;49:925–35.
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 315

[40] Chertkow H, Bergman H, Schipper HM, Gauthier S, Bouchard R, the Alzheimer type: a prospective population-based study. Brain
Fontaine S, et al. Assessment of suspected dementia. Can J Neurol 2005;128(Pt 5):1093–101.
Sci 2001;28(Suppl 1):28 – 41. [59] Chen P, Ratcliff G, Belle SH, Cauley JA, DeKosky ST, Ganguli M.
[41] McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan Patterns of cognitive decline in presymptomatic Alzheimer disease:
EM. Clinical diagnosis of Alzheimer’s disease: report of the a prospective community study. Arch Gen Psychiatry 2001;58:
NINCDS-ADRDA Work Group under the auspices of Department 853– 8.
of Health and Human Services Task Force on Alzheimer’s Disease. [60] Maruff P, Collie A, Darby D, Weaver-Cargin J, Masters C, Currie J.
Neurology 1984;34:939 – 44. Subtle memory decline over 12 months in mild cognitive impair-
[42] Levy R. Aging-associated cognitive decline: Working Party of the ment. Dement Geriatr Cogn Disord 2004;18:342– 8.
International Psychogeriatric Association in collaboration with the [61] Fowler KS, Saling MM, Conway EL, Semple JM, Louis WJ. Paired
World Health Organization. Int Psychogeriatr 1994;6:63– 8. associate performance in the early detection of DAT. J Int Neuro-
[43] Peters KR, Graf P, Hayden S, Feldman H. Neuropsychological psychol Soc 2002;8:58 –71.
characterization of cognitively-impaired-not-demented (CIND) in- [62] Bowen J, Teri L, Kukull W, McCormick W, McCurry SM, Larson
dividuals: clinical comparison data. Clin Neuropsychol 2004;18: EB. Progression to dementia in patients with isolated memory loss.
208 –28. Lancet 1997;349:763–5.
[44] Peters KR, Graf P, Hayden S, Feldman H. Neuropsychological [63] Tierney MC, Yao C, Kiss A, McDowell I. Neuropsychological tests
subgroups of cognitively-impaired-not-demented (CIND) individu- accurately predict incident Alzheimer disease after 5 and 10 years.
als: delineation, reliability, and predictive validity. J Clin Exp Neu- Neurology 2005;64:1853–9.
ropsychol 2005;27:164 – 88. [64] Tierney MC, Szalai JP, Snow WG, Fisher RH, Nores A, Nadon
[45] Lopez OL, Becker JT, Jagust WJ, Fitzpatrick A, Carlson MC, G, et al. Prediction of probable Alzheimer’s disease in memory-im-
DeKosky ST, et al. Neuropsychological characteristics of mild cog- paired patients: a prospective longitudinal study. Neurology 1996;46:
nitive impairment subgroups. J Neurol Neurosurg Psychiatry 2006; 661–5.
77:159 – 65. [65] Arnaiz E, Almkvist O, Ivnik RJ, Tangalos EG, Wahlund LO, Win-
[46] Grundman M, Petersen RC, Ferris SH, Thomas RG, Aisen PS, blad B, et al. Mild cognitive impairment: a cross-national compar-
Bennett DA, et al. Mild cognitive impairment can be distinguished ison. J Neurol Neurosurg Psychiatry 2004;75:1275– 80.
from Alzheimer disease and normal aging for clinical trials. Arch [66] Artero S, Ritchie K. The detection of mild cognitive impairment in
Neurol 2004;61:59 – 66. the general practice setting. Aging Ment Health 2003;7:251– 8.
[47] Kramer JH, Nelson A, Johnson JK, Yaffe K, Glenn S, Rosen HJ, [67] Elias MF, Beiser A, Wolf PA, Au R, White RF, D’Agostino RB.
et al. Multiple cognitive deficits in amnestic mild cognitive impair- The preclinical phase of Aalzheimer disease: a 22-year prospective
ment. Dement Geriatr Cogn Disord 2006;22:306 –11. study of the Framingham Cohort. Arch Neurol 2000;57:808 –13.
[48] Collie A, Maruff P, Currie J. Behavioral characterization of mild [68] Howieson DB, Dame A, Camicioli R, Sexton G, Payami H, Kaye
cognitive impairment. J Clin Exp Neuropsychol 2002;24:720 –33. JA. Cognitive markers preceding Alzheimer’s dementia in the
[49] Welsh K, Butters N, Hughes J, Mohs R, Heyman A. Detection of healthy oldest old. J Am Geriatr Soc 1997;45:584 –9.
abnormal memory decline in mild cases of Alzheimer’s disease using [69] Blackwell AD, Sahakian BJ, Vesey R, Semple JM, Robbins TW,
CERAD neuropsychological measures. Arch Neurol 1991;48:278 – 81. Hodges JR. Detecting dementia: novel neuropsychological markers
[50] Cahn DA, Salmon DP, Butters N, Wiederholt WC, Corey-Bloom J, of preclinical Alzheimer’s disease. Dement Geriatr Cogn Disord
Edelstein SL, et al. Detection of dementia of the Alzheimer type in 2004;17:42– 8.
a population-based sample: neuropsychological test performance. [70] Palmer K, Backman L, Winblad B, Fratiglioni L. Detection of
J Int Neuropsychol Soc 1995;1:252– 60. Alzheimer’s disease and dementia in the preclinical phase: popula-
[51] Swearer JM, O’Donnell BF, Kane KJ, Hoople NE, Lavoie M. tion based cohort study. BMJ 2003;326:245.
Delayed recall in dementia: sensitivity and specificity in patients [71] Devanand DP, Folz M, Gorlyn M, Moeller JR, Stern Y. Question-
with higher than average general intellectual abilities. Neuropsychi- able dementia: clinical course and predictors of outcome. J Am
atry Neuropsychol Behav Neurol 1998;11:200 – 6. Geriatr Soc 1997;45:321– 8.
[52] American Psychiatric Association. Diagnostic and statistical manual [72] Bozoki A, Giordani B, Heidebrink JL, Berent S, Foster NL. Mild
of mental disorders, fourth edition, text revision (DSM-IV-TR). cognitive impairments predict dementia in nondemented elderly
Arlington, VA. American Psychiatric Association; 2000. patients with memory loss. Arch Neurol 2001;58:411– 6.
[53] Steenhuis RE, Ostbye T. Neuropsychological test performance of [73] Backman L, Jones S, Berger AK, Laukka EJ, Small BJ. Cognitive
specific diagnostic groups in the Canadian Study of Health and impairment in preclinical Alzheimer’s disease: a meta-analysis.
Aging (CSHA). J Clin Exp Neuropsychol 1995;17:773– 85. Neuropsychology 2005;19:520 –31.
[54] Lambon Ralph MA, Patterson K, Graham N, Dawson K, Hodges JR. [74] Herlitz A, Small BJ, Fratiglioni L, Almkvist O, Viitanen M,
Homogeneity and heterogeneity in mild cognitive impairment and Backman L. Detection of mild dementia in community surveys:
Alzheimer’s disease: a cross-sectional and longitudinal study of 55 is it possible to increase the accuracy of our diagnostic instru-
cases. Brain 2003;126(Pt 11):2350 – 62. ments? Arch Neurol 1997;54:319 –24.
[55] Small BJ, Fratiglioni L, Viitanen M, Winblad B, Backman L. The [75] Tian J, Bucks RS, Haworth J, Wilcock G. Neuropsychological
course of cognitive impairment in preclinical Alzheimer disease: prediction of conversion to dementia from questionable dementia:
three- and 6-year follow-up of a population-based sample. Arch statistically significant but not yet clinically useful. J Neurol Neu-
Neurol 2000;57:839 – 44. rosurg Psychiatry 2003;74:433– 8.
[56] Hall CB, Lipton RB, Sliwinski M, Stewart WF. A change point [76] Fabrigoule C, Rouch I, Taberly A, Letenneur L, Commenges D,
model for estimating the onset of cognitive decline in preclinical Mazaux JM, et al. Cognitive process in preclinical phase of demen-
Alzheimer’s disease. Stat Med 2000;19:1555– 66. tia. Brain 1998;121(Pt 1):135– 41.
[57] Rubin EH, Storandt M, Miller JP, Kinscherf DA, Grant EA, Morris JC, [77] Ritchie K, Leibovici D, Ledesert B, Touchon J. A typology of sub-
et al. A prospective study of cognitive function and onset of dementia clinical senescent cognitive disorder. Br J Psychiatry 1996;168:470 – 6.
in cognitively healthy elders. Arch Neurol 1998;55:395– 401. [78] Ivnik RJ, Smith GE, Cerhan JH, Boeve BF, Tangalos EG, Petersen
[58] Amieva H, Jacqmin-Gadda H, Orgogozo JM, Carret N, Helmer C, RC. Understanding the diagnostic capabilities of cognitive tests.
Letenneur L, et al. The 9 year cognitive decline before dementia of Clin Neuropsychol 2001;15:114 –24.
316 C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317

[79] Neary D, Snowden J, Mann D. Frontotemporal dementia. Lancet [99] Helkala EL, Laulumaa V, Soininen H, Riekkinen PJ. Recall and
Neurol 2005;4:771– 80. recognition memory in patients with Alzheimer’s and Parkinson’s
[80] Feldman H, Kertesz A. Diagnosis, classification and natural history diseases. Ann Neurol;24:214 –7.
of degenerative dementias. Can J Neurol Sci 2001;28(Suppl 1): [100] Pillon B, Deweer B, Agid Y, Dubois B. Explicit memory in Alz-
S17–27. heimer’s, Huntington’s, and Parkinson’s diseases. Arch Neurol
[81] Gustafson L. Frontal lobe degeneration of non-Alzheimer type. II. 1993;50:374 –9.
Clinical picture and differential diagnosis. Arch Gerontol Geriatr [101] Tierney MC, Black SE, Szalai JP, Snow WG, Fisher RH, Nadon
1987;6:209 –23. G, et al. Recognition memory and verbal fluency differentiate
[82] Neary D, Snowden JS, Northen B, Goulding P. Dementia of frontal probable Alzheimer disease from subcortical ischemic vascular
lobe type. J Neurol Neurosurg Psychiatry 1988;51:353– 61. dementia. Arch Neurol 2001;58:1654 –9.
[83] Aarsland D, Litvan I, Salmon D, Galasko D, Wentzel-Larsen T, [102] Branconnier RJ, Cole JO, Spera KF, DeVitt DR. Recall and recog-
Larsen JP. Performance on the dementia rating scale in Parkinson’s nition as diagnostic indices of malignant memory loss in senile
disease with dementia and dementia with Lewy bodies: comparison dementia: a Bayesian analysis. Exp Aging Res 1982;8:189 –93.
[103] Davis PE, Mumford SJ. Cued recall and the nature of the memory
with progressive supranuclear palsy and Alzheimer’s disease. J Neu-
disorder in dementia. Br J Psychiatry 1984;144:383– 6.
rol Neurosurg Psychiatry 2003;74:1215–20.
[104] Chen P, Ratcliff G, Belle SH, Cauley JA, DeKosky ST, Ganguli
[84] Ballard CG, Ayre G, O’Brien J, Sahgal A, McKeith IG, Ince PG, et
M. Cognitive tests that best discriminate between presymptom-
al. Simple standardised neuropsychological assessments aid in the
atic AD and those who remain nondemented. Neurology 2000;
differential diagnosis of dementia with Lewy bodies from Alzhei-
55:1847–53.
mer’s disease and vascular dementia. Dement Geriatr Cogn Disord
[105] Grossman M. Frontotemporal dementia: a review. J Int Neuropsy-
1999;10:104 – 8. chol Soc 2002;8:566 – 83.
[85] Calderon J, Perry RJ, Erzinclioglu SW, Berrios GE, Dening TR, [106] Kertesz A, Davidson W, McCabe P, Munoz D. Behavioral quanti-
Hodges JR. Perception, attention, and working memory are dispropor- tation is more sensitive than cognitive testing in frontotemporal
tionately impaired in dementia with Lewy bodies compared with Alz- dementia. Alzheimer Dis Assoc Disord 2003;17:223–9.
heimer’s disease. J Neurol Neurosurg Psychiatry 2001;70:157– 64. [107] Faber-Langendoen K, Morris JC, Knesevich JW, LaBarge E, Miller
[86] Litvan I, Mohr E, Williams J, Gomez C, Chase TN. Differential JP, Berg L. Aphasia in senile dementia of the Alzheimer type. Ann
memory and executive functions in demented patients with Parkin- Neurol 1988;23:365–70.
son’s and Alzheimer’s disease. J Neurol Neurosurg Psychiatry 1991; [108] Kertesz A, Appell J, Fisman M. The dissolution of language in
54:25–9. Alzheimer’s disease. Can J Neurol Sci 1986;13(Suppl):415– 8.
[87] Looi JC, Sachdev PS. Differentiation of vascular dementia from AD [109] Appell J, Kertesz A, Fisman M. A study of language functioning in
on neuropsychological tests. Neurology 1999;53:670 – 8. Alzheimer patients. Brain Lang 1982;17:73–91.
[88] Corkin S. Some relationships between global amnesia and the mem- [110] Cummings JL, Darkins A, Mendez M, Hill MA, Benson DF. Alz-
ory impairment in Alzheimer’s disease. In: Corkin S, Davis K, heimer’s disease and Parkinson’s disease: comparison of speech and
Growdon JH, eds. Alzheimer’s disease: a report of progress on language alterations. Neurology 1988;38:680 – 4.
research. New York: Raven Press; 1982. p. 149 – 64. [111] Huber SJ, Shuttleworth EC, Freidenberg DL. Neuropsychological
[89] Karlsson T, Backman L, Herlitz A, Nilsson LG, Winblad B, differences between the dementias of Alzheimer’s and Parkinson’s
Osterlind PO. Memory improvement at different stages of Alz- diseases. Arch Neurol 1989;46:1287–91.
heimer’s disease. Neuropsychologia 1989;27:737– 42. [112] Grossman M, Carvell S, Gollomp S, Stern MB, Vernon G, Hurtig
[90] Martin A, Brouwers P, Cox C, Fedio P. On the nature of the HI. Sentence comprehension and praxis deficits in Parkinson’s dis-
verbal memory deficit in Alzheimer’s disease. Brain Lang 1985; ease. Neurology 1991;41:1620 – 6.
25:323– 41. [113] Grossman M, Carvell S, Stern MB, Gollomp S, Hurtig HI. Sentence
[91] Hodges JR. Frontotemporal dementia (Pick’s disease): clinical fea- comprehension in Parkinson’s disease: the role of attention and
tures and assessment. Neurology 2001;56(Suppl 4):6 –10. memory. Brain Lang 1992;42:347– 84.
[92] Pasquier F, Grymonprez L, Lebert F, Van der Linden M. Memory [114] Howard D, Patterson K. Pyramids and palm trees: a test of semantic
impairment differs in frontotemporal dementia and Alzheimer’s access from pictures and words. St Edmunds, Suffolk, UK: Thames
Valley Test Company; 1992.
disease. Neurocase 2001;7:161–71.
[115] Shimomura T, Mori E, Yamashita H, Imamura T, Hirono N, Hashi-
[93] Collerton D, Burn D, McKeith I, O’Brien J. Systematic review and
moto M, et al. Cognitive loss in dementia with Lewy bodies and
meta-analysis show that dementia with Lewy bodies is a visual-
Alzheimer disease. Arch Neurol 1998;55:1547–52.
perceptual and attentional-executive dementia. Dement Geriatr
[116] Cormack F, Aarsland D, Ballard C, Tovee MJ. Pentagon drawing
Cogn Disord 2003;16:229 –37.
and neuropsychological performance in dementia with Lewy bodies,
[94] Doubleday EK, Snowden JS, Varma AR, Neary D. Qualitative per-
Alzheimer’s disease, Parkinson’s disease and Parkinson’s disease
formance characteristics differentiate dementia with Lewy bodies and with dementia. Int J Geriatr Psychiatry 2004;19:371–7.
Alzheimer’s disease. J Neurol Neurosurg Psychiatry 2002;72:602–7. [117] Stern Y, Richards M, Sano M, Mayeux R. Comparison of cognitive
[95] Emre M. Dementia associated with Parkinson’s disease. Lancet changes in patients with Alzheimer’s and Parkinson’s disease. Arch
Neurol 2003;2:229 –37. Neurol 1993;50:1040 –5.
[96] Ballard CG, Aarsland D, McKeith I, O’Brien J, Gray A, Cormack F, [118] Stern Y, Mayeux R, Rosen J, Ilson J. Perceptual motor dysfunction
et al. Fluctuations in attention: PD dementia vs DLB with parkin- in Parkinson’s disease: a deficit in sequential and predictive volun-
sonism. Neurology 2002;59:1714 –20. tary movement. J Neurol Neurosurg Psychiatry 1983;46:145–51.
[97] Mendez MF, Cummings JL. Dementia: a clinical approach, 3 ed. [119] Rascovsky K, Salmon DP, Ho GJ, Galasko D, Peavy GM, Hansen
Philadelphia: Butterworth-Heinemann (Elsevier); 2003. LA, et al. Cognitive profiles differ in autopsy-confirmed frontotem-
[98] Hamilton JM, Salmon DP, Galasko D, Delis DC, Hansen LA, poral dementia and AD. Neurology 2002;58:1801– 8.
Masliah E, et al. A comparison of episodic memory deficits in [120] Bathgate D, Snowden JS, Varma A, Blackshaw A, Neary D. Be-
neuropathologically-confirmed dementia with Lewy bodies and Alz- haviour in frontotemporal dementia, Alzheimer’s disease and vas-
heimer’s disease. J Int Neuropsychol Soc 2004;10:689 –97. cular dementia. Acta Neurol Scand 2001;103:367–78.
C. Jacova et al. / Alzheimer’s & Dementia 3 (2007) 299 –317 317

[121] Bozeat S, Gregory CA, Ralph MA, Hodges JR. Which neuropsy- [141] Freedman M, Leach L, Kaplan E, Shulman KI. Clock drawing. a
chiatric and behavioural features distinguish frontal and temporal neuropsychological analysis. New York: Oxford University Press;
variants of frontotemporal dementia from Alzheimer’s disease? 1994.
J Neurol Neurosurg Psychiatry 2000;69:178 – 86. [142] Cahn-Weiner DA, Williams K, Grace J, Tremont G, Westervelt H,
[122] Kertesz A, Davidson W, Fox H. Frontal behavioral inventory: di- Stern RA. Discrimination of dementia with Lewy bodies from Alz-
agnostic criteria for frontal lobe dementia. Can J Neurol Sci 1997; heimer disease and Parkinson disease using the clock drawing test.
24:29 –36. Cogn Behav Neurol 2003;16:85–92.
[123] Kertesz A, Nadkarni N, Davidson W, Thomas AW. The Frontal [143] Rouleau I, Salmon DP, Butters N, Kennedy C, McGuire K. Quan-
Behavioral Inventory in the differential diagnosis of frontotemporal titative and qualitative analyses of clock drawings in Alzheimer’s
dementia. J Int Neuropsychol Soc 2000;6:460 – 8. and Huntington’s disease. Brain Cogn 1992;18:70 – 87.
[124] Levy ML, Miller BL, Cummings JL, Fairbanks LA, Craig A. Alz- [144] Rouleau I, Salmon DP, Butters N. Longitudinal analysis of clock
heimer disease and frontotemporal dementias: behavioral distinc- drawing in Alzheimer’s disease patients. Brain Cogn 1996;31:17–34.
tions. Arch Neurol 1996;53:687–90. [145] Gregory CA, Hodges JR. Clinical features of frontal lobe dementia
[125] Miller BL, Ikonte C, Ponton M, Levy M, Boone K, Darby A, et al. A in comparison to Alzheimer’s disease. J Neural Transm Suppl 1996;
study of the Lund-Manchester research criteria for frontotemporal de- 47:103–23.
mentia: clinical and single-photon emission CT correlations. Neurology [146] Miller BL, Cummings JL, Villanueva-Meyer J, et al. Frontal lobe
1997;48:937– 42. degeneration: clinical, neuropsychological, and SPECT characteris-
[126] Davies RR, Hodges JR, Kril JJ, Patterson K, Halliday GM, Xuereb tics. Neurology 1991;41:1374 – 82.
JH. The pathological basis of semantic dementia. Brain 2005;128(Pt [147] Gregory C, Lough S, Stone V, Erzinclioglu S, Martin L, Baron-Cohen
9):1984 –95. S, et al. Theory of mind in patients with frontal variant frontotemporal
[127] Marczinski CA, Davidson W, Kertesz A. A longitudinal study of dementia and Alzheimer’s disease: theoretical and practical implica-
behavior in frontotemporal dementia and primary progressive apha- tions. Brain 2002;125(Pt 4):752– 64.
sia. Cogn Behav Neurol 2004;17:185–90. [148] Colosimo C, Hughes AJ, Kilford L, Lees AJ. Lewy body cortical
[128] Devanand DP. The interrelations between psychosis, behavioral involvement may not always predict dementia in Parkinson’s dis-
disturbance, and depression in Alzheimer disease. Alzheimer Dis ease. J Neurol Neurosurg Psychiatry 2003;74:852– 6.
Assoc Disord 1999;13(Suppl 2):S3– 8. [149] Lezak MD, Howieson DB, Loring DW. Neuropsychological Assess-
[129] Mendez MF, Martin RJ, Smyth KA, Whitehouse PJ. Psychiatric
ment. New York: Oxford University Press; 2004.
symptoms associated with Alzheimer’s disease. J Neuropsychiatry
[150] Morgan CD, Baade LE. Neuropsychological testing and assessment
Clin Neurosci 1990;2:28 –33.
scales for dementia of the Alzheimer’s type. Psychiatr Clin North
[130] Paulsen JS, Salmon DP, Thal LJ, Romero R, Weisstein-Jenkins C,
Am 1997;20:25– 43.
Galasko D, et al. Incidence of and risk factors for hallucinations and
[151] Sano M. Neuropsychological testing in the diagnosis of dementia. J
delusions in patients with probable AD. Neurology 2000;54:1965–71.
Geriatr Psychiatry Neurol 2006;19:155–9.
[131] Ballard C, Holmes C, McKeith I, Neill D, O’Brien J, Cairns N, et al.
[152] De Vreese LP, Neri M, Fioravanti M, Belloi L, Zanetti O. Memory
Psychiatric morbidity in dementia with Lewy bodies: a prospective
rehabilitation in Alzheimer’s disease: a review of progress. Int J
clinical and neuropathological comparative study with Alzheimer’s
Geriatr Psychiatry 2001;16:794 – 809.
disease. Am J Psychiatry 1999;156:1039 – 45.
[153] Christensen H, Griffiths K, Mackinnon A, Jacomb P. A quantitative
[132] Aarsland D, Ballard C, Larsen JP, McKeith I. A comparative study
review of cognitive deficits in depression and Alzheimer-type de-
of psychiatric symptoms in dementia with Lewy bodies and Parkin-
mentia. J Int Neuropsychol Soc 1997;3:631–51.
son’s disease with and without dementia. Int J Geriatr Psychiatry
2001;16:528 –36. [154] Simard M, van RR, Cohen T. A review of the cognitive and behav-
[133] Aarsland D, Cummings JL, Larsen JP. Neuropsychiatric differences ioral symptoms in dementia with Lewy bodies. J Neuropsychiatry
between Parkinson’s disease with dementia and Alzheimer’s dis- Clin Neurosci 2000;12:425–50.
ease. Int J Geriatr Psychiatry 2001;16:184 –91. [155] Kramer JH, Jurik J, Sha SJ, Rankin KP, Rosen HJ, Johnson JK, et
[134] Fenelon G, Mahieux F, Huon R, Ziegler M. Hallucinations in Par- al. Distinctive neuropsychological patterns in frontotemporal de-
kinson’s disease: prevalence, phenomenology and risk factors. Brain mentia, semantic dementia, and Alzheimer disease. Cogn Behav
2000;123(Pt 4):733– 45. Neurol 2003;16:211– 8.
[135] Ballard C, Neill D, O’Brien J, McKeith IG, Ince P, Perry R. Anxiety, [156] Mathuranath PS, Nestor PJ, Berrios GE, Rakowicz W, Hodges JR.
depression and psychosis in vascular dementia: prevalence and as- A brief cognitive test battery to differentiate Alzheimer’s disease
sociations. J Affect Disord 2000;59:97–106. and frontotemporal dementia. Neurology 2000;55:1613–20.
[136] Cummings JL, Miller B, Hill MA, Neshkes R. Neuropsychiatric [157] Gorno-Tempini ML, Dronkers NF, Rankin KP, Ogar JM, Phen-
aspects of multi-infarct dementia and dementia of the Alzheimer grasamy L, Rosen HJ, et al. Cognition and anatomy in three variants
type. Arch Neurol 1987;44:389 –93. of primary progressive aphasia. Ann Neurol 2004;55:335– 46.
[137] Starkstein SE, Sabe L, Vazquez S, Teson A, Petracca G, Chemer- [158] Hodges JR, Patterson K, Ward R, Garrard P, Bak T, Perry R,
inski E, et al. Neuropsychological, psychiatric, and cerebral blood Gregory C. The differentiation of semantic dementia and frontal
flow findings in vascular dementia and Alzheimer’s disease. Stroke lobe dementia (temporal and frontal variants of frontotemporal de-
1996;27:408 –14. mentia) from early Alzheimer’s disease: a comparative neuropsy-
[138] Sultzer DL, Levin HS, Mahler ME, High WM, Cummings JL. A chological study. Neuropsychology 1999;13:31– 40.
comparison of psychiatric symptoms in vascular dementia and Alz- [159] Sjogren M, Wallin A, Edman A. Symptomatological characteristics
heimer’s disease. Am J Psychiatry 1993;150:1806 –12. distinguish between frontotemporal dementia and vascular dementia
[139] Srikanth S, Nagaraja AV, Ratnavalli E. Neuropsychiatric symptoms with a dominant frontal lobe syndrome. Int J Geriatr Psychiatry
in dementia-frequency, relationship to dementia severity and com- 1997;12:656 – 61.
parison in Alzheimer’s disease, vascular dementia and frontotem- [160] Butters N, Wolfe J, Martone M, Granholm E, Cermak LS. Memory
poral dementia. J Neurol Sci 2005;236:43– 8. disorders associated with Huntington’s disease: verbal recall, verbal
[140] Rao R, Howard R. Vascular dementia: dead or alive? Int J Geriatr recognition and procedural memory. Neuropsychologia 1985;23:
Psychiatry 1998;13:277– 84. 729 – 43.
Alzheimer’s & Dementia 3 (2007) 318 –324

Cobalamin and homocysteine in older adults: Do we need to test


for serum levels in the work-up of dementia?
Angeles Garcia*
Department of Medicine (Geriatrics), Queen’s University, Kingston, Ontario, Canada

Abstract This article reviews available information on the relationship between cobalamin (Cbl), homo-
cysteine (tHcy) and cognitive decline and dementia in older adults with the aim to propose
recommendations as to the need to perform such determinations in the work-up for dementia. The
article includes brief reviews of the magnitude of the problem, effects of the Canadian folic acid
fortification program on the Cbl and tHcy status in older adults, relation between Cbl and tHcy and
cognition and cognitive changes, and available data on treatment trials up to March 2007. Recom-
mendations and levels of evidence were assigned and approved by consensus following the
directives of the CCCDTD3. The review concludes that determination of Cbl (vit B12) levels are
recommended in the work-up for dementia and cognitive decline because of the high prevalence of
Cbl deficits in this population, independently of the possible effects of normalization of Cbl levels
on cognitive function. Even though elevated tHcy is a risk factor for dementia, there is no prooof
that normalization of tHcy levels changes the course of the disease. Clinical trials on that regard are
on going.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Diagnosis; Dementia; Cobalamin; Vitamin B12; Homocysteine; Folate

1. Introduction of dementia more frequent among younger adults. Aspects


that might differentiate Canadian population from other
The Third Canadian Consensus Conference on the Diag-
countries’ populations have been emphasized.
nosis and Treatment of Dementia started from the assump-
Low cobalamin (Cbl or B12), a commonly occurring
tion that previously published guidelines advocating a lim-
metabolic deficiency in the elderly [1,2], has been consid-
ited battery of lab tests (complete blood count, electrolytes,
thyroid-stimulating hormone, calcium, glucose) for routine ered a “reversible cause of dementia,” but the fact that the
dementia diagnosis remain valid. This review is aimed at cognitive deficits of many patients with low Cbl and de-
summarizing the available evidence for the need to perform mentia do not improve after the Cbl levels are normalized
cobalamin (Cbl or B12), folate, and homocysteine serum has prompted questions about the relationship between the
determinations in patients whose cognitive function is under two and the relevance of Cbl in cognitive function.
investigation. The review and recommendations are cen- The association between Cbl and cognitive function
tered on older adults because this is the age group most has recently been analyzed in light of studies involving
affected by dementia, cognitive decline, and the metabolic total homocysteine (tHcy). Total homocysteine is a sulfur-
changes considered; the review takes a holistic approach to containing amino acid that accumulates in states of low
investigations and treatment of this population. Therefore, Cbl and/or folic acid [3,4]. Large epidemiologic studies
the recommendations might not be applicable to patients have found that elevated tHcy is a risk factor for the
with early-onset familiar Alzheimer’s disease or other types development of cerebrovascular disease, dementia of the
Alzheimer’s type (AD), and vascular dementia [5– 8],
*Corresponding author. Tel.: (613) 548-7222 x2229; Fax: (613)
although others have failed to confirm the association
544-4017. between AD and tHcy [9]. High levels of tHcy have been
E-mail address: garciaa@providencecare.ca associated with white matter abnormalities [10,11].
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.003
A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324 319

The inverse relationships between Cbl, folic acid, and tologic and neurologic abnormalities is found in only 41%
tHcy levels have prompted studies on the effects of B- of cases [18]. Analysis of cohort populations has shown that
vitamin treatment (Cbl, folic acid, and B6) on tHcy, demen- there is no significant relationship between clinical neuro-
tia, and cognitive decline in the elderly population. Since logic or hematologic signs or symptoms of Cbl deficiency
the implementation in 1998 of the Canadian program of and Cbl levels [19,20]; therefore, presence or absence of
folic acid fortification of grain products, there is virtually no those signs and symptoms should not be used to guide
folic acid deficiency in our population [12,13]; therefore, request for serum determinations of Cbl or assume Cbl
low Cbl remains the most important factor determining status. Moreover, a large percentage of older persons with
elevated tHcy in elderly Canadians. Cbl deficiency (determined either by low levels of Cbl or by
This article will summarize previous investigations on elevated MMA) have neither classic hematologic nor neu-
Cbl, tHcy, and cognitive function as well as the available rologic symptoms of Cbl deficit.
results of treatment studies, some of which are still ongoing.
The aim is to analyze the need to perform Cbl, folic acid,
4. Cobalamin and cognitive function
and/or tHcy determinations in the standard work-up for
dementia and cognitive decline in older adults in Canada. Low Cbl is more frequent among older persons with
dementia than among cognitively normal older adults, but
2. Methods this does not imply that low Cbl is the cause of dementia or
that the dementia is reversible with Cbl treatment. Although
MedLine database was searched for articles on Vitamin individual patients with low Cbl levels and dementia might
B12 and elderly, Vitamin B12 and cognition, Vitamin B12 benefit from Cbl treatment, there is no epidemiologic evi-
treatment, Homocysteine and elderly, Homocysteine and dence that low Cbl levels per se cause cognitive decline or
cognition, Homocysteine treatment, and Folic acid fortifi- increase the risk of dementia among older adults [21] or that
cation. Articles and presentations at various meetings were treatment with Cbl improves cognition in patients with
reviewed. Personal communications of unpublished results dementia.
and ongoing research are also noted when applicable. There are reports of dementia patients with low Cbl
The review includes a brief description of background whose dementia improved after normalization of Cbl levels
biologically relevant factors and changes during the last 6 [22,23]. Others have failed to find a significant effect of Cbl
years in Canada (folic acid fortification) and covers two treatment among dementia patients with low Cbl, although
outcomes: dementia (including AD, mixed and vascular a significant improvement was seen among persons with
dementia) and cognitive performance in older adults. cognitive impairment, not dementia [24].
There are no large randomized placebo-controlled trials
3. Cobalamin deficit in older adults assessing the effects of Cbl treatment on patients with de-
mentia and low Cbl, although two small, short-term trials
Human Cbl levels depend on intake and absorption from with negative results have been published [25]. A long-
animal-derived proteins. Cbl deficiency is an age-related term, appropriately designed randomized placebo control
condition. Epidemiologic studies have shown that the prev- trial of patients with dementia and low Cbl might be uneth-
alence of low Cbl serum concentrations rises with age ical at this stage because low Cbl can cause multiple pa-
[14,15]. With 200 pg/mL as a normal cutoff point, the thologies and normalization of Cbl levels is desirable for
incidence of low Cbl serum concentration among the elderly many reasons besides improving cognition.
population ranges from 7% to 16% [1,2,14]. It is unclear whether there is a subgroup of patients with
It is relevant to recognize that levels of Cbl might not be dementia and low Cbl who might benefit from Cbl treat-
a good marker of Cbl status/function and that methylma- ment, and what are the cognitive characteristics of these
lonic acid (MMA) appears to be the best indicator of Cbl patients. Nevertheless, because of the high prevalence of
function [15,16]. With elevated MMA as indicator of Cbl low Cbl as comorbidity among older adults, other available
deficiency in various elderly populations, the prevalence of guidelines for dementia recommend determinations of Cbl
Cbl deficiency can be much higher, up to 40% [17]. levels in the work-up for dementia and treatment if low Cbl
Cobalamin deficiency might cause or contribute to mul- is found [26].
tiple abnormalities in different tissues/organs, including Among cognitively normal older persons, results on the
megaloblastic anemia, gastrointestinal changes, peripheral relationship between Cbl and cognitive decline are available.
neuropathy, subacute combined degeneration, fatigue, de- Although we did not find that low Cbl affects cognitive decline
lirium, and depression. Traditionally, megaloblastic anemia in normal older persons [27], two trials have shown improve-
has served as a surrogate marker for the clinical investiga- ment of cognitive function with Cbl treatment. In a single blind
tion of Cbl deficiency. However, the effects of Cbl defi- trial among cognitively normal older subjects with low Cbl,
ciency at the hematologic and the neurologic levels are not 1-month placebo followed by 5 months of treatment with Cbl
parallel; in fact, the concurrent presentation of both hema- resulted in improved cognitive performance [28]. In a short-
320 A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324

term randomized controlled trial in cognitively normal to be a risk factor for dementia (both AD and vascular
women of all ages with unknown Cbl levels, B-vitamin dementia), with an increase in relative risk of 1.4 (confi-
treatment was found to slightly improve memory [29]. dence interval, 1.1 to 1.9) per 1 standard deviation increase
It appears that the effects of Cbl on cognitive function in the log-transformed tHcy level (tHcy levels are not nor-
might be related to levels of metabolites such as tHcy and mally distributed). Of note, the study period included pre-
MMA as a result of intracellular depletion of Cbl, folate, or fortification and postfortification years in the U.S. Similar
both. results were found in a recently published large cohort study
of older Italians with normal cognitive function at baseline
5. Homocysteine and aging and a follow-up of 4 years [31]. In a study by Luchsinger
et al [9] on Medicare recipients in the U.S., however, no
Total homocysteine levels also increase with age [4]. The association between tHcy and AD was found.
most important factors determining tHcy concentrations in Among the studies investigating the relation between
older age are Cbl and folate levels and renal function. tHcy levels and cognitive scores, results of the Rotterdam
Homocysteine is an amino acid predominantly derived from cohort showed an association between tHcy and decline in
the methionine pathway where deficits of folate or Cbl will scores of executive function and visual memory tests [32].
result in increases of tHcy levels. Other factors such as Another study in the U.S. involving 1,789 persons of Latino
smoking and dehydration also increase tHcy levels. Genetic origin older than the age of 60 years (SALSA study) found
markers of tHcy levels include methyltetrahydrofolate re- inverse relationships between levels of tHcy and scores in a
ductase (MTHFR) polymorphisms (677C¡T). The impact test of executive function and a test of global cognitive
of this genetic polymorphism in older age is still being function [33]. In another large cohort study including 1,241
debated. Total homocysteine is higher in men than in French people age 60 years or older, Dufouil et al [34]
women. As in Cbl deficiency, there are no reliable clinical found an odds ratio of 2.8 (confidence interval, 1.2 to 6.2) of
signs or symptoms that predict the presence of elevated cognitive decline at 4 years in persons with elevated tHcy
tHcy; therefore, there are no clinical indicators of elevated levels. In a cohort of normal elderly of Southeastern Ontario
tHcy to guide performing determinations [20]. we found a significant correlation between tHcy levels at
Prevalence of elevated tHcy among older adults varies baseline, tHcy changes over time, and decline in a test of
slightly according to country and folic acid fortification executive function [27]. Another cohort study from the U.S.
status. In Canada, levels of tHcy declined shortly after involving 321 men only also found an independent relation-
fortification was implemented in 1998, but they have risen ship between elevated tHcy and cognitive decline [35] in-
in subsequent years in spite of continuous increase in intra- cluding memory. Luchsinger et al [9], however, did not find
cellular folic acid levels (red blood cell folate). Between a relation between tHcy and decline in scores of memory,
2003 and 2004 the prevalence of older adults older than the whereas other studies have found significant relationship be-
age of 65 years in Southeastern Ontario with elevated tHcy tween tHcy and decline in scores of various cognitive tests
was 9%, similar to the prevalence found before fortification [36 –38]. Elevated tHcy levels also appear to affect frontal-
(10%) [13]. executive function decline in patients with strokes [39].
Age is an important determinant of the relationship be-
6. Homocysteine and cognition tween tHcy and cognitive decline. In a recently published
study from the Framingham cohort, Elias et al [40] found an
A full review of published data on the relationship be- association between tHcy and decline in multiple cognitive
tween tHcy and dementia and cognitive decline can be areas only in individuals younger than the age of 60. It
found in recently published reviews [30]. Only the most appears that the effects of homocysteine on cognition might
important findings of prospective cohort studies only are occur earlier than the target age for dementia screening,
summarized here. indicating that perhaps primary prevention treatment trials
The association between tHcy levels and cognitive func- should include younger subjects.
tion in older adults has been investigated by using two types
of outcomes: (1) dementia and (2) decline in cognitive test
7. Effects of B vitamins on tHcys
scores (without implying a diagnosis of dementia). The
studies investigating decline of cognitive scores as primary Total homocysteine levels can be lowered with B vita-
outcome used a variety of psychometric tests; therefore, mins (folic acid, Cbl, and B6).
there is no uniformity of results. Deficits of folic acid cause elevated tHcy. In 1998 for-
In the Framingham cohort study [7], 1,092 subjects cog- tification of grain products with folic acid became manda-
nitively normal at baseline were followed for 8 years. Di- tory in Canada. The aim of the fortification program was to
agnosis of dementia by Diagnostic and Statistical Manual reduce the incidence of neural tube defects in newborns.
[of Mental Disorders], Fourth Edition criteria was used as Canada, USA, and Chile are the only countries in the world
the primary outcome. Elevated tHcy at baseline was found with national folic acid fortification programs for more than
A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324 321

5 years. This fact might explain some differences encoun- in patients at high risk of dementia [46]. Final results of this
tered in studies from different countries and over time. trial are pending. Several large trials in different groups of
Shortly after fortification was initiated in Canada, tHcy patients include cognitive outcomes, but none has preven-
levels in older adults (and in the population at large) were tion or treatment of dementia as a primary outcome. These
significantly lower than levels before fortification [12]. include the VITATOPS in which B vitamins are adminis-
Analysis of tHcy levels in older adults in Southeastern tered for secondary prevention of cerebrovascular events
Ontario during the years thereafter, however, has shown that [47]. The HOPE study, recently published, showed negative
levels of tHcy have increased again to the same level seen results of B vitamins for secondary prevention of cardio-
in the prefortification period in spite of constantly increas- vascular events [48]. Although a Mini-Mental State Exam-
ing concentrations of red blood cell folate that are, at ination was conducted in a subgroup of the trial population,
present, above normal in 92% of older adults. By the end of those results have not been published yet. Stott et al [49]
2004, in spite of fortification, 16% of older adults in our recently published results of a 3-month randomized con-
population of Southeastern Ontario not taking oral Cbl sup- trolled trial (RCT) in patients with vascular disease. No
plements had elevated tHcy (3% had low Cbl levels), effects of B vitamins on cognitive function were seen. An
whereas only 2% of those taking oral Cbl supplements had efficacy study from the VISP trial has shown that a sub-
elevated tHcy (0% had low Cbl levels) [13]. None had low group of participants (those with Cbl levels between 250
red blood cell folate. It appears that in the presence of and 637 pmol/L) might benefit from B vitamins [50], but it
excess intracellular folic acid, a large percentage of older is unclear whether this beneficial effect would result in
adults not taking Cbl supplements in Canada have elevated cognitive improvement.
tHcy with normal Cbl levels. Current RCTs are analyzing the effects of B vitamins as
The largest majority of older adults absorb oral crystal- primary prevention of cognitive decline in older adults. One
line Cbl because it is not bound to protein, because the Cbl such trial [51] conducted in Holland (a country without folic
deficiency is most frequently due to atrophic gastritis. In acid fortification) showed a significant difference in cogni-
patients with low Cbl levels, oral Cbl decreases tHcy in a tive decline between the placebo and the active treatment
dose-dependent manner, with 1,000 ␮g/day being the most (folic acid), with worse outcomes in the placebo arm, at 3
effective dose to normalize tHcy [41]. It appears that ele- years. A short (4 months) RCT on primary prevention of
vated tHcy and low Cbl levels in older adults can be cor- cognitive decline in older adults failed to show significant
rected with oral Cbl supplements. Smaller doses can be effects of treatment on cognitive performance [52]. Other
effective in preventing elevated tHcy in persons with nor- trials have been completed and are pending publication
mal Cbl. Among community-dwelling older persons with (personal communication).
normal Cbl levels taking between 25 and 37.5 ␮g/day of
oral Cbl, we did not find any subjects with elevated tHcy,
whereas 13.2% of those with normal Cbl levels not taking 9. Conclusions
any Cbl oral supplements had elevated tHcy [42].
The role of vitamin B6 in tHcy levels has not been fully 1. Low Cbl is a frequent comorbidity among cogni-
investigated, but it appears that B6 alone does not signifi- tively normal older adults and older patients with
cantly decrease tHcy levels [43]. dementia.
2. Low Cbl can be corrected with either oral or paren-
teral Cbl.
8. Effects of lowering tHcys treatment on cognition
3. The direct relationship between low Cbl and cogni-
and dementia
tive function remains unclear.
There are no published large, long-term randomized 4. MMA is a better indicator of Cbl function than Cbl
placebo-controlled trials of reduction of tHcy levels with levels.
B vitamins and cognition as primary outcome in patients 5. Treatment with Cbl in patients with low Cbl and
with elevated tHcy and dementia or in patients with dementia has yielded variable results.
dementia regardless of their tHcy level. 6. There are no large randomized placebo-controlled
In a recently published case series of patients with dementia trials of Cbl treatment in patients with low Cbl and
and elevated tHcy levels, the combination treatment of B dementia, and because of the wide range of abnor-
vitamins and N-acetylcysteine (antioxidant) resulted in cogni- malities found in Cbl deficiency states, such trials
tive improvement in 5 of 7 patients [44]. Successful improve- might be unethical.
ment of executive cognitive function with homocysteine- 7. Treatment of low Cbl in older adults with normal
lowering treatment in an individual patient has been reported cognitive function might improve cognitive scores.
[45]. 8. Elevated tHcy is frequent among older adults, even
The VITAL trial is aimed at analyzing the effects of oral in the presence of elevated intracellular folic acid
high dose B vitamins and aspirin on cognitive performance levels (red blood cell folate).
322 A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324

9. Elevated tHcy is a risk factor for dementia. baseline and a battery of 8 cognitive tests as end-points. The
10. Elevated tHcy seems to affect cognitive decline in treatment arm consisted of daily high dose of Folic acid,
older adults. B12 and B6. The authors did not find any improvement, but
11. Elevated tHcy can be corrected with B vitamins/Cbl decline in one test (the Part B Reitan Trails Making Test) in
even in the presence of elevated red blood cell the active treatment arm compared to placebo, raisings
folate. questions that remain unexplained [54]. The second [55]
12. There are case reports of successful treatment of publishes the results obtained in a 3-years RCT with high
elevated tHcy in patients with dementia. dose folic acid in 818 cognitively normal subjects ages 50 to
13. There are no published RCTs of B-vitamin treatment 70 with elevated tHcy. The authors found a significant
in patients with dementia of the Alzheimer’s type improvement in the composite scores of memory, informa-
and elevated tHcy. Such trials are ongoing. tion processing speed and sensorimotor speed in the active
14. Short-term RCTs in patients with vascular dementia treatment group.
showed no effects of B vitamins on cognition.
15. Results from primary prevention RCTs (in cogni-
Author Disclosures
tively normal adults with or without elevated tHcy)
are inconclusive. Larger studies are needed. Angeles Garcia has received support from Janssen-Ortho
16. Red blood cell folate/total folate levels are elevated (Consultant, Advisor), Pfizer (Consultant, Advisor), Novartis
in the largest majority of older adults after imple- (Consultant, Advisor), and Lundbeck (Consultant, Advisor).
mentation of the folic acid fortification program.
Acknowledgments
10. Recommendations reaching consensus at the Third
This work was supported by The Ontario Mental Health
Canadian Consensus Conference on Diagnosis and
Foundation and Queen’s University.
Treatment of Dementia

1. It is recommended that serum Cbl levels be deter- References


mined in all older adults suspected of dementia or
cognitive decline (Grade B, Level 2). [1] Lindenbaum J, Rosenberg IH, Wilson PWF, Stabler SP, Allen RH.
Prevalence of cobalamin deficiency in the Framingham elderly pop-
2. Older adults found to have low Cbl levels should be ulation. Am J Clin Nutr 1994;60:2–11.
treated with Cbl (either oral or parenteral forms) be- [2] Joosten E, van den Berg A, Riezler R, Naurath HJ, Lindenbaum J,
cause of potential improvement of cognitive function Stabler SP, Allen RH. Metabolic evidence that deficiencies of B12
and the deleterious effects of low Cbl levels on mul- (cobalamin), folate, and vitamin B6 occur commonly in elderly peo-
tiple organ systems, besides the effects on cognition ple. Am J Clin Nutr 1993;58:468 –76.
[3] Stabler SP, Marcell PD, Podell ER, Allen RH, Savage DG, Linder-
(Grade B, Level 2). baum J. Elevations of total homocysteine in the serum of patients with
3. There is currently insufficient evidence to support the cobalamin or folate deficiency detected by capillary gas chromatog-
need for serum tHcy levels to be determined in older raphy-mass spectrometry. J Clin Invest 1988;81:466 –74.
adults suspected of dementia or cognitive decline [4] Selhub J, Jaques PF, Rosenberg IH, Rogers G, Bowman BA, Gunter
(Grade C, Level 3). EW, et al. Serum total homocysteine concentrations in the third
national health and nutrition examination survey (1991–1994): pop-
4. There is currently insufficient evidence that treatment ulation reference ranges and contribution of vitamin status to high
of elevated serum tHcy levels affects cognition serum concentrations. Ann Intern Med 1999;131:331–9.
(Grade C, Level 3). [5] The Homocysteine Studies Collaboration. Homocysteine and risk of
5. Determination of serum folic acid or red blood cell ischemic heart disease and stroke: a meta-analysis. JAMA 2002;288:
folate in older adults in Canada is optional and might 2015–22.
[6] Boysen G, Brander T, Christensen H, Gideon R, Truelsen T. Homo-
be reserved for patients with celiac disease, inade- cysteine and risk of recurrent stroke. Stroke 2003;34:1258 – 61.
quate diets, or other conditions that prevent them [7] Seshadri S, Beiser A, Selhub J, et al. Plasma homocysteine as a risk
from ingesting grain products (Grade E, Level 2). factor for dementia and Alzheimer’s disease. N Engl J Med 2002;
346:476 – 83.
[8] Wald DS, Law M, Morris JK. Homocysteine and cardiovascular
11. Addendum disease: evidence on causality from a meta-analysis. BMJ 2002;325:
1202– 8.
Since the submission of this paper, two other papers have [9] Luchsinger JA, Tang MX, Shea S, Miller J, Green R, Mayeux R.
been published that are relevant to the topic discussed. Plasma homocysteine levels and risk of Alzheimer disease. Neurol-
Although their conclusions do not change ours, or the rec- ogy 2004;62:1972– 6.
[10] Wright CB, Paik MC, Brown TR, Stabler SP, Allen RH, Sacco RL,
ommendations, it is relevant to note them. The first [53] is et al. Total homocysteine is associated with white matter hyperinten-
the results of a 2-year randomized placebo controlled trial sity volume: the Northern Manhattan Study. Stroke 2005;36:1207–
(RCT) on cognitively normal elderly with elevated tHcy at 11.
A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324 323

[11] Vermeer SE, Den Heijer T, Koudstaal PJ, Oudkerk M, Hofman A, [32] Prins ND, den Heijer T, Hofman A, Koudstaal PJ, Jolles J, Clarke R,
Breteler MMB. Rotterdam Scan Study: incidence and risk factors Breteler MMB. Homocysteine and cognitive function in the elderly:
of silent brain infarcts in the population-based Rotterdam Scan the Rotterdam scan study. Neurology 2002;59:1375– 80.
Study. Stroke 2003;34:392– 6. [33] Miller JW, Green R, Ramos MI, Allen LH, Mungas DM, Jagust WJ,
[12] Ray JG, Vermeulen MJ, Boss SC, Cole DEC. Declining rate of folate et al. Homocysteine and cognitive function in the Sacramento Area
insufficiency among adults following increased folic acid food forti- Latino Study on Aging. Am J Clin Nutr 2003;78:441–7.
fication in Canada. Can J Public Health 2002;93:249 –53. [34] Dufouil C, Alperovitch A, Ducros V, Tzourio C. Homocysteine,
[13] Day A, Zanibbi K, Garcia A. Red blood cell folate, homocysteine, white matter hyperintensities, and cognition in healthy elderly people.
cobalamin and methylmalonic acid through the folic acid fortification Ann Neurol 2003;53:214 –21.
years. Hematol Rep 2005;1:58. [35] Tucker KL, Qiao N, Scott T, Rosenberg I, Spiro A 3rd. High homo-
[14] Pennypacker LC, Allen RH, Kelly JP, Matthews LM, Grigsby J, Kaye cysteine and low B vitamins predict cognitive decline in aging men:
K, et al. High prevalence of cobalamin deficiency in elderly outpa- the Veterans Affairs Normative Aging Study. Am J Clin Nutr 2005;
tients. J Am Geriatr Soc 1994;40:1197–204. 82:627–35.
[15] Clarke R, Evans JG, Schneede J, Nexo E, Bates C, Fletcher A, et al. [36] Kalmijn S, Launer LJ, Lindemans J, Michiel B, Hofman A, Breteler
Vitamin B12 and folate deficiency in later life. Age Ageing 2004;33: MM. Total homocysteine and cognitive decline in a community-
34 – 41. based sample of elderly subjects: the Rotterdam Study. Am J Epide-
[16] Allen RH, Stabler SP, Savage DG, Lindenbaum J. Diagnosis of miol 1999;150:283–9.
cobalamin deficiency I: usefulness of serum methylmalonic acid and [37] McCaddon A, Hudson P, Davies G, Hughes A, Williams JH, Wilkin-
total homocysteine concentrations. Am J Hematol 1990;34:90 – 8. son C. Homocysteine and cognitive decline in healthy elderly. De-
[17] Andres E, Loukili NH, Noel E, Kaltenbach G, Abdelgheni MB, Perrin ment Geriatr Cogn Disord 2001;12:309 –13.
AE, et al. Vitamin B12 (cobalamin) deficiency in elderly patients. [38] Teunissen CE, Blom AHJ, Van Boxtel PJ, Bosma H, de Bruijn C,
CMAJ 2004;171:251–9. Jolles J, et al. Homocysteine: a marker for cognitive performance? a
[18] Healton EB, Savage DG, Brust JCM, Garrett TJ, Lindenbaum J. Neu- longitudinal follow-up study. J Nutr Health Aging 2003;7:153–9.
rologic aspects of cobalamin deficiency. Medicine 1991;70:229 – 44. [39] Sachdev PS, Valenzuela MJ, Brodaty H, Wang XL, Looi J, Lorentz
[19] Bjorkegren K, Svardsudd K. Reported symptoms and clinical findings L, et al. Homocysteine as a risk factor for cognitive impairment in
in relation to serum cobalamin, folate, methylmalonic acid and total
stroke patients. Dement Geriatr Cogn Disord 2003;15:155– 62.
homocysteine among elderly Swedes: a population-based study. J In-
[40] Elias MF, Sullivan LM, D’Agostino RB, Elias PK, Jacques PF,
tern Med 2003;254:343–52.
Selhub J, et al. Homocysteine and cognitive performance in the
[20] Garcia A, Haron Y. Predictive values of signs and symptoms in
Framingham offspring study: age is important. Am J Epidemiol
cobalamin deficiency. Gerontology 2002;42:34.
2005;162:644 –53.
[21] Crystal HA, Ortof E, Frishman WH, Gruber A, Hershman D, Aronson
[41] Eussen SPM, de Groot LCPG, Clarke R, Schneede J, Ueland PM,
M. Serum vitamin B12 levels and incidence of dementia in a healthy
Hoefnagels WHL, et al. Oral cyanocobalamin supplementation in
elderly population: a report from the Bronx Longitudinal Aging
older people with vitamin B12 deficiency. Arch Intern Med 2005;
Study. J Am Geriatr Soc 1994;42:933– 6.
165:1167–72.
[22] Cunha UG, Rocha FL, Peixoto JM, Motta MF, Barbosa MT. Vitamin
[42] Garcia A, Evans L, Paris-Pombo A, Freedman, M. Is low dose oral
B12 deficiency and dementia. Int Psychogeriatr 1995;7:85– 8.
cobalamin enough to normalize cobalamin levels and function in the
[23] Abyad A. Prevalence of vitamin B12 deficiency among demented
elderly? J Am Geriatr Soc 2002:50:1401– 4.
patients and cognitive recovery with cobalamin replacement. J Nutr
[43] Malouf R, Grimley Evans J. The effect of vitamin B6 on cognition.
Health Aging 2002;6:254 – 60.
[24] Eastley R, Wilcock GK, Bucks RS. Vitamin B12 deficiency in de- Cochrane Database Syst Rev 2003.CD004393.
mentia and cognitive impairment: the effects of treatment on neuro- [44] McCaddon A. Homocysteine and cognitive impairment: a case series
psychological function. Int J Geriatr Psychiatry 2000;15:226 –33. in a general practice setting. Nutr J 2006;5:6 –13.
[25] Malouf R, Areosa Sastre A. Vitamin B12 for cognition. Cochrane [45] Boxer AL, Kramer JH, Johnston K, Goldman J, Finley R, Miller
Database Syst Rev 2003.CD004326. BL. Executive dysfunction in hyperhomocystinemia responds to
[26] Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom J, homocysteine-lowering treatment. Neurology 2005;64:1431– 4.
Relkin N, et al. Practice parameter: diagnosis of dementia (an evidence- [46] VITAL collaborative group: effects of vitamins and aspirin on mark-
based review): Report of the Quality Standards Subcommittee of the ers of platelet activation, oxidative stress and homocysteine in people
American Academy of Neurology. Neurology 2001;56:1143–53. at high risk for dementia. J Intern Med 2003;254:67–75.
[27] Garcia A, Haron Y, Pulman K, Hua L, Freedman M. Increases in [47] VITATOPS Trial Study Group. The VITATOPS (Vitamins to Pre-
homocysteine are related to worsening of Stroop scores in normal vent Stroke) Trial: rationale and design of an international, large,
elderly subjects: a prospective follow-up study. J Gerontol A Biol Sci simple, randomized trial of homocysteine-lowering multivitamin
Med Sci 2004;59:1323–7. therapy in patients with recent transient ischaemic attack or stroke.
[28] van Asselt DZ, Pasman JW, van Lier HJ, Vingerhoets DM, Poels Cerebrovasc Dis 2002;13:120 – 6.
PJ, Kuin Y, et al. Cobalamin supplementation improves cognitive [48] Lonn E, Yusuf S, Arnold MJ, Sheridan P, Pogue J, Micks M, et al.
and cerebral function in older, cobalamin-deficient persons. J Heart Outcomes Prevention Evaluation (HOPE) 2 investigators: ho-
Gerontol A Biol Sci Med Sci 2001;56:775–9. mocysteine lowering with folic acid and B vitamins in vascular
[29] Bryan J, Calvaresi E, Hughes D. Short-term folate, vitamin B-12 or disease. N Engl J Med 2006;354:1567–77.
vitamin B-6 supplementation slightly affects memory performance [49] Stott DJ, MacIntosh G, Lowe GD, Rumley A, McMahon AD, Lang-
but not mood in women of various ages. J Nutr 2002;132:1345–56. horne P, et al. Randomized controlled trial of homocysteine-lowering
[30] Garcia A, Zanibbi K. Homocysteine and cognitive function in elderly vitamin treatment in elderly patients with vascular disease. Am J Clin
people. CMAJ 2004;171:897–904. Nutr 2005;82:1320 – 6.
[31] Ravaglia G, Forti P, Maioli F, Martelli M, Servadei L, Brunetti N, et [50] Spence JD, Bang H, Chambless LE, Stampher MJ. Vitamin Interven-
al. Homocysteine and folate as risk factors for dementia and Alzhei- tion For Stroke Prevention trial: an efficacy analysis. Stroke 2005;
mer disease. Am J Clin Nutr 2005;82:636 – 43. 36:2404 –9.
324 A. Garcia / Alzheimer’s & Dementia 3 (2007) 318 –324

[51] Durga J, van Boxtel MPJ, Schouten EG, Kok FJ, Jolles J, Katan MB, [53] McMahon JA, Green TJ, Skeaff CM, Knight RG, Mann JI, Williams
et al. The effect of 3-year folic acid supplementation on cognitive SM. A controlled trial of homocysteine lowering and cognitive per-
function, a randomized controlled trial. Hematol Rep 2005;1:1. formance. N Engl J Med 2006;354:2764 –72.
[52] Lewerin C, Matousek M, Steen G, Johansson B, Steen B, Nilsson- [54] Clarke R. Vitamin B12, folic acid, and the prevention of dementia.
Ehle H. Significant correlations of plasma homocysteine and serum N Engl J Med. 2006;354:2817–9.
methylmalonic acid with movement and cognitive performance in [55] Durga J, van Boxtel MPJ, Schouten EG, Kok FJ, Jolles J, Katan MB,
elderly subjects but no improvement from short-term vitamin therapy: et al. Effect of 3-year folic acid supplementation on cognitive func-
a placebo-controlled randomized study. Am J Clin Nutr 2005;81: tion in older adults in the FACIT trial: a randomised, double blind,
1155– 62. controlled trial. Lancet 2007;369:208 –16.
Alzheimer’s & Dementia 3 (2007) 325–332

The role of biologic markers in the diagnosis of Alzheimer’s disease


Hyman M. Schipper*
Centre for Neurotranslational Research and Bloomfield Centre for Research in Aging, Lady Davis Institute for Medical Research, Sir Mortimer B.
Davis-Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University,
Montreal, Quebec, Canada

1. Introduction role to play in the management of patients with the far more
common sporadic form of the illness. The presence of the
The objective of this report is to establish consensus
apolipoprotein E (APOE) e4 allele, a genetic modifier, is a
criteria for the use of biologic markers in the diagnosis of
strong risk factor for the development of sporadic AD and
Alzheimer’s disease (AD). To address this issue, we will
predicts earlier onset of clinical dementia in AD patients.
first provide background information concerning the signif-
However, the presence of the e4 allele does not confer AD,
icance of biologic AD markers, differences between genetic
markers, genetic modifiers, and biologic markers, criteria and its absence does not exclude this disorder. Thus, testing
for an ideal biologic marker, strengths and weaknesses of for the e4 allele cannot be used as a diagnostic marker of
biologic markers currently available, and promising future sporadic AD. Unlike the genetic markers and modifiers, true
research developments in AD biomarker discovery. Specific biologic markers of AD are “state” indicators that inform of
recommendations to primary care physicians and specialists the presence or absence of AD at the time of measurement
will follow. and are thus appropriate for use as diagnostic modalities for
sporadic (and possibly familial) forms of this condition.

2. What is a biologic marker?


A biologic marker of disease is a measurable change in 3. How would a biologic marker assist in the
the physical constitution of an organism that indicates the management of AD?
presence of the disease. Current biologic indices under in-
vestigation for the early diagnosis of AD include chemical The advent of a biologic marker that reliably indicates
markers detected in body fluids and brain volume or activity the presence of AD and differentiates the latter from other
measurements derived from neuroimaging modalities such dementing conditions would represent a major achievement
as magnetic resonance imaging (MRI) or positron emission in the management of this common neurodegenerative dis-
tomography (PET) of relevant brain regions. Neuroimaging order. Specific benefits accruing from successful implemen-
modalities are expensive, labor-intensive, and not univer- tation of such a marker in clinical practice would include
sally available, prompting a search for reliable chemical (1) more rapid and accurate diagnosis of sporadic AD;
biomarkers and other practical neurodiagnostic modalities. (2) ability to diagnose AD in patients with major affective
Chemical markers of AD can be conceptualized within three disorders, clouded sensorium, depressed level of conscious-
general categories: (1) genetic markers, (2) genetic modifiers, ness, and other clinical conditions that often preclude es-
and (3) biologic markers. Genetic markers such as mutant tablishment of a dementia diagnosis by conventional means;
forms of amyloid precursor protein (APP), presenilin-1, and (3) possible monitoring of the progression and severity of
presenilin-2 are excellent for predicting disease in rare kin- AD and responses to therapeutic interventions; (4) potential
dreds with familial AD but are not useful in tracking disease prognostication of mild cognitive impairment (MCI) sub-
progression or efficacy of therapeutic intervention in these jects at highest risk for early conversion to incipient AD;
subjects. Moreover, these genetic markers have little or no (5) assembly of homogeneous patient cohorts for therapeu-
tic clinical trials; and (6) considerable cost savings related to
*Corresponding author. Tel.: 514-340-8260; Fax: 514-340-8925. curtailment of ancillary biochemical and imaging modalities
E-mail address: hyman.schipper@mcgill.ca currently used to exclude other causes of dementia.
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.015
326 H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332

4. What are the characteristics of an ideal biologic remains considerably lower when differentiating AD from
marker for use in AD? control groups with other causes of dementia. For example,
in a multi-center study involving 150 AD, 100 normal
The quest for the development of reliable AD biomarkers elderly controls (NEC), and 79 cases of non-AD dementia
worldwide is guided, in large measure, by principles set [10], the specificity of CSF A␤1-42 in differentiating AD
forth in a Consensus Report on Molecular and Biochemical from NEC and patients with other dementias was 81% and
Markers of AD sponsored by the Alzheimer’s Association 59%, respectively. This is because CSF A␤1-42 might be
(US) and the National Institute on Aging (Neurobiology of decreased in other degenerative and nondegenerative CNS
Aging 1998;19:107– 67). In this landmark report it was disorders including Lewy body dementia (LBD) [11–13],
suggested an ideal biologic marker of AD fulfill the follow- Creutzfeldt-Jakob disease (CJD) [12,14], multisystem atro-
ing criteria [1]: phy [15], and amyotrophic lateral sclerosis (ALS) [16]. The
(1) Reflect a fundamental aspect of central nervous sys- advent of mass spectrometry techniques has led to the recent
tem (CNS) pathophysiology in AD (plausibility) identification of novel amyloid peptide fragments in AD
(2) Indicate the actual presence of AD and not merely CSF that might prove to be of diagnostic significance [17].
increased risk
(3) Exhibit high sensitivity and specificity (in the range 5.2. CSF total tau
of 80% or better for each)
Increased concentrations of total tau protein (t-tau) have
(4) Be efficacious in early or pre-clinical AD (e.g. MCI)
been consistently observed in AD CSF. A meta-analysis
(5) Monitor disease severity or rate of progression
performed by Sunderland et al [9] involving 35 studies of
(6) Indicate efficacy of therapeutic intervention
CSF tau as an AD diagnostic disclosed an effect size of 1.31
(7) Be noninvasive, inexpensive, and readily available.
(95% CI, 1.23 to 1.39). In 2003 Blennow and Hampel [4]
In the aftermath of this consensus report, it was also deemed reviewed CSF t-tau data from 41 studies (more than 4,000
desirable that (8) the efficacy of the putative biomarker is AD and control subjects) that used either of the two most
corroborated by at least one other independent laboratory, and commonly available t-tau assays, the Innogenetics or
that its accuracy (Criterion 3) is demonstrated in discriminating Athena enzyme-linked immunosorbent assay (Athena Neu-
AD not only from cognitively healthy controls but from pa- rosciences, Worcester, MA). Overall, the sensitivity and
tients with various non-AD dementias [2]. specificity of these assays for the diagnosis of AD were in
the range of 80% and 90%, respectively (similar to those of
5. What biologic markers are currently available for CSF A␤1-42). Although CSF t-tau distinguished AD patients
the diagnosis of AD? from neurologically healthy controls with high accuracy,
the marker performs considerably less well in discriminat-
5.1. CSF A␤1-42 ing AD from other dementias, with reported specificities of
57% for suspected non-AD dementias [10] and 69% for
Increased plasma ␤-amyloid1-42 (A␤1-42) levels have
autopsy-proven cases [12]. High levels of CSF t-tau might
been documented in several kindreds with familial AD [3],
also predict progression of cognitive decline in MCI, espe-
but these families represent a very small subset of the total
cially in patients without extensive periventricular white
AD population. Measurements of total A␤ peptide, A␤1-40,
matter lesions [18]. CSF t-tau appears to be a general
or soluble APP␣/␤ concentrations in cerebrospinal fluid
marker for neuronal destruction in a wide range of degen-
(CSF) or blood have not proved useful in the diagnosis of
erative and nondegenerative CNS conditions and is there-
sporadic AD [4 – 8]. Amyloid fragments are plausible AD
fore fairly nonspecific for AD. In addition to AD, elevated
biomarkers because they directly reflect a known patho-
CSF t-tau levels might occur in vascular dementia [19],
physiologic process in AD brain (senile plaque formation).
frontotemporal dementia (FTD) [20], CJD, and (transiently)
Numerous laboratories have demonstrated abnormally low
in acute ischemic stroke [21]. In a recent study, 34% of
levels of the amyloid peptide fragment A␤1-42 in the CSF of
subjects with FTD exhibited significantly low levels of CSF
sporadic AD patients and individuals with MCI [5]. A
tau, a finding not observed in the AD cohort [22]. CSF t-tau
meta-analysis performed by Sunderland et al [9] involving
values in vascular dementia [23] and LBD [11] are reportedly
18 studies of CSF A␤1-42 as an AD diagnostic disclosed an
intermediate between those of NEC and subjects with AD.
effect size of 1.56 (95% confidence interval [CI]: 1.43
to 1.69). In 2003 Blennow and Hampel [4] reviewed CSF 5.3. CSF phospho-tau
A␤1-42 data from 13 studies (⬃600 AD and 450 control
subjects) that used the Innogenetics enzyme-linked immu- By using antibodies against various phosphorylated
nosorbent assay (Gent, Belgium). They calculated an over- epitopes of tau (p-tau), a number of laboratories have doc-
all sensitivity and specificity of 80% and 90%, respectively, umented significant increases in levels of hyperphosphory-
for differentiating AD from healthy cognitive aging on the lated tau in AD CSF relative to NEC and, more importantly,
basis of this assay. However, the specificity of this test patients with other dementing and nondementing neurologic
H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332 327

disorders [4,24,25]. Thus, in contrast to t-tau, augmented biomarker studies are deemed more valuable than
CSF p-tau levels might discriminate AD from FTD [26,27], retrospective analyses, the former are less likely to
vascular dementia [28], LBD [29], Parkinson’s disease (PD) entail neuropathologic diagnoses [46].
[30], ALS, acute stroke [31], major depression [30], and (2) CSF examination by lumbar puncture (spinal tap) is
schizophrenia [32]. CSF levels of threonine 181 p-tau have relatively invasive and therefore generally not consid-
recently been reported to be elevated in sporadic and variant ered suitable for mass screening of elderly individuals
CJD [33], despite an earlier report to the contrary [34]. with AD risk factors or mild memory impairment. Yet,
These phospho-tau isoforms are plausible AD biomarkers in a recent study of 342 AD, MCI, and NEC subjects
because they directly reflect a known pathophysiologic pro- who underwent 428 research lumbar punctures, there
cess in AD brain (neurofibrillary tangle formation). They was a very low rate of adverse effects (eg, post–lumbar
are relatively early markers of the disease inasmuch as their puncture headaches in 0.93%), and the procedure
concentrations in CSF are significantly elevated in incipient was well-tolerated (low anxiety and pain ratings in
AD [35] and MCI [36,37]. On the other hand, CSF p-tau visual analog scales) by all patient groups [47].
might not be useful in monitoring disease progression in AD (3) The immunoassay techniques used to measure CSF
(possibly as a result of dilutional factors) unless combined A␤1-42 and tau are not trivial, and CSF specimens
with MRI measurements of hippocampal atrophy in these usually must be shipped to specialized laboratories
patients [38]. adept at performing these immunoassays in a reliable
and reproducible fashion.
5.4. CSF A␤1-42 and p-tau combined
(4) The cost per sample of the combined CSF A␤1-42
When measured together, CSF A␤1-42 and phospho-tau and tau assays at Athena Neurosciences is US$1130
exhibit sensitivities for detecting AD when present and to insurance companies and $765 to MDs. The cost
specificities for excluding other dementing disorders in the of an AD biomarker might not be prohibitive if
respectable range of 80% to 90% [39]. At a prevalence rate it obviates the need for additional testing (eg,
of 45%, the positive and negative predictive values of the neuroimaging).
combined test are 90% and 95%, respectively [5]. In addi-
5.5. Urine AD7C-neuronal thread protein
tion, these markers identify AD in early stages (eg, MCI),
reflect the underlying pathophysiology of the disease (see Measurement of elevated AD7C–neuronal thread protein
above), and are relatively inexpensive. It has been argued (NTP) in urine, an assay commercialized by Nymox Cor-
that CSF A␤1-42 reflects the stage of the disease (with levels poration (Montreal, Quebec, Canada), purportedly differen-
declining progressively as a function of disease duration), tiates AD from control subjects with high sensitivity and
whereas t-tau and p-tau reflect the intensity of the disease specificity. The protein induces neuritic sprouting and apo-
process (with higher CSF levels connoting more rapid pro- ptosis in transfected neuronal cells [48] and might nonspe-
gression) [40]. Also, the degree of CSF A␤1-42 suppression cifically reflect AD pathogenesis. However, measurement of
and tau elevation might correlate with the APOE e4 allele urinary AD7C levels requires extensive protein purification
burden [41], although whether and to what extent the ge- before customized immunoassay, and the NYMOX findings
netic data should be considered in the analysis of the CSF await confirmation from independent laboratories. In July
biomarker levels remain unclear. Importantly, elevation of 2005 the Food & Drug Administration denied approval of
CSF tau and suppressed CSF A␤1-42 in MCI might prog- the urinary AD7C-NTP measurement as an AD neurodi-
nosticate for incipient conversion to AD with sensitivities/ agnostic, citing excessive overlap of values between di-
specificities in the range of 83% to 90% [42,43]. This agnostic groups, lack of autopsy confirmation, and ques-
marker combination showed efficacy in identifying “non- tionable clinical utility (http://www.fdaadvisorycommittee.
progressors” in “mixed” (amnestic and non-amnestic) MCI com/FDC/AdvisoryCommittee/Committees/Device⫹Panels/
during a 3-year median follow-up period [44] and might be 071505_Alzheimertest/071505_AlzheimAlertR.htm). The test
helpful in discriminating pre-AD MCI from anxiety/depression remains available to physicians through Nymox’s Clinical
in the elderly [45]. We are aware of no studies implicating Laboratories Improvement Act– certified laboratory in New
CSF A␤1-42 and tau measurements as useful indices of Jersey.
therapeutic efficacy in AD.
5.4.1. Caveats 6. Are there any promising AD biomarkers currently
(1) In the vast majority of these AD biomarker studies, under development?
receiver operating characteristic curves (plotting the 6.1. Serum p97 protein (melanotransferrin)
relationship between sensitivities and specificities)
were determined solely on the basis of clinical diag- At the time of the Second Canadian Consensus Confer-
noses without autopsy confirmation, thereby limiting ence on Diagnosis and Treatment of Dementia [49], there
the validity of the data. Although prospective AD was considerable enthusiasm regarding the potential use of
328 H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332

elevated serum levels of the iron-binding protein p97 (mela- including Huntington disease, CJD, and multiple
notransferrin) for the diagnosis of sporadic AD [50,51]. sclerosis [73].
Unfortunately, the interest has dissipated on recognition that (3) A clinical test based on CSF or urinary isoprostane
the immune-based measurements of this circulating protein measurements might be difficult to standardize and
were probably artifactual [52]. make readily available because quantification of these
lipid products requires complex gas chromatography/
6.2. Platelet APP ratios
mass spectrometry techniques using internal standards
Blood platelets contain many of the same amyloid- that are often difficult to procure.
processing enzymes present in brain and generate small
amounts of A␤ from APP. Several laboratories have docu- 6.4. Plasma heme oxygenase-1 suppressor factor
mented reductions in the 130-kd APP isoform:110-kd APP In 2000 our laboratory reported that suppressed levels of
isoform ratios in AD platelets relative to NEC and non-AD heme oxygenase-1 (HO-1) mRNA (measured by Northern
dementias [53–55]. In a study of platelet APP ratios involv- blot) in peripheral blood mononuclear cells distinguish spo-
ing 85 probable AD patients and 95 healthy and neurologic radic AD from NEC and patients with nonAD dementias,
controls, Padovani et al [56] successfully diagnosed AD with a sensitivity and specificity of 88% and 75%, respec-
with 88.2% sensitivity and 89.4% specificity. Additional tively [74]. These findings have been replicated by an in-
studies provided evidence that low APP 130:110 ratios dependent laboratory with a sensitive reverse transcriptase
correlate with the severity of AD dementia [57,58] and polymerase chain reaction technique [75]. However, HO-1
predict conversion to AD in subjects with MCI [59,60]. mRNA procured from AD blood cells was subsequently
Intriguingly, there are at least three reports of APP ratio shown to display chemical instability [76], confounding its
“normalization” in AD patients treated with cholinesterase development as a clinical neurodiagnostic. More recently, a
inhibitors, implicating platelet APP as a possible reporter plasma heme oxygenase-1 suppressor (HOS) factor was
molecule of effective therapeutic intervention (Section 4, identified, characterized, and observed to be significantly
Criterion #6) [61– 63]. On the negative side, platelet APP elevated in patients with early AD relative to subjects with
isoform ratios are determined by analyses of band densities PD and NEC. MCI subjects exhibited plasma HOS levels
obtained on Western blotting, a labor-intensive and rela- that were intermediate between NEC and AD values [77].
tively expensive technique that resists accurate protein Unlike HO-1 mRNA, the HOS protein (␣1-antitrypsin) is
quantification, inter-laboratory standardization, and high- chemically stable and is undergoing further testing as a
throughput processing. putative peripheral AD biomarker. HO-1 and the HOS pro-
6.3. CSF and urine F2-isoprostanes tein reflect AD pathophysiology in so far as HO-1 protein is
augmented and co-localizes to senile plaques and neurofi-
Oxidative molecular damage has been implicated in brillary tangles in AD-affected neural tissues [78].
the pathophysiology of numerous aging-related neurode-
generative disorders [64,65]. Several indices of oxidative 6.5. Biospectroscopy
substrate damage, such as 8-hydroxy-2-deoxyguanosine and
4-hydroxynonenal, might be augmented in AD CSF relative Biospectroscopy is an optical-based analytic technology
to control values. However, many oxidative metabolites are that provides a simultaneous and integrated measure of all
either nonspecific for AD pathology or difficult to measure metabolic activities taking place in a biologic sample or
because they are present in trace amounts, chemically un- field and then uses the accruing spectral data to create a
stable, or require elaborate technology [66,67]. Elevated profile of molecular biomarkers. Advantages of biospectros-
concentrations of F2-isoprostanes, specific indices of lipid copy include simplicity of use, short analysis time, low-cost
peroxidation, have been reported in the CSF, plasma, and/or instrumentation, simultaneous ascertainment of multiple
urine of patients with sporadic AD [68,69] and MCI [38,70] metabolites/biomarkers in a single specimen, and opportu-
relative to control values. In a recent (but relatively small) nity for noninvasive, real-time in vivo monitoring both
study, inclusion of CSF isoprostane data facilitated dis- remotely and at bedside. In preliminary studies, biospectro-
crimination of probable AD from NEC and non-AD de- scopic analyses in the near-infrared performed on small
mentia above and beyond that conferred by measure- volumes of human plasma revealed metabolomic signatures
ments of A␤1-42/tau alone [71]. that distinguished early sporadic AD from PD and NEC
with high sensitivity and specificity (Schipper HM and
6.3.1. Caveats Burns D, manuscript in preparation).
(1) A laboratory at the University of Washington was
6.6. Other putative biomarkers
able to reproduce the data of Pratico et al [70] in
CSF, but not in blood or urine [72]. An extensive array of endogenous substances detected in
(2) F2-isoprostane levels might be elevated in the CSF of CSF and/or blood have been touted as possible AD biomar-
other potentially dementing neurologic disorders, kers. The latter include various acute phase reactants, cyto-
H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332 329

kines, oxidative stress markers, advanced glycation end (5) Platelet APP isoform ratios, plasma and urinary F2-
products, ubiquitin-proteasomal constituents, trophic fac- isoprostane levels, systemic alterations in HOS/
tors, complement components, glial and synaptic proteins, HO-1 profiles, and blood biospectrosopy might ful-
cholesterol metabolites and lipoproteins, as well as transi- fill several key criteria for an ideal biologic marker
tion metals [2,39,66,67,79 – 85]. In many of the published of early sporadic AD. However, further experimen-
reports, the chemical species in question were evaluated for tation will be required before these candidate bi-
their possible role in the pathophysiology of AD, with omarkers can be considered for routine clinical use.
deliberations concerning their biomarker potential arising Similarly, all AD biomarker candidates emerging
post facto [46,86]. Mere determination that a given com- from the application of mass spectrometry and other
pound is abnormally elevated or suppressed in AD body proteomic techniques [87,88] will require rigorous
fluids is insufficient to implicate the substance as a viable clinical evaluation for their suitability as bonafide
biologic marker of the disease in the absence of other diagnostic tools.
criteria listed in Section 4. (6) The pathology of AD is highly complex, and it is
therefore likely that combinations of individual bi-
omarkers will provide more accurate diagnostic and
prognostic information than any single marker mea-
7. Conclusions
sured in isolation (akin to use of multiple biochem-
ical indices to characterize connective tissue disease,
(1) AD is a public health problem of epidemic propor- liver failure, or cardiac ischemia).
tions for which current diagnostic and therapeutic (7) In October 2004 the National Institute on Aging
modalities remain inadequate. announced the Alzheimer Disease Neuroimaging
(2) The advent of a biologic marker that differentiates Initiative (ADNI), a large, multi-institutional pro-
early, sporadic AD from normal aging and other spective study proposing to delineate imaging and
dementing illnesses would represent a significant chemical biomarkers in 800 rigorously ascertained
achievement in the evaluation and management of subjects with normal cognition, MCI, and AD. This
this debilitating neurodegenerative disorder. An ac- initiative represents the most concerted effort yet to
curate, minimally invasive biologic marker of early develop accurate diagnostic, prognostic, and surro-
sporadic AD would serve the public interest by fa- gate markers of sporadic AD [89] and might identify
cilitating patient and family counseling, optimizing presymptomatic (pre-MCI) individuals at risk for
stratification of subgroups for enrollment in clinical this condition [90].
drug trials, and interpretation of treatment outcome
measures. The development of a biomarker that dif-
ferentiates “malignant” MCI cases at high risk for 8. Recommendations reaching consensus at the Third
conversion to AD from neuropsychologically iden- Canadian Consensus Conference on Diagnosis and
tical cases destined to manifest “benign” aging- Treatment of Dementia
related memory loss would be a particularly signif-
8.1. To primary care physicians
icant accomplishment. On theoretical grounds,
biomarkers might also prove useful in situations in
which concurrent medical conditions preclude or (1) Biologic markers for the diagnosis of AD should not,
confound cognitive and neuropsychological testing. at this juncture, be included in the battery of tests
Examples of the latter include patients with major routinely used by primary care physicians to evalu-
depression, delirium, suppressed consciousness, or ate subjects with memory loss (Grade C, Level 3).
who are otherwise uncooperative for detailed cogni- Consideration for such specialized testing in an in-
tive testing. dividual case should prompt referral of the patient to
(3) Although several candidate biomarkers of sporadic a specialist engaged in dementia evaluations or a
AD have been identified and commercialized, none memory clinic.
currently fulfills criteria for an ideal test.
(4) Decreased A␤1-42 and increased phospho-tau protein 8.2. To specialists
levels in the CSF are, at present, the most accurate
and reproducible chemical neurodiagnostics of early (2) Although highly desirable, there currently exist no
sporadic AD. These biomarkers also show promise blood- or urine-based AD diagnostics that can be
as prognosticators in subjects with MCI. However, unequivocally endorsed for the routine evaluation of
CSF evaluation by lumbar puncture remains imprac- memory loss in the elderly (Grade C, Level 3). The
tical for mass screening of elderly individuals with noninvasiveness of such tests, if and when they be-
memory impairment or AD risk factors. come available, would be suitable for mass screening
330 H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332

of subjects with memory loss presenting to special- [7] Mehta PD, Pirttila T, Mehta SP, Sersen EA, Aisen PS, Wisniewski
ists in their offices and memory clinics. HM. Plasma and cerebrospinal fluid levels of amyloid beta proteins
1-40 and 1-42 in Alzheimer disease. Arch Neurol 2000;57:100 –5.
(3) Because of their relative invasiveness and availabil- [8] Tamaoka A, Fukushima T, Sawamura N, Ishikawa K, Oguni E,
ity of other fairly accurate diagnostic modalities Komatsuzaki Y, et al. Amyloid beta protein in plasma from patients
(clinical, neuropsychological, and neuroimaging), with sporadic Alzheimer’s disease. J Neurol Sci 1996;141:65– 8.
CSF biomarkers should not be routinely performed [9] Sunderland T, Linker G, Mirza N, Putnam KT, Friedman DL, Kim-
in all subjects undergoing evaluation for memory mel LH, et al. Decreased beta-amyloid1-42 and increased tau levels in
cerebrospinal fluid of patients with Alzheimer disease. JAMA 2003;
loss (Grade D, Level 2).
289:2094 –103.
(4) CSF biomarkers might be considered in cases in [10] Hulstaert F, Blennow K, Ivanoiu A, Schoonderwaldt HC, Riemen-
which there are atypical features and diagnostic con- schneider M, De Deyn PP, et al. Improved discrimination of AD
fusion. CSF biomarkers might be useful in differen- patients using beta-amyloid(1-42) and tau levels in CSF. Neurology
tiating frontal variants of AD from FTD (Grade B, 1999;52:1555– 62.
Level 2). [11] Mollenhauer B, Cepek L, Bibl M, Wiltfang J, Schulz-Schaeffer WJ,
Ciesielczyk B, et al. Tau protein, Abeta42 and S-100B protein in
(5) When a decision to obtain CSF biomarkers is made, cerebrospinal fluid of patients with dementia with Lewy bodies.
combined A␤1-42 and p-tau concentrations should be Dement Geriatr Cogn Disord 2005;19:164 –70.
measured by validated enzyme-linked immunosor- [12] Clark CM, Xie S, Chittams J, Ewbank D, Peskind E, Galasko D,
bent assay (Grade A, Level 1). It might be best to et al. Cerebrospinal fluid tau and beta-amyloid: how well do these
convey the CSF samples to a centralized facility biomarkers reflect autopsy-confirmed dementia diagnoses? Arch
Neurol 2003;60:1696 –702.
(commercial or academic) with a track record in
[13] Gomez-Tortosa E, Gonzalo I, Fanjul S, Sainz MJ, Cantarero S,
generating high quality, reproducible data. Cemillan C, et al. Cerebrospinal fluid markers in dementia with lewy
(6) CSF biomarker data in isolation are insufficient to bodies compared with Alzheimer disease. Arch Neurol 2003;60:
diagnose or exclude AD (Grade C, Level 3). They 1218 –22.
should be interpreted in light of clinical, neuropsy- [14] Otto M, Esselmann H, Schulz-Shaeffer W, Neumann M, Schroter A,
chological, other laboratory, and neuroimaging data Ratzka P, et al. Decreased beta-amyloid1-42 in cerebrospinal fluid of
patients with Creutzfeldt-Jakob disease. Neurology 2000;54:1099 –
available for the individual under investigation. 102.
[15] Holmberg B, Johnels B, Blennow K, Rosengren L. Cerebrospinal
fluid Abeta42 is reduced in multiple system atrophy but normal in
Author Disclosures Parkinson’s disease and progressive supranuclear palsy. Mov Disord
2003;18:186 –90.
Hyman M. Schipper has received support from Capiron
[16] Sjogren M, Andreasen N, Blennow K. Advances in the detection of
Pharmaceuticals Inc. (Consultant), Osta Biotechnologies Alzheimer’s disease-use of cerebrospinal fluid biomarkers. Clin Chim
Inc. (Consultant), Molecular Biometrics, and LLC (Found- Acta 2003;332:1–10.
ing Scientist, Equity Holder). [17] Lewczuk P, Esselmann H, Groemer TW, Bibl M, Maler JM, Steinacker
P, et al. Amyloid beta peptides in cerebrospinal fluid as profiled with
surface enhanced laser desorption/ionization time-of-flight mass spec-
Acknowledgments trometry: evidence of novel biomarkers in Alzheimer’s disease. Biol
Psychiatry 2004;55:524 –30.
Hyman M. Schipper’s laboratory is supported by the [18] Maruyama M, Matsui T, Tanji H, Nemoto M, Tomita N, Ootsuki M,
Canadian Institutes of Health Research. et al. Cerebrospinal fluid tau protein and periventricular white matter
lesions in patients with mild cognitive impairment: implications for 2
major pathways. Arch Neurol 2004;61:716 –20.
References [19] Andreasen N, Minthon L, Clarberg A, Davidsson P, Gottfries J,
Vanmechelen E, et al. Sensitivity, specificity, and stability of CSF-tau
[1] Klunk WE. Biological markers of Alzheimer’s disease. Neurobiol in AD in a community-based patient sample. Neurology 1999;53:
Aging 1998;19:145–7. 1488 –94.
[2] Frank RA, Galasko D, Hampel H, Hardy J, de Leon MJ, Mehta PD, [20] Green AJ, Harvey RJ, Thompson EJ, Rossor MN. Increased tau in the
et al. Biological markers for therapeutic trials in Alzheimer’s disease: cerebrospinal fluid of patients with frontotemporal dementia and
proceedings of the biological markers working group—NIA initiative Alzheimer’s disease. Neurosci Lett 1999;259:133–5.
on neuroimaging in Alzheimer’s disease. Neurobiol Aging 2003;24: [21] Hesse C, Rosengren L, Vanmechelen E, Vanderstichele H, Jensen C,
521–36. Davidsson P, et al. Cerebrospinal fluid markers for Alzheimer’s
[3] Iwatsubo T. Amyloid beta protein in plasma as a diagnostic marker disease evaluated after acute ischemic stroke. J Alzheimers Dis 2000;
for Alzheimer’s disease. Neurobiol Aging 1998;19:161–3. 2:199 –206.
[4] Blennow K, Hampel H. CSF markers for incipient Alzheimer’s dis- [22] Grossman M, Farmer J, Leight S, Work M, Moore P, Van Deerlin V, et
ease. Lancet Neurol 2003;2:605–13. al. Cerebrospinal fluid profile in frontotemporal dementia and Alzhei-
[5] Shoji M. Cerebrospinal fluid Abeta40 and Abeta42: natural course mer’s disease. Ann Neurol 2005;57:721–9.
and clinical usefulness. Front Biosci 2002;7:997–1006. [23] Leszek J, Malyszczak K, Janicka B, Kiejna A, Wiak A. Total tau in
[6] Olsson A, Hoglund K, Sjogren M, Andreasen N, Minthon L, Lannfelt cerebrospinal fluid differentiates Alzheimer’s disease from vascular
L, et al. Measurement of alpha- and beta-secretase cleaved amyloid dementia. Med Sci Monit 2003;9:CR484 – 8.
precursor protein in cerebrospinal fluid from Alzheimer patients. Exp [24] Hampel H, Buerger K, Zinkowski R, Teipel SJ, Goernitz A, Andreasen
Neurol 2003;183:74 – 80. N, et al. Measurement of phosphorylated tau epitopes in the differential
H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332 331

diagnosis of Alzheimer disease: a comparative cerebrospinal fluid study. proteins identify Alzheimer disease in subjects with mild cognitive
Arch Gen Psychiatry 2004;61:95–102. impairment. Arch Neurol 2002;59:1729 –34.
[25] Lewczuk P, Esselmann H, Bibl M, Beck G, Maler JM, Otto M, et al. [43] Hampel H, Teipel SJ, Fuchsberger T, Andreasen N, Wiltfang J, Otto
Tau protein phosphorylated at threonine 181 in CSF as a neurochem- M, et al. Value of CSF beta-amyloid1-42 and tau as predictors of
ical biomarker in Alzheimer’s disease: original data and review of the Alzheimer’s disease in patients with mild cognitive impairment. Mol
literature. J Mol Neurosci 2004;23:115–22. Psychiatry 2004;9:705–10.
[26] Vanmechelen E, Vanderstichele H, Davidsson P, Van Kerschaver E, [44] Herukka SK, Hallikainen M, Soininen H, Pirttila T. CSF Abeta42 and
Van Der Perre B, Sjogren M, et al. Quantification of tau phosphor- tau or phosphorylated tau and prediction of progressive mild cogni-
ylated at threonine 181 in human cerebrospinal fluid: a sandwich tive impairment. Neurology 2005;64:1294 –7.
ELISA with a synthetic phosphopeptide for standardization. Neurosci [45] Ivanoiu A, Sindic CJ. Cerebrospinal fluid TAU protein and amyloid
Lett 2000;285:49 –52. beta42 in mild cognitive impairment: prediction of progression to
[27] Schoonenboom NS, Pijnenburg YA, Mulder C, Rosso SM, Van Elk Alzheimer’s disease and correlation with the neuropsychological ex-
EJ, Van Kamp GJ, et al. Amyloid beta(1-42) and phosphorylated tau amination. Neurocase 2005;11:32–9.
in CSF as markers for early-onset Alzheimer disease. Neurology [46] Klunk WE. Blood and CSF biomarkers for AD revisited: what’s new,
2004;62:1580 – 4. what’s good, and is this where we should be looking? Neurobiol
[28] Kurz A, Riemenschneider M, Wallin A. Potential biological markers for Aging 2002;23:517–9; discussion 521–2.
cerebrovascular disease. Int Psychogeriatr 2003;15(Suppl 1):89 –97. [47] Peskind ER, Riekse R, Quinn JF, Kaye J, Clark CM, Farlow MR, et
[29] Parnetti L, Lanari A, Amici S, Gallai V, Vanmechelen E, Hulstaert F. al. Safety and acceptability of the research lumbar puncture. Alzhei-
CSF phosphorylated tau is a possible marker for discriminating Alz- mer Dis Assoc Disord 2005;19:220 –5.
heimer’s disease from dementia with Lewy bodies: Phospho-Tau [48] De La Monte SM, Wands JR. The AD7c-NTP neuronal thread protein
International Study Group. Neurol Sci 2001;22:77– 8. biomarker for detecting Alzheimer’s disease. J Alzheimers Dis 2001;
[30] Blennow K, Wallin A, Agren H, Spenger C, Siegfried J, Van- 3:345–53.
mechelen E. Tau protein in cerebrospinal fluid: a biochemical marker [49] Chertkow H, Bergman H, Schipper HM, Gauthier S, Bouchard R,
for axonal degeneration in Alzheimer disease? Mol Chem Neuro- Fontaine S, et al. Assessment of suspected dementia. Can J Neurol Sci
pathol 1995;26:231– 45. 2001;28(Suppl 1):S28 – 41.
[31] Hesse C, Rosengren L, Andreasen N, Davidsson P, Vanderstichele H,
[50] Kennard ML, Feldman H, Yamada T, Jefferies WA. Serum levels of
Vanmechelen E, et al. Transient increase in total tau but not phospho-
the iron binding protein p97 are elevated in Alzheimer’s disease. Nat
tau in human cerebrospinal fluid after acute stroke. Neurosci Lett
Med 1996;2:1230 –5.
2001;297:187–90.
[51] Kim DK, Seo MY, Lim SW, Kim S, Kim JW, Carroll BJ, et al. Serum
[32] Schonknecht P, Hempel A, Hunt A, Seidl U, Volkmann M, Pantel J,
melanotransferrin, p97 as a biochemical marker of Alzheimer’s dis-
et al. Cerebrospinal fluid tau protein levels in schizophrenia. Eur Arch
ease. Neuropsychopharmacology 2001;25:84 –90.
Psychiatry Clin Neurosci 2003;253:100 –2.
[52] Desrosiers RR, Bertrand Y, Nguyen QT, Demeule M, Gabathuler R,
[33] Goodall CA, Head MW, Everington D, Ironside JW, Knight RS,
Kennard ML, et al. Expression of melanotransferrin isoforms in
Green AJ. Raised CSF phospho-tau concentrations in variant
human serum: relevance to Alzheimer’s disease. Biochem J 2003;
Creutzfeldt-Jakob disease: diagnostic and pathological implications.
374(Pt 2):463–71.
J Neurol Neurosurg Psychiatry 2006;77:89 –91.
[53] Di Luca M, Pastorino L, Bianchetti A, Perez J, Vignolo LA, Lenzi
[34] Riemenschneider M, Wagenpfeil S, Vanderstichele H, Otto M,
GL, et al. Differential level of platelet amyloid beta precursor protein
Wiltfang J, Kretzschmar H, et al. Phospho-tau/total tau ratio in
isoforms: an early marker for Alzheimer disease. Arch Neurol 1998;
cerebrospinal fluid discriminates Creutzfeldt-Jakob disease from
other dementias. Mol Psychiatry 2003;8:343–7. 55:1195–200.
[35] Schonknecht P, Pantel J, Hunt A, Volkmann M, Buerger K, Hampel [54] Di Luca M, Pastorino L, Cattabeni F, Zanardi R, Scarone S, Racagni
H, et al. Levels of total tau and tau protein phosphorylated at threo- G, et al. Abnormal pattern of platelet APP isoforms in Alzheimer
nine 181 in patients with incipient and manifest Alzheimer’s disease. disease and Down syndrome. Arch Neurol 1996;53:1162– 6.
Neurosci Lett 2003;339:172– 4. [55] Rosenberg RN, Baskin F, Fosmire JA, Risser R, Adams P, Svetlik D,
[36] Buerger K, Teipel SJ, Zinkowski R, Blennow K, Arai H, Engel R, et et al. Altered amyloid protein processing in platelets of patients with
al. CSF tau protein phosphorylated at threonine 231 correlates with Alzheimer disease. Arch Neurol 1997;54:139 – 44.
cognitive decline in MCI subjects. Neurology 2002;59:627–9. [56] Padovani A, Pastorino L, Borroni B, Colciaghi F, Rozzini L, Mon-
[37] Arai H, Ishiguro K, Ohno H, Moriyama M, Itoh N, Okamura N, et al. astero R, et al. Amyloid precursor protein in platelets: a peripheral
CSF phosphorylated tau protein and mild cognitive impairment: a marker for the diagnosis of sporadic AD. Neurology 2001;
prospective study. Exp Neurol 2000;166:201–3. 57:2243– 8.
[38] de Leon MJ, DeSanti S, Zinkowski R, Mehta PD, Pratico D, Segal S, [57] Baskin F, Rosenberg RN, Iyer L, Hynan L, Cullum CM. Platelet APP
et al. MRI and CSF studies in the early diagnosis of Alzheimer’s isoform ratios correlate with declining cognition in AD. Neurology
disease. J Intern Med 2004;256:205–23. 2000;54:1907–9.
[39] Blennow K. Cerebrospinal fluid protein biomarkers for Alzheimer’s [58] Borroni B, Colciaghi F, Archetti S, Marcello E, Caimi L, Di Luca M, et
disease. NeuroRx 2004;1:213–25. al. Predicting cognitive decline in Alzheimer disease. Role of platelet
[40] Wahlund LO, Blennow K. Cerebrospinal fluid biomarkers for disease amyloid precursor protein. Alzheimer Dis Assoc Disord 2004;18:
stage and intensity in cognitively impaired patients. Neurosci Lett 32– 4.
2003;339:99 –102. [59] Borroni B, Colciaghi F, Caltagirone C, Rozzini L, Broglio L, Cat-
[41] Ganzer S, Arlt S, Schoder V, Buhmann C, Mandelkow EM, Finckh tabeni F, et al. Platelet amyloid precursor protein abnormalities in
U, et al. CSF-tau, CSF-Abeta1-42, ApoE-genotype and clinical pa- mild cognitive impairment predict conversion to dementia of Alzhei-
rameters in the diagnosis of Alzheimer’s disease: combination of mer type: a 2-year follow-up study. Arch Neurol 2003;60:1740 – 4.
CSF-tau and MMSE yields highest sensitivity and specificity. J Neu- [60] Padovani A, Borroni B, Colciaghi F, Pettenati C, Cottini E, Agosti C,
ral Transm 2003;110:1149 – 60. et al. Abnormalities in the pattern of platelet amyloid precursor
[42] Riemenschneider M, Lautenschlager N, Wagenpfeil S, Diehl J, protein forms in patients with mild cognitive impairment and Alz-
Drzezga A, Kurz A. Cerebrospinal fluid tau and beta-amyloid 42 heimer disease. Arch Neurol 2002;59:71–5.
332 H.M. Schipper / Alzheimer’s & Dementia 3 (2007) 325–332

[61] Borroni B, Colciaghi F, Pastorino L, Pettenati C, Cottini E, Rozzini disease by measuring lymphocyte heme oxygenase-1 expression with
L, et al. Amyloid precursor protein in platelets of patients with real-time RT-PCR. Ann N Y Acad Sci 2002;977:173– 8.
Alzheimer disease: effect of acetylcholinesterase inhibitor treatment. [76] Schipper HM, Mawal Y, Chertkow H, Bergman H. Blood HO-1
Arch Neurol 2001;58:442– 6. mRNA in AD and MCI. Neurology 2001;57:2142–3.
[62] Liu HC, Chi CW, Ko SY, Wang HC, Hong CJ, Lin KN, et al. [77] Maes OC, Kravitz S, Mawal Y, Su H, Liberman A, Mehindate K, et
Cholinesterase inhibitor affects the amyloid precursor protein iso- al. Characterization of alpha(1)-antitrypsin as a heme oxygenase-1
forms in patients with Alzheimer’s disease. Dement Geriatr Cogn suppressor in Alzheimer plasma. Neurobiol Dis 2006;24:89 –100.
Disord 2005;19:345– 8. [78] Schipper HM, Cissé S, Stopa EG. Expression of heme oxygenase-1 in
[63] Zimmermann M, Borroni B, Cattabeni F, Padovani A, Di Luca M. the senescent and Alzheimer-diseased brain. Ann Neurol 1995;37:
Cholinesterase inhibitors influence APP metabolism in Alzheimer 758 – 68.
disease patients. Neurobiol Dis 2005;19:237– 42. [79] Small DH. Alzheimer’s disease biomarkers: their value in diagnosis
[64] Schipper HM. Brain iron deposition and the free radical-mitochondrial and clinical trials. Front Biosci 2002;7:986 – 8.
theory of ageing. Ageing Res Rev 2004;3:265–301. [80] Irizarry MC. Biomarkers of Alzheimer disease in plasma. NeuroRx
[65] Markesbery WR, Carney JM. Oxidative alterations in Alzheimer’s 2004;1:226 –34.
disease. Brain Pathol 1999;9:133– 46. [81] Davidsson P, Sjogren M. The use of proteomics in biomarker dis-
[66] Galasko D. Biological markers and the treatment of Alzheimer’s covery in neurodegenerative diseases. Dis Markers 2005;21:81–92.
disease. J Mol Neurosci 2001;17:119 –25. [82] Ho L, Sharma N, Blackman L, Festa E, Reddy G, Pasinetti GM. From
[67] Maccioni RB, Lavados M, Maccioni CB, Mendoza-Naranjo A. Bio-
proteomics to biomarker discovery in Alzheimer’s disease. Brain Res
logical markers of Alzheimer’s disease and mild cognitive impair-
Brain Res Rev 2005;48:360 –9.
ment. Curr Alzheimer Res 2004;1:307–14.
[83] Wada H. Analyses of serum concentrations of apolipoproteins in the
[68] Tuppo EE, Forman LJ, Spur BW, Chan-Ting RE, Chopra A, Cavalieri
demented elderly. Intern Med 2000;39:220 –2.
TA. Sign of lipid peroxidation as measured in the urine of patients
[84] Meli M, Perier C, Ferron C, Parssegny F, Denis C, Gonthier R, et al.
with probable Alzheimer’s disease. Brain Res Bull 2001;54:565– 8.
Serum pentosidine as an indicator of Alzheimer’s disease. J Alzhei-
[69] Pratico D, V MYL, Trojanowski JQ, Rokach J, Fitzgerald GA. In-
mers Dis 2002;4:93– 6.
creased F2-isoprostanes in Alzheimer’s disease: evidence for en-
[85] Mulder C, Scheltens P, Visser JJ, van Kamp GJ, Schutgens RB.
hanced lipid peroxidation in vivo. Faseb J 1998;12:1777– 83.
Genetic and biochemical markers for Alzheimer’s disease: recent
[70] Pratico D, Clark CM, Liun F, Rokach J, Lee VY, Trojanowski JQ.
Increase of brain oxidative stress in mild cognitive impairment: a developments. Ann Clin Biochem 2000;37(Pt 5):593– 607.
possible predictor of Alzheimer disease. Arch Neurol 2002;59:972– 6. [86] Padovani A, Borroni B, Di Luca M. Advances on biological markers
[71] Montine TJ, Kaye JA, Montine KS, McFarland L, Morrow JD, Quinn in early diagnosis of Alzheimer disease. Adv Clin Chem 2005;39:
JF. Cerebrospinal fluid abeta42, tau, and f2-isoprostane concentra- 107–29.
tions in patients with Alzheimer disease, other dementias, and in [87] Choe LH, Dutt MJ, Relkin N, Lee KH. Studies of potential cerebro-
age-matched controls. Arch Pathol Lab Med 2001;125:510 –2. spinal fluid molecular markers for Alzheimer’s disease. Electrophore-
[72] Montine TJ, Quinn JF, Milatovic D, Silbert LC, Dang T, Sanchez S, sis 2002;23:2247–51.
et al. Peripheral F2-isoprostanes and F4-neuroprostanes are not in- [88] Yu H-L, Chertkow HM, Bergman H, Schipper HM. Aberrant profiles
creased in Alzheimer’s disease. Ann Neurol 2002;52:175–9. of native and oxidized glycoproteins in Alzheimer plasma. Proteom-
[73] Greco A, Minghetti L, Levi G. Isoprostanes, novel markers of oxi- ics 2003;3:2240 – 8.
dative injury, help understanding the pathogenesis of neurodegenera- [89] Trojanowski JQ, Lee M-Y. Commentary on “Diagnosis of Alzhei-
tive diseases. Neurochem Res 2000;25:1357– 64. mer’s disease: Two decades of progress”: Alzheimer’s disease
[74] Schipper HM, Chertkow H, Mehindate K, Frankel D, Melmed C, research—new opportunities for early diagnosis and drug discovery.
Bergman H. Evaluation of heme oxygenase-1 as a systemic biological Alz Dement 2005;1:119 –20.
marker of sporadic AD. Neurology 2000;54:1297–304. [90] Morris JC, Kimberly A, Quaid K, Holtzman DM, Kantarci K, Kaye
[75] Ishizuka K, Kimura T, Yoshitake J, Akaike T, Shono M, Takamatsu J, et al. Role of biomarkers in studies of presymptomatic Alzheimer’s
J, et al. Possible assessment for antioxidant capacity in Alzheimer’s disease. Alz Dement 2005;1:145–51.
Alzheimer’s & Dementia 3 (2007) 333–335

Structural neuroimaging in the diagnosis of dementia


Tiffany Chow*
The Rotman Research Institute Baycrest Centre for Geriatric Care; Department of Medicine, Neurology Division, University of Toronto, Toronto,
Ontario, Canada

Abstract This article reviews recent use of structural imaging in the diagnosis of dementia and presents
evidence-based recommendations approved at the meeting of the Third Consensus Conference on
the Diagnosis and Treatment of Dementia (CCCDTD3) held in Montreal in March 2006. Although
the consensus group concurs that it is possible to specify circumstances in which structural imaging
has a role to rule out pathology, it also has a role in ruling in concomitant cerebrovascular disease
for patient management.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Alzheimer’s disease; Vascular dementia; MRI; CT; Structural imaging

The decision to exclude structural neuroimaging from a review was derived from neither primary care nor research
dementia evaluation must weigh several opposing factors: environments exclusively, some indications for imaging
the physician’s confidence that the history and neurologic have implications for any clinical setting, whereas other
examination have sufficiently ruled out the likelihood of indications are currently mandated for research purposes
finding a neurosurgical lesion that would reverse the pa- only.
tient’s cognitive complaints, the availability of the imaging The following indications could not be separated into
procedure, the need to identify comorbidity, and the practice applications for primary care versus research setting or CT
setting (clinical vs research). Primary care providers and of head versus MRI. Justifications for ordering a diagnostic
specialists will perceive this set of considerations differ- CT of head or MRI of brain in the past focused on the
ently. Indicators to increase the yield from structural imag- importance of ruling out etiologies of dementia that could
ing while decreasing the number of studies ordered and be reversed with a neurosurgical procedure (normal pres-
minimizing the number of treatable dementia cases missed sure hydrocephalus, subdural hematoma, and neoplasm).
might be useful to all physicians (Appendix 1) [1– 4]. Such One cost analysis calculated a work-up individualized to
indicators would also decrease the costs of dementia work- course of illness that offers imaging only to those subjects
ups. younger than age 65 years [6]. Other studies have since
Search criteria for the indications listed below included shown with Class II and III evidence that only a small
original articles or clinical trials examining the utility of number of cases with neurosurgical lesions have dementia
structural magnetic resonance imaging (MRI) or computed as the sole presenting symptom, and that even fewer of
tomography (CT). Utility could be reported in terms of those cases enjoy significant resolution of the cognitive
benefit to patient outcome or cost/benefit analyses. All but impairment after treatment [2,5–10]. In this sense, there is
two [5,6] of the 27 references used for this review pertain to fair evidence to recommend against structural neuroimaging
patients diagnosed through specialty clinics. Most studies to rule out a neurosurgical lesion during a work-up for
are from research cohorts; four were from clinical cohorts dementia (Grade D, Level 2). This recommendation did not
[2,7–9]. Although most of the evidence discovered for this
reach consensus at the CCCDTD3 meeting, however.
Although four studies report Class II evidence that atro-
*Corresponding author. Tel.: (416) 785-2500 x3459; Fax: (416) 785-
phy as measured on CT [11] or MRI [12–14] distinguishes
2862. Alzheimer’s disease (AD) from nondemented healthy el-
E-mail address: tchow@rotman-baycrest.on.ca derly, neuroimaging has seldom changed the initial clinical
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.016
334 T. Chow / Alzheimer’s & Dementia 3 (2007) 333–335

diagnosis of AD. Instead, there is fair evidence to support [2] Freter SBH, Gold S, Chertkow H, Clarfield AM. Prevalence of
use of structural neuroimaging to rule in concomitant cere- potentially reversible dementias and actual reversibility in a memory
clinic cohort. CMAJ 1998;159:657– 62.
brovascular disease that can affect patient management
[3] Gifford DR, Holloway RG, Vickrey BG. Systematic review of clin-
(Grade B, Level 2). Two studies provide Class II evidence ical prediction rules for neuroimaging in the evaluation of dementia.
that CT and MRI scans identify cerebrovascular disease that Arch Intern Med 2000;160:2855– 62.
was not suspected clinically, resulting in the addition of [4] Martin DC, Miller J, Kapoor W, Karpf M, Boller F. Clinical predic-
stroke risk management to the care plan [1,15–17]. tion rules for computed tomographic scanning in senile dementia.
Within a research setting, there is fair evidence to sup- Arch Intern Med 1987;147:77– 80.
[5] Alexander EM, Buchner DM, Cain KC, Larson EB. Do surgical brain
port the use of structural neuroimaging to predict conver-
lesions present as isolated dementia? a population-based study. J Am
sion of mild cognitive impairment (MCI) to AD and pro- Geriatr Soc 1995;43:138 – 43.
gression through stages of AD (Grade B, Level 2). Three [6] Foster GR, Scott DA, Payne S. The use of CT scanning in demen-
labs have replicated Class II evidence to support volumetric tia: a systematic review. Int J Technol Assess Health Care 1999;
analyses of entorhinal cortex and hippocampus to identify 15:406 –23.
those patients with MCI who are most likely to progress to [7] Walstra GJM, Teunisse S, van Gool HvC. Reversible dementia in
elderly patients referred to a memory clinic. J Neurol 1997;244:
AD within several years [18 –20]. There is also fair evi-
17–22.
dence to support the use of structural neuroimaging to track [8] Clarfield AM. The decreasing prevalence of reversible dementias: an
the progression of AD in clinical trials, especially if the updated meta-analysis. Arch Intern Med 2003;163:2219 –29.
morphometry is combined with neuropsychological testing [9] Chong MS, Sahadevan S. An evidence-based clinical approach to the
(Grade B, Level 2) [12,21–23]. diagnosis of dementia. Ann Acad Med Singapore 2003;32:740 – 48.
[10] Simon D. Routine imaging in suspected dementia: diagnostic and
therapeutic yield. Clin Res 1992;40:565A.
1. Recomendations supported at the Third [11] Jobst KA, Barnetson LP, Shepstone BJ. Accurate prediction of his-
tologically confirmed Alzheimer’s disease and the differential diag-
CCCDTD meeting
nosis of dementia: the use of NINCDS-ADRDA and DSM-III-R
criteria, SPECT, X-ray CT, and APO E4 medial temporal lobe
1. There is fair evidence to support the selective use of dementias—the Oxford Project to Investigate Memory and Aging. Int
CT or MRI scanning in the work-up for dementia per Psychogeriatr 1997;(Suppl 1):191–222; discussion 247–52.
[12] Zakzanis KK. Quantitative evidence for neuroanatomic and neuro-
1999 guidelines (Grade B, Level 2).
psychological markers in dementia of the Alzheimer’s type. J Clin
2. There is fair evidence to support use of structural Exp Neuropsychol 1998;20:259 – 69.
neuroimaging to rule in concomitant cerebrovascular [13] Frisoni GB, Weiss C, Geroldi C, Bianchetti A, Trabucchi M. Linear
disease that can affect patient management (Grade B, measures of atrophy in mild Alzheimer disease. AJNR Am J Neuro-
Level 2). radiol 1996;17:913–23.
3. There is fair evidence to support the use of structural [14] Laakso MP, Soininen H, Partanen K, Lehtovirta M, Hallikanen M,
Hanninen T, HMHTHEVPRP. MRI of the hippocampus in Alzheimer’s
neuroimaging to track the progression of AD in clin-
disease: sensitivity, specificity, and analysis of the incorrectly classified
ical trials, especially if the morphometry is combined subjects. Neurobiol Aging 1998;19:23–31.
with neuropsychological testing (Grade B, Level 2). [15] Massoud F, Devi G, Moroney JT, Stern Y, Lawton A, Bell K, et al.
The role of routine laboratory studies and neuroimaging in the diag-
nosis of dementia: a clinicopathological study. J Am Geriatr Soc
Author Disclosures 2000;48:1204 –10.
[16] Wilcock G, Mobius HJ, Stoffler A, MMM 500 group. A double-blind,
Tiffany Chow has received support from with Novartis placebo-controlled multicentre study of memantine in mild to mod-
(Speaker’s Bureau), Janssen Ortho Inc. (Consultant), and erate vascular dementia (MMM500). Int Clin Psychopharmacol 2002;
CanCog Technologies (Consultant). 17:297–305.
[17] Lopez OL, Kuller LH, Becker JT, Jagust WJ, DeKosky ST, Fitz-
patrick A, et al. Classification of vascular dementia in the Cardiovas-
Acknowledgments cular Health Study Cognition Study. Neurology 2005;64:1539 – 47.
[18] Killiany RJ, Gomez-Isla T, Moss MB, Kikinis R, Jolesz F, Tanzi R,
Tiffany Chow is supported by NIA grant F32 et al. MRI measures of entorhinal cortex vs hippocampus in preclin-
AG022802; the University of Toronto Dean’s Fund for New ical AD. Neurology 2002;58:1188 –96.
[19] Rusinek H, Frid D, Tsui WH, Tarshish CY, Convit A, de Leon MJ.
Faculty (#457494), and an endowment to the Sam and Ida
Regional brain atrophy rate predicts future cognitive decline: 6-year
Ross Memory Clinic. longitudinal MR imaging study of normal aging. Radiology 2003;
229:691– 6.
[20] Jack CR Jr, Shiung MM, Weigand SD, O’Brien PC, Gunter JL, Boeve
References BF, et al. Brain atrophy rates predict subsequent clinical conversion
in normal elderly and amnestic MCI. Neurology 2005;65:1227–31.
[1] Chui H, Zhang Q. Evaluation of dementia: a systematic study of the [21] Frisoni GB, Scheltens P, Galluzzi S, Nobili FM, Fox NC, Robert PH,
usefulness of the American Academy of Neurology’s practice param- et al. Neuroimaging tools to rate regional atrophy, subcortical cere-
eters. Neurology 1997;49:925–35. brovascular disease, and regional cerebral blood flow and metabo-
T. Chow / Alzheimer’s & Dementia 3 (2007) 333–335 335

lism: consensus paper of the EADC. J Neurol Neurosurg Psychiatry Rapid (eg, during 1 to 2 months) unexplained decline
2003;74:1371– 81. in cognition or function
[22] Cardenas VA, Hardin D, Ezekiel F, Weber P, Jagust WJ, Chui HC,
et al. Comparison of methods for measuring longitudinal brain
“Short” duration of dementia (less than 2 years)
change in cognitive impairment and dementia. Neurobiol Aging Recent/significant head trauma
2003;24:537– 44. Unexplained neurologic symptoms (eg, new onset of
[23] Zamrini E, De Santi S, Tolar M. Imaging is superior to cognitive severe headache or seizures)
testing for early diagnosis of Alzheimer’s disease. Neurobiol Aging History of cancer (especially in sites and types that
2004;25:685–91.
[24] Organizing Committee CCCotAoD. Assessing dementia: the Cana-
metastasize to the brain)
dian Consensus. CMAJ 1991;144:851–3. Use of anticoagulants or history of a bleeding disorder
History of urinary incontinence and gait disorder early
in the course of dementia (as might be found in
normal pressure hydrocephalus)
Appendix 1. CCCAD indicators [24] Any new localizing sign (eg, hemiparesis or a Babin-
A cranial CT scan is recommended if one or more of the ski’s reflex)
following criteria are present: Unusual or atypical cognitive symptoms or presenta-
tion (eg, progressive aphasia)
Age younger than 60 years Gait disturbance
Alzheimer’s & Dementia 3 (2007) 336 –340

Functional neuroimaging in the diagnosis of dementia


Michael Borrie*
Division of Geriatric Medicine, St. Joseph’s Health Care London-Parkwood Site; Professor, Department of Medicine, University of Western Ontario,
London, Ontario, Canada

The functional neuroimaging modalities of fluoro-2- The decline from normal cognition to MCI has been
deoxy-D-glucose positron emission tomography (FDG-PET), predicted by baseline entorhinal cortex MRglc with 84%
single photon emission computed tomography (SPECT), accuracy, 83% sensitivity, and 85% specificity [5]. With
functional magnetic resonance imaging (fMRI), magnetic ROIs, it is possible to find significant differences when
resonance spectroscopy (MRS), and their roles in diagnos- comparing normal controls with patients with MCI or AD,
ing dementia and predicting the progression from mild cog- but in this study there were no significant differences be-
nitive impairment (MCI) to dementia and tracking its pro- tween MCI and AD [7]. However, in studying MRglc re-
gression have recently been reviewed [1– 4]. The question ductions in ROIs in normal elderly and in subjects with MCI
asked by this evidence-based review was “Is there sufficient and AD, De Santi et al [6] reported a diagnostic accuracy of
evidence to recommend the use of a functional neuroimag- 85% (80% MCI, 92% AD).
ing technique by family physicians or specialists in Canada There are fewer studies that follow the decline from MCI
to make or differentiate a diagnosis of dementia in people to AD, with the numbers of subjects in these studies ranging
presenting with cognitive impairment?” from 17 to 52 [2]. Overall, the parietotemporal area meta-
bolic reductions predicted an overall accuracy of 86%, sen-
1. Methods sitivity and specificity ranging from 75% to 100%.
A recent meta-analysis of PET articles, published be-
The literature review used PubMed and EMBASE data- tween 1989 and 2003, found 15 articles that reported
bases including articles in English and human research from sensitivity and specificity that met the study inclusion/
January 1996 to February 2006.Search terms included de- exclusion criteria. The summary sensitivity of PET was
mentia, mild cognitive impairment, PET, SPECT, fMRI, 86% confidence (95% confidence interval [CI], 76% to
and MRS. A separate search used the same parameters but 93%), and the summary specificity was 86% (95% CI, 72%
restricted it to review articles to identify recent evidence- to 93%) [8]. The limitations included use of an imperfect
based reviews. reference standard, verification bias, and variability in the
choice of the disease spectrum. This limited generalizability
2. FDG-PET to a clinical setting. The authors recommended further re-
search on the use of PET in the diagnosis of AD, focusing
Reduction in brain glucose metabolism (MRglc) cor- on these limitations in clinical settings.
rected for atrophy, in the parietotemporal, frontal and A four-point visual rating scale to evaluate the presence
posterior cingulate cortices, is the typical pattern in Alz- and severity of medial temporal lobe (MTL) hypometabo-
heimer’s disease (AD). High resolution PET scanning,
lism on FDG-PET scans has been evaluated in normal
coregistered with MRI, has resulted in reports of reductions
controls and subjects with MCI and AD. The visual MTL
in MRglc in more specific regions of interest (ROIs). The
ratings were compared with MRglc data from ROIs in the
hippocampus, known to be affected early in AD, has reduc-
MRI-coregistered PET scans of all subjects as well as stan-
tions in MRglc in patients with mild cognitive impairment
dard rating of neocortical evaluation. There was high intra-
(MCI) compared with normal elderly [5–7].
rater and inter-rater reliabilities between the MTL and cor-
tical ratings (P ⫽ .001). MTL ratings were also highly
*Corresponding author. Tel.: (519) 685-4021; Fax: 519-685-4093. correlated with ROI MRglc. The diagnostic accuracy or
E-mail address: michael.borrie@sjhc.london.on.ca cortical ratings alone was improved by combining MTL
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.004
M. Borrie / Alzheimer’s & Dementia 3 (2007) 336 –340 337

with cortical ratings, correctly identifying 100% of AD, pooled [15]. The weighted sensitivity for brain SPECT
77% of MCI, and 85% of normal elderly. If these obser- against neuropathology was 74%, with a weighted specific-
vations are confirmed in other studies, this scale has ity of 91%. This finding suggests that clinical criteria are
promise as a clinically useful instrument in patients with more sensitive than brain SPECT (81% versus 74%), and
MCI undergoing PET scans [9]. brain SPECT has a higher specificity than clinical criteria
The cost, availability, and, to a lesser extent, radiation (91% versus 70%) [15]. These are conservative estimates
exposure are three present barriers for PET scanning to be because, in practice, SPECT scans are not usually inter-
used clinically in Canada. The approval in 2004 in the U.S. preted without clinical information. The authors concluded
by Medicare and Medicaid for the use of PET, in cases that that in dementia patients who present with possible comor-
remain uncertain after standard clinical evaluation for de- bid symptoms of vascular disease or who do not fit clinical
mentia, will increase the clinical use and reporting of its criteria, SPECT scanning might help rule in AD when the
diagnostic utility. SPECT scan is positive and rule it out when the SPECT
Ligand molecules that cross the blood-brain barrier and scan is negative for AD [15].
combine with amyloid are being developed and tested for One SPECT 2-year longitudinal study in MCI subjects
use with PET scanning to quantify in vivo amyloid depos- reported that the initial findings in the hippocampal-
ited in human brains [10 –12]. If these molecules are equally amygdaloid complex, the posterior cingulate, the anterior
successful with SPECT in defining amyloid load, the bar- thalamus, and the caudal portion of the anterior cingulate
riers of cost and availability of PET might be removed. predicted progression to dementia in 78% of those who
A recent review of the role of neuroimaging in MCI [4] progress to AD [4,18].
concluded that although neuroimaging modalities had po- There have been few SPECT studies reporting cross-
tential to become valuable tools for the early diagnosis of sectional differences between MCI and controls [19,20].
AD, larger prospective longitudinal studies are needed. The Some SPECT studies that used conventional clinical routine
Alzheimer’s Disease Neuroimaging Initiative (ADNI), be- techniques, found computed tomography or MRI had better
gun in the United States and Canada in 2005, is one study discrimination between normal controls, MCI, and dementia
that will establish standardization of neuroimaging with than SPECT [21] and in discriminating between progressive
MRI at 1.5 T and 3 T and PET and might realize this compared with stable MCI [19,22].
potential.
4. fMRI
3. SPECT
Functional MRI scans visualize the location of activity in
SPECT uses radiopharmacologic perfusion agents to the brain in response to defined sensory stimulation or cogni-
image blood flow patterns in the brain. 99mTechnetium- tive tasks compared with scans taken at rest. Areas of increased
hexamethyl-propylenamine oxime (99mTc-HMPAO) is the intensity on the scans correspond to the brain areas activated by
most commonly used agent, which is lipophilic and quickly the stimulus. Neuronal activity is measured indirectly through
crosses the blood-brain barrier into the neurons and glia and the blood oxygenation level dependent (BOLD) hemodynamic
is metabolized into a “trapped” lipophobic compound. response to the defined stimulus [23].
Clearance of 99mTc-HMPAO from the vascular pool al- Recently published fMRI studies have examined the re-
lows the imaging of brain tissue, which has a symmetrical sponse in people with AD [24], MCI [25], MCI compared
pattern with a gray to white matter flow pattern of 4:1 with mild AD [26 –28], FTD versus early AD [29], MCI and
[13]. AD has a characteristic pattern with perfusion def- a cholinesterase inhibitor [30], AD and a cholinesterase
icits in the parietal and temporal lobes [14]. A recent inhibitor [31], and normal older adults with or without
systematic review examined the added value of SPECT to APOE e4 allelle [32]. In each of these studies, the number
discriminate between clinically defined AD, vascular de- of subjects was small (range, 7 to 41 subjects per group) and
mentia (VD), frontotemporal dementia (FTD), and other recruited from university centers or tertiary referral centers.
non-dementia comparison groups [15]. Randomized con- In the one study of normal older adults, the subjects were
trolled trials have not been conducted because of the use recruited by print advertisements [32].
of ionizing radiation, leaving case-control and cohort Only one study had a longitudinal follow-up, mean time
studies as the next best alternative [15]. 2.5 years, to compare those with MCI who “declined” and
SPECT discriminated between AD, VD, FTD, and non- those who remained stable [25]. In this study, activation in
dementia patient comparison (PC) groups. The pooled the hippocampus and parahippocampal gyrus (PHG) corre-
weighted sensitivities ranged from 65.7% (AD versus PC) lated with better memory performance. However, paradox-
to 71.5% (AD versus FTD). Specificity was lowest overall ically those who “progressed” on the clinical dementia rat-
for AD versus VD at 75.9%, increasing to 79.1% for AD ing scale activated the PHG to a greater extent. This was
versus PC [15]. Data from two studies that compared clin- interpreted as a possible compensatory response to an un-
ical, SPECT, and histopathologic verification [16,17] were derlying progressive AD pathology. In a normal older adult
338 M. Borrie / Alzheimer’s & Dementia 3 (2007) 336 –340

study comparing those with and without the APOE e4 al- ingly in this study, measurements from the hippocampal
lelle, those with the e4 allelle had a greater magnitude and cortex were not predictive. These findings need to be du-
extent of BOLD brain response during the picture learning plicated.
stimulus compared with those matched with the e3 allelle. In cognitively intact subjects older than 85 years, MRS
The result suggested that the APOE e4 genotype directly has shown that subjects with small hippocampal volumes
influences the brain’s response and was consistent with the have a significantly lower NAA/MI ratio in parietal and
hypothesis of a compensatory mechanism in those at genetic temporal lobes compared with the other subjects. This study
risk of AD [32]. did not report a longitudinal component; therefore, it is
These fMRI studies are providing insights into how brain unclear whether these subjects are at greater risk of devel-
ROIs respond to stimuli in response to different degrees of oping dementia [46].
impairment and assist in hypothesis generation. They are MRS holds considerable promise as a modality for dif-
not applicable as a diagnostic tool to primary or specialist ferentiating between MCI and AD and predicting those who
practice but will continue to advance research understand- might progress from MCI to AD. Future standardization of
ing of brain function. MRS variables and neuroanatomic regions of study will
allow comparability between studies.
5. MRS
MRS is an application of MR imaging that can measure 6. Summary
the concentration of cerebral metabolites of the brain in
vivo. Although various nuclei can be used, the hydrogen Of the four functional neuroimaging modalities re-
proton 1H and phosphorus 31P are the nuclei that are in high viewed, SPECT is the most available to family physicians
enough concentration to measure routinely. 1H is most fre- and specialists. It might provide added value in patients with
quently used in MRS for cognitive research because it comorbid symptoms of vascular disease or who do not fit
provides the most information on metabolic markers of the clinical criteria for dementia. Amyloid ligand binding in
neurons, myelin, and glia and also has higher sensitivity and SPECT is still speculative. PET scanning for dementia di-
spatial resolution [33–36]. Compounds with a concentration agnosis is not available for clinical use in Canada except to
of at least 0.5 to 1.0 mmol that are commonly measured a very limited degree, but its utility clinically in the United
with the standard 1.5 T strength magnet include N-acetyl States will be followed with interest. It has also been
aspartate (NAA), myoinositol (MI), choline, and creatine. encouraged for use in clinical trials. MRS and fMRI
NAA, an amino acid present only in the central nervous continue in the research arena and hold considerable
system neurons of adults, reflects neuronal density or neu- promise in being able to predict progression from MCI to
ronal viability. It is reduced in a number of disease states, dementia, determine response to research compounds,
including degenerative central nervous system diseases. and are useful in hypothesis generation. In combination
MI is a component of membrane lipids and is considered with structural MRI and neuropsychological tests, func-
a glial cell marker. Choline is thought to reflect cellular tional neuroimaging holds promise for an earlier diagno-
density. Creatine is an osmolyte and has been used as a sis of cognitive impairment.
“constant” internal reference, although its constancy has
been questioned [34].
Higher magnetic field MR systems at 3 and 4 T strength 7. Recommendations approved at the 3rd
(even up to 7 T) have been used for human studies, provid- CCCDTD Conference
ing better signal/noise and contrast/noise ratios and im-
proved resolution [33,37]. Most studies in AD have shown
a reduction in NAA. 1. There is fair evidence that functional imaging with
Studies of brain regions affected early in AD have shown PET or SPECT scanning might assist specialists in the
reductions in NAA [37– 41], whereas MI has been shown to differential diagnosis of dementia, particularly those
increase [37,38,40,42]. Also, correlations between NAA with questionable early stage dementia or those with
measured in the MTL and cognitive measures have been FTD. There is variability across centers, with requi-
reported in AD [41,43]. NAA and MI ratios show differ- site expertise in these modalities that needs to be
ences between normal elderly controls, subjects with MCI, taken into account in determining utility (Grade B,
and AD with NAA/MI decreased in AD, with MCI mean Level 2).
levels falling between controls and AD [37,44]. 2. fMRI and MRS scanning are not recommended for
An MCI longitudinal study, mean period of 3 years, use by family physicians or specialists to make or
measuring occipital cortex NAA/creatine ratios predicted differentiate a diagnosis of dementia in people pre-
dementia at 100% sensitivity and 75% specificity (area senting with cognitive impairment. They remain very
under the curve, 0.91; 95% CI, 0.80 to 0.97) [45]. Surpris- promising research tools (Grade D, Level 3).
M. Borrie / Alzheimer’s & Dementia 3 (2007) 336 –340 339

Author Disclosures [13] Morano GN and Seibyl JP. Technical overview of brain SPECT
imaging: Improving acquisition and processing data. J Nucl Med
Michael Borrie has received support from Pfizer Canada Technol 2003;31:191–5.
(Medical Advisory Board Member, Speaker, Clinical Trial [14] Small GW, Rabins PV, Barry PP, Buckholtz NS, DeKosky ST, Ferris
Investigator), Janssen Ortho Inc. (Medical Advisory Board SH, et al. Diagnosis and treatment of Alzheimer disease and related
disorders: Consensus statement of the American Association for Ge-
Member, Speaker, Clinical Trial Investigator), Novartis
riatric Psychiatry, the Alzheimer’s Association, and the American
(Medical Advisory Board Member, Speaker, Clinical Trial Geriatrics Society. JAMA 1997;278:1363–71.
Investigator), Lundbeck Canada (Medical Advisory Board [15] Dougall NJ, Bruggink S, Ebmeier KP. Systematic review of the
Member, Speaker, Clinical Trial Investigator), Neurochem diagnostic accuracy of 99mTc-HMPAO-SPECT in dementia. Am J
(Clinical Trials Investigator), Sanofi Aventis (Clinical Tri- Geriatr Psychiatry 2004;12:554 –70.
als Investigator), GlaxoSmithKline (Clinical Trials Investi- [16] Bonte FJ, Weiner MF, Bigio E, White CL. Brain blood flow in the
dementias: SPECT with histopathologic correlation in 54 patients.
gator), ONO (Clinical Trials Investigator), Myriad (Clinical
Radiology 1997;202:793–7.
Trials Investigator), and HMR (Clinical Trials Investigator). [17] Jagust W, Thisted R, Devous MD Sr, Van Heertum R, Mayberg H,
Jobst K, et al. SPECT perfusion imaging in the diagnosis of
Alzheimer’s disease: a clinical-pathologic study. Neurology 2001;
Acknowledgments 56:950 – 6.
[18] Johnson KA, Jones K, Holman BL, Becker JA, Spiers PA, Satlin A,
The author would like to acknowledge the contributions et al. Preclinical prediction of Alzheimer’s disease using SPECT.
to the writing of this paper of Matthew Smith, Research Neurology 1998;50:1563–71.
Coordinator; Howard Chertkow, MD FRCP(C); and Judy [19] Kogure D, Matsuda H, Ohnishi T, Asada T, Uno M, Kunihiro T, et al.
Longitudinal evaluation of early Alzheimer’s disease using brain
McCallum, Administrative Assistant.
perfusion SPECT. J Nucl Med 2000;41:1155– 62.
[20] Celsis P, Agniel A, Cardebat D, Demonet JF, Ousset PJ, Puel M.
Age-related cognitive decline: A clinical entity? A longitudinal study
References of cerebral blood flow and memory performance. J Neurol Neurosurg
Psychiatry 1997;62:601– 8.
[1] Petrella JR, Coleman RE, Doraiswamy PM. Neuroimaging and early [21] Scheltens P, Launer LJ, Barkhof F, Weinstein HC, Jonker C. The
diagnosis of Alzheimer disease: A look to the future1. Radiology diagnostic value of magnetic resonance imaging and technetium 99m-
2003;226:315–36. HMPAO single-photon-emission computed tomography for the diag-
[2] Mosconi L. Brain glucose metabolism in the early and specific diag- nosis of Alzheimer disease in a community-dwelling elderly popula-
nosis of Alzheimer’s disease. FDG-PET studies in MCI and AD. Eur tion. Alzheimer Dis Assoc Disord 1997;11:63–70.
J Nucl Med Mol Imaging 2005;32;486 –510. [22] McKelvey R, Bergman H, Stern J, Rush C, Zahirney G, Chertkow H.
[3] Zakzanis KK, Graham SJ, Campbell Z. A meta-analysis of structural Lack of prognostic significance of SPECT abnormalities in non-
and functional brain imaging in dementia of the Alzheimer’s type: a demented elderly subjects with memory loss. Can J Neurol Sci 1999;
neuroimaging profile. Neuropsychology Review 2003;13;1. 26:23– 8.
[4] Wolf H, Jelic V, Gertz HJ, Nordberg A, Julin P, Wahlund LO. A [23] Smith SM. Overview of fMRI analysis. Br J Radiol 2004;77:S167–
critical discussion of the role of neuroimaging in mild cognitive S75.
impairment. Acta Neurol Scand Suppl 2003;107:52–76. [24] Golby A, Silverberg G, Race E, Gabrieli S, O’Shea J, Knierim K, et
[5] de Leon MJ, Convit A, Wolf OT, Tarshish CY, DeSanti S, Rusinek al. Memory encoding in Alzheimer’s disease: an fMRI study of
H, et al. Prediction of cognitive decline in normal elderly subjects explicit and implicit memory. Brain 2005;128:773– 87.
with 2-[18F]fluoro-2-deoxy-D-glucose/positron-emission tomography [25] Dickerson BC, Salat DH, Bates JF, Mtiya M, Killiany RJ, Greve DN,
(FDG/PET). Proc Natl Acad Sci U S A 2001;98:10966 –71. et al. Medical temporal lobe function and structure in mild cognitive
[6] De Santi S, de Leon MJ, Rusinek H, Convit A, Tarshish CY, Roche
impairment. Ann Neurol 2004;56:27–35.
A, et al. Hippocampal formation glucose metabolism and volume
[26] Rombouts SARB, Barkhof F, Goekoop R, Stam CJ, Scheltens P.
losses in MCI and AD. Neurobiol Aging 2001;22:529 –39.
Altered resting state networks in mild cognitive impairment and mild
[7] Nestor PJ, Fryer TD, Smielewski P, Hodges JR. Limbic hypometab-
Alzheimer’s disease: An fMRI study. Hum Brain Mapp 2005;26:
olism in Alzheimer’s disease and mild cognitive impairment. Ann
231–9.
Neurol 2003;54:343–51.
[27] Yetkin FZ, Rosenberg RN, Weiner MF, Purdy PD, Cullum CM.
[8] Patwardhan MB, McCrory DC, Matchar DB, Samsa GP, Rutschmann
OT. Alzheimer disease: Operating characteristics of PET - A meta- FMRI of working memory in patients with mild cognitive impairment
analysis. Radiology 2004;231:73– 80. and probably Alzheimer’s disease. Eur Radiol 2006;16:193–206.
[9] Mosconi L, De Santi S, Li Y, Li J, Zhan J, Tsui WH, et al. Visual [28] Rombouts SARB, Goekoop R, Stam CJ, Barkhof F, Scheltens P.
rating of medial temporal lobe metabolism in mild cognitive impair- Delayed rather than decreased BOLD response as a marker for early
ment and Alzheimer’s disease using FDG-PET. Eur J Nucl Med Mol Alzheimer’s disease. NeuroImage 2005;26:1078 – 85.
Imaging 2006;33:210 –21. [29] Rombouts SARB, van Swieten JC, Pijnenburg YAL, Goekoop R,
[10] Nordberg A. PET imaging of amyloid in Alzheimer’s disease. Lancet Barkhof F, Scheltens P. Loss of frontal fMRI activation in early
Neurol 2004;3:519 –27. frontotemporal dementia compared to early AD. Neurology 2003;60:
[11] Verhoeff N, Wilson AA, Takeshita S, Trop L, Mussey D, Singh K, et al. 1904 – 8.
In-Vivo imaging of Alzheimer disease ␤-Amyloid with [11C]SB-13 [30] Goekoop R, Rombouts SARB, Jonker C, Hibbel A, Knol DL, Truyen
PET. AM J Geriatr Psychiatry 2004;12:584 –95. L, et al. Challenging the cholinergic system in mild cognitive impair-
[12] Wang Y, Klunk WE, Debnath ML, Huang GF, Holt DP, Shao L, et ment: a pharmacological fMRI study. NeuroImage 2004;23:1450 –9.
al. Development of a PET/SPECT agent for amyloid imaging in [31] Kircher TTJ, Erb M, Grodd W, Leube DT. Cortical activation during
Alzheimer’s disease. J Mol Neurosci. 2004;24:55– 62. cholinesterase-inhibitor treatment in Alzheimer disease. Preliminary
340 M. Borrie / Alzheimer’s & Dementia 3 (2007) 336 –340

findings from a pharmaco-fMRI study. Am J Geriatr Psychiatry [40] Kantarci K, Jack CR, Jr. Xu YC, Campeau NG, O’Brien PC, Smith
2005;13:1006 –13. GE, et al. Regional metabolic patterns in mild cognitive impairment
[32] Bondi MW, Houston WS, Eyler LT, Brown GG. fMRI evidence of and Alzheimer’s disease: a 1H MRS study. Neurology 2000;55:
compensatory mechanisms in older adults at genetic risk for Alzhei- 210 –7.
mer disease. Neurology 2005;64:501– 8. [41] Jessen F, Block W, Traber F, Keller E, Flacke S, Papassotiropoulos
[33] Di Costanzo A, Trojsi F, Tosetti M, Giannatempo GM, Nemore F, A, et al. Proton MR spectroscopy detects a relative decrease of
Piccirillo M, et al. High-field proton MRS of human brain. Eur J N-acetyl aspartate in the medial temporal lobe of patients with AD.
Radiol 2003;48:146 –53. Neurology 2000;55:684 – 8.
[34] Ross AJ and Sachdev PS. Magnetic resonance spectroscopy in cog- [42] Rose SE, de Zubicaray GI, Greig I, Wang D, Galloway GJ, Chalk JB,
nitive research. Brain Research Reviews 2004;44:83–102. et al. A 1H MRS study of probable Alzheimer’s disease and normal
[35] Gujar SK, Maheshwari S, Bjorkman-Burtscher I, Sundgren PC. Mag- aging: implications for longitudinal monitoring of dementia progres-
netic resonance spectroscopy. J Neuro-Ophthalmol 2005;25:217–26. sion. Magn Reson Imaging 1999;17:291–9.
[36] Mason GF. Magnetic resonance spectroscopy for studies of neuro- [43] Chantal S, Labelle M, Bouchard RW, Braun CM, Boulanger Y.
transmission in vivo. Psychopharmacol Bull 2003;37:26 – 40. Correlation of regional proton magnetic resonance spectroscopic met-
[37] Kantarci K, Reynolds G, Petersen RC, Boeve BF, Knopman DS, abolic changes with cognitive deficits in mild Alzheimer disease.
Edland SD, et al. Proton MR spectroscopy in mild cognitive impair- Arch Neurol 2002;59:955– 62.
ment and Alzheimer disease: Comparison of 1.5 and 3 T. Am J [44] Catani M, Cherubini A, Howard R, Tarducci R, Pelliccioli GP,
Neuroradiol 2003;24:843–9. Piccirilli M, et al. 1H-MR spectroscopy differentiates mild cognitive
[38] Parnetti L, Tarducci R, Presciutti O. Proton magnetic resonance impairment from normal brain aging. NeuroReport 2001;12:2315–7.
spectroscopy can differentiate Alzheimer’s disease from normal ag- [45] Modrego PJ, Fayed N, Pina MA. Conversion from mild cognitive
ing. Mech. Ageing Dev 1997;97:9 –14. impairment to probable Alzheimer’s disease predicted by brain mag-
[39] Frederick BB, Satlin A, Yurgelun-Todd DA, Renshaw PF. In vivo netic resonance spectroscopy. Am J Psychiatry 2005;162:667–75.
proton magnetic resonance spectroscopy of Alzheimer’s disease in [46] Spencer DC, Zitzelberger T, Spielman D, Kaye J. MRS in relation to
the parietal and temporal lobes. Biol Psychiatry 1997;42:147–50. hippocampal volume in the oldest old. Neurology 2003;60:1194 – 6.
Alzheimer’s & Dementia 3 (2007) 341–347

General risk factors for dementia: A systematic evidence review


Christopher Pattersona,*, John Feightnerb, Angeles Garciac, Chris MacKnightd
a
Division of Geriatric Medicine, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
b
Department of Family Medicine, Faculty of Medicine, University of Western Ontario, London, Ontario, Canada
c
Department of Medicine (Geriatrics), Queen’s University, Kingston, Ontario, Canada
d
Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Abstract This review identifies and quantifies general (ie, nongenetic) risk factors for all-cause dementia,
Alzheimer’s disease, and vascular dementia specifically.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Alzheimer’s disease; Vascular dementia; Risk factors

1. Background reason, the present systematic review focuses solely on


longitudinal cohort studies.
Since the Canadian Consensus Conference on Demen-
We posed the question “What modifiable risk factors are
tia was convened in 1999, many advances have been
associated with dementia, Alzheimer’s disease, or vascular
made in the understanding of dementing disorders. One
dementia?” A comprehensive electronic literature search
area that was considered worthy of further investigation
was performed on Medline and EMBASE from 1966 to
was that of risk factors for dementia, because knowledge
December 2005. The search string is listed in Appendix 1.
of these risk factors has direct relevance to the primary
This search identified 3,424 articles. It was possible to reject
prevention of these disorders. The topic of preventing
1,705 of these as obviously irrelevant. The remaining 1,719
dementia was previously addressed at the Canadian Con-
references with their abstracts were reviewed by a single
sensus Conference on Dementia [1], and an update on
primary prevention appears as a companion piece to the reader (C.P.), who identified possibly relevant articles and
present article [2]. set aside the remainder. These latter references and abstracts
were then reviewed by three other readers (J.F., A.G., C.M.)
to ensure that no possibly relevant articles had been dis-
2. Methods carded. The following criteria were used for relevance: (1)
longitudinal cohort studies; (2) a population broadly repre-
The identification and relative importance of risk factors
sentative of Canadian demographics; (3) dementia, Alzhei-
are best addressed through longitudinal cohort studies. Al-
mer’s disease, or vascular dementia as outcome; and (4)
though case-control studies can be used to explore risk
general risk factors identified (eg, hypertension, educational
factors for disease, significant differences have been ob-
status, occupation, chemical exposure.) Articles addressing
served when conclusions of case-controlled studies are
genetic risk factors were excluded.
compared with longitudinal studies. An example of differ-
After assembling all possibly relevant publications, the
ent conclusions is found with tobacco, where case-control
full articles were obtained, and each was assessed indepen-
studies have suggested that tobacco is associated with a
dently by two reviewers for quality. Additional articles from
lower risk of dementia, whereas numerous longitudinal
personal files and those identified by scrutiny of bibliogra-
studies have shown that the risk is increased [3]. For this
phies of retrieved articles were added to the pool for quality
assessment. After consulting several key references on ap-
praisal of risk factor literature, the following criteria were
*Corresponding author. Tel.: (905) 521-7931; Fax: (905) 521-7905. chosen to determine whether an article was good, fair, poor,
E-mail address: pattec@hhsc.ca or ineligible.
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.001
342 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347

2.1. Population characteristics (DBP) levels have been implicated in increased risks of
subsequent dementia. For example, from the Kungsholmen
Inclusion criteria were defined, including ages, locations, project, SBP in excess of 180 mm Hg is associated with an
dates; exclusion of dementia at inception; and population increased RR of all-cause dementia (ACD) of 1.6 and for
broadly representative of Canadian demographics. Alzheimer’s disease (AD) of 1.5 after 6 years [4]. However,
2.2. Follow-up in the same study population, a SBP of less than 140 mm Hg
was also associated with increased risk of ACD (RR, 1.9)
All participants were accounted for at end of follow-up, and AD (RR, 2.2) [5]. From the combined results of the
without excessive “preventable” losses. No differences in Rotterdam and Gothenberg H-70 studies in individuals who
follow-up were found between those developing dementia are taking antihypertensive agents, each increase of 10
and those with no dementia. mm Hg in SBP appears to lower the RR of ACD (0.93) and
2.3. Exposures (risk factors) AD (0.89) [6]. A longitudinal study in Finland reported an
increased relative risk of AD of 2.3 in the long term after
List of risk factors was considered. Exposures were mea- elevated SBP [7]. However, a decline in SBP of 15 or more
sured in the same way for those with and without dementia. mm Hg appears to predate the onset of both ACD and AD,
according to data from the Kungsholmen project [8]. From
2.4. Outcomes
the same study group a DBP of less than 65 mm Hg is also
Dementia and subtypes were defined by standardized associated with a RR of 1.54 for ACD and 1.7 for AD, as
criteria. Ascertainment of outcome was independent of ex- does a pulse pressure of less than 70 mm Hg [9]. Thus, it
posure status. would appear that increased SBP increases the risk of sub-
sequent ACD, AD, and vascular dementia (VaD) in women,
2.5. Analysis but so apparently does reduced systolic, diastolic, and pulse
Important confounders were included in analysis (age, pressure below normal levels, overall a somewhat contra-
sex, education at minimum). Statistical analysis was appro- dictory and confusing situation.
priate (ie, multiple logistic regression). Specific measures of
risk are stated (eg, relative risk). 4.2. Diabetes mellitus
The overall quality rating was determined as good (all Several studies have linked the presence of type 2 dia-
criteria fulfilled adequately); fair (most criteria fulfilled ad- betes mellitus to subsequent development of dementia.
equately, no fatal flaw); or poor (fatal flaw or multiple minor Analysis of data from the Canadian Study of Health and
inadequacies). Each article was reviewed for quality inde- Aging (CSHA) concluded that diabetes increased the risk of
pendently by two readers, and a consensus was reached if ACD and AD, although the increased risks did not reach
disagreement occurred. Articles graded as good or fair were statistical significance. For VaD, however, the RR was 2.03
then submitted for data abstraction by two independent [10]. Diabetes in midlife was shown to increase the subse-
reviewers. quent risk of ACD (OR, 2.83) in a stratified sample of civil
servants from Tel Aviv [11]. From the Kungsholmen Study
3. Results in Sweden, Xu et al [12] concluded that there was an
increased risk of ACD and VaD (HR, 1.5 and 2.6, respec-
Risk factors are either immutable (eg, age, gender, eth- tively), although the increased risk for AD did not reach
nicity) or potentially modifiable. This article deals only with statistical significance. Data collected within a large ran-
potentially modifiable risk factors. A total of 60 articles of domized controlled trial of raloxifene suggested an in-
single longitudinal studies were identified to be of good or creased risk of ACD in those with diabetes mellitus, but the
fair quality. In addition, eight systematic reviews of good or increased OR of 2.38 did not reach statistical significance
fair quality also addressed relevant risk factors. Table 1 [13]. Thus, there is fair evidence that the presence of type 2
summarizes risk factors and their relative risk (RR) for diabetes mellitus increases the risk of ACD and VaD.
dementia of different types. Other measures of risk (eg,
odds ratio [OR], hazard ratio [HR]) are indicated where 4.3. Stroke
necessary.
The presence of clinical strokes or of silent infarctions on
neuroimaging increases the risk of subsequent dementia.
4. Potentially modifiable risk factors The temporal relationship between stroke and onset of de-
4.1. Blood pressure mentia is a diagnostic criterion for VaD. Longitudinal stud-
ies have shown that stroke increases the subsequent risk of
The relationship between blood pressure and subsequent ACD (RR, 2.4) [14] and AD (RR, 1.83) [15]. The presence
development of dementia is somewhat confusing. Both of silent brain infarctions visible on magnetic resonance
higher and lower systolic (SBP) and diastolic blood pressure imaging scanning is associated with an increased risk of
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347 343

Table1
Risk factors for dementia: RR (95% CI) [references]
ACD AD VAD

Systolic hypertension 1.6 (1.1 to 2.2) [4] RR, 1.5 (1.0 to 2.3) [4]; 2.05 (1.2 to 3.53) [25],
OR, 2.3 (1.0 to 5.5) [7] women only
DBP ⬍ 65 mm Hg 1.5 (1.0 to 2.1) [4] 1.7 (1.1 to 2.4) [4]
PP ⬎ 84 mm Hg 1.3 (0.9 to 1.7) [9] 1.4 (1.0 to 2.0) [9]
PP ⬍ 70 mm Hg 1.4 (1.0 to 1.9) [9] 1.7 (1.2 to 2.3) [9]
SBP decline ⱖ 15 mm Hg 3.1 (1.5 to 6.3) [8] 3.1 (1.3 to 7.0) [8]
SBP ⬍ 140 mm Hg 1.9 (1.2 to 3.2) [5] 2.2 (1.2 to 3.8) [5]
Diabetes mellitus, type 2 1.26 (0.9 to 1.76) [10] 1.3 (0.83 to 2.03) [10] 2.03 (1.15 to 3.57) [10]
2.38 (0.92 to 6.12) [13] 1.3 (0.9 to 2.1) [12] HR, 2.6 (1.2 to 6.1) [12]
2.83 (1.4 to 5.71) [11]
Stroke 2.4 (1.6 to 3.7) [14] 1.83 (1.14 to 2.95) [15] Required for diagnosis
Silent infarcts on neuroimaging 2.17 (1.04 to 4.54) [18]
HR, 2.26 (1.09 to 4.70) [16]
Serum cholesterol 1 3.1 (1.5 to 6.3) [19] 2.1 (1.0 to 4.4) [7]
3.1 (1.2 to 8.5) [19]
Serum homocysteine 1 2.08 (1.31 to 3.30) [20] 2.11 (1.19 to 3.76) [20]
Serum thyroid-stimulating hormone 2 3.5 (1.1 to 11.5) [21]
Sex hormones: higher total estrogen 1.45 (1.08 to 1.94) [22] 1.30 (0.88 to 1.99) [22]
level (women)
Sex hormones: higher free testosterone 0.74 (0.57 to 0.96) [23]
index (men)
Depression 1.87 (1.09 to 3.20) [26] Men: OR, 4.2 (2.1 to 8.8) OR, 2.41 (1.22 to 4.52) [25]
Men: OR, 3.5 (1.9 to 6.5) Women: OR, 1.0 (0.5 to 1.9) [24]
Women: OR, 1.2 (0.7 to 2.0) [24]
High fat intake 2.4 (1.1 to 5.4) [27]
Fish consumption ⬎ once/week 0.4 (0.2 to 0.9) [27] 0.3 (0.1 to 0.9) [27] Shellfish: OR, 0.45
0.73 (0.52 to 1.03) [28] (.22 to .88) [25]
Vitamin E: lowest tertile serum level 2.54 (1.06 to 6.10) [29] 2.10 (0.81 to 5.54) [29]
Moderate wine consumption, 250 to OR, 0.19 (.05 to 0.66) [30] 0.53 (0.3 to 0.95) [28]
500 mL/day 0.56 (0.36 to 0.92) [28]
Activity: highest level, physical 0.58 (0.41 to 0.83) [33] 0.5 (0.28 to 0.90) [32] OR, 0.46 (0.25 to 0.82),
0.63 (0.40 to 0.98) [32] 0.55 (0.34 to 0.88) [33] women [25]
0.69 (0.5 to 0.96) [31]
Activity: highest level, mental, daily 0.59 (0.37 to 0.96) [34]
Smoking 1.10 (0.94 to 1.29) [37]
1.99 (1.33 to 2.98) [3]
Occupation Manual work: 1.6 (1.0 to 2.5) [49] Manual work: 1.4 (0.9 to 2.1) [49]
Exposure to toxins (pesticides, 4.35 (1.05 to 17.90) [36] OR, 2.05 (1.03 to 3.85) [25]
fertilizers, fumigants), defoliants
Head injury With LOC: 1.14 (0.84 to 1.56) [37] With LOC: 1.02 (0.68 to 1.15) [37]
Moderate: HR, 2.39 (1.24 to 4.58) [38] Moderate: HR, 2.32 (1.04 to 5.1) [38]
Severe: HR, 4.48 (2.09 to 9.63) [38] Severe: HR, 4.51 (1.77 to 11.47) [38]
Education ⬎ 15 vs ⬍ 12 0.64 (0.4 to 1.0) [48] 0.48 (0.27 to 0.84) [48]
Statin drugs 1.19 (0.82 to 1.75) [41] 0.82 (0.46 to 1.46) [41]
HR, 1.19 (0.53 to 2.34) [40] HR, 1.19 (0.35 to 2.96) [40]
All lipid-lowering drugs OR, 0.26 (0.08 to 0.88) [39], age less
than 80 y
NSAIDs 0.51 (0.37 to 0.70) [44] 0.42 (0.26 to 0.66) [42]
Benzodiazepines Ever used: OR, 1.7 (1.2 to 2.4)
Former use: OR, 2.3 (1.2 to 4.5) [45]
Vaccination Any vaccination: 0.40
(0.17 to 0.96) [36]
Influenza: 0.75 (0.54 to 1.04)
Poliomyelitis: 0.6 (.37 to .99)
Diphtheria or tetanus: 0.41
(.27 to .62) [47]

Abbreviation: LOC, loss of consciousness.


344 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347

ACD (HR, 2.26) [16]. From a prospective study of stroke of eating fish regularly. In the PAQUID longitudinal study,
patients in hospital in Lille, France, in which prestroke weekly consumption of fish was associated with an RR of
cognitive function was assessed with the IQCODE [17], 0.73 for ACD, although this was of marginal significance
the presence of silent infarctions increased the risk of sub- [28]. Regular consumption of shellfish was also associated
sequent ACD (RR, 2.7) [18]. Thus, there is compelling with a reduced risk of VaD in the CSHA, with an OR of
evidence that the presence of strokes increases the risk of 0.45 [25]. There appears to be consistent epidemiologic
both ACD and VaD. evidence that regular consumption of fish and seafood is
associated with reduced risk of dementia. A single study
4.4. Serum cholesterol
reported an association between the lowest tertile of serum
Midlife elevation in serum cholesterol has been associ- vitamin E levels and elevated risk of both ACD and AD, but
ated with increased risk of subsequent AD (OR, 2.1) [7] and statistical significance was not reached [29].
(OR, 3.1) [19]. The risk of ACD was also elevated (RR, 3.1)
with higher levels of cholesterol [19]. There appears to be 4.9.2. Wine
good evidence that elevated serum cholesterol is associated The PAQUID study in France established that the con-
with an increased risk of ACD and AD. sumption of moderate amounts of red wine (250 to 500
mL/day) was associated with a RR of 0.56 for ACD and a
4.5. Hyperhomocysteinemia
RR of 0.53 for AD [28]. In the same study population,
A single prospective study has established an association moderate consumption, compared with other degrees of
between serum homocysteine levels in excess of 15 ␮mol/L usage, was associated with a lower risk of ACD (RR, 0.19)
and ACD (RR, 2.08) and AD (RR, 2.11; 95% confidence [30]. Furthermore, in the CSHA population, consumption of
interval [CI], 1.19 to 3.76) [20]. wine at least weekly was associated with a reduced risk of
AD (OR, 0.49), although this lost statistical significance
4.6. Hyperthyroidism
when decedents were included in the analysis [31].
A single study showed an association between a de-
pressed level of serum thyroid-stimulating hormone and 4.9.3. Activity
subsequent development of AD [21]. There is evidence that regular physical activity is asso-
ciated with a reduced risk of dementia. In the CSHA, reg-
4.7. Sex hormone levels
ular physical activity was associated with an OR of 0.69 for
In women, levels of total estrogen are associated with a AD [31], and when this was quantified, those with the
higher risk of ACD (RR, 1.45) [22], whereas in men, higher highest level of physical activity had a reduced risk of ACD
levels of free testosterone index are associated with lower (OR, 0.63) and of AD (OR, 0.5) [32]. In the Cardiovascular
risk of AD (RR, 0.74) [23]. Health Study from the USA, when those expending the
highest quartile of energy expenditure were compared with
4.8. Depression the lowest, the RR was 0.58 for ACD and 0.55 for AD [33].
The presence of depressive symptoms was associated Regular exercise was also associated with RR of VaD in the
with subsequent development of dementia only in men in CSHA; in women only, OR for VaD was 0.46 [25].
the PAQUID Study. For men, the OR of developing ACD Daily mental activities were associated with a RR of 0.59
was 3.5, and the RR for AD was 4.3. In women there was for ACD in the Kungsholmen Study [34]. In the Washington
no statistically significant association [24]. For VaD, data Heights District of New York, a longitudinal study found an
from the CSHA indicated that the presence of depression association between high leisure activities and decreased
increased the risk of VaD (OR, 2.41) [25]. A systematic incidence of ACD, with a RR of 0.62 [35]. It is thus
evidence review concluded that depression is associated plausible that maintaining a high level of mental activity
with an RR of 1.87 for ACD [26]. might be associated with reduced subsequent incidence of
ACD.
4.9. Lifestyle factors
4.9.1. Diet 4.9.4. Smoking
A high intake of total fat (⬎ 85.5 g/day) increases the RR A systematic review [3] compared the association of
for ACD by 2.4 compared with a fat intake of ⬍ 75.5 g/day smoking with dementia in case-controlled and prospective
[27], but in this longitudinal population study from the cohort studies. Although there appeared to be a protective
Netherlands, increased amounts of saturated fat and choles- effect in the case-controlled studies, the prospective cohort
terol were not established as definite risk factors. The con- studies established an increased risk for AD (RR, 1.99) in
sumption of fish greater than 18.5 g/day compared with less studies that described the number of smokers at baseline [3].
than 3.0 g/day lowered the risk of both ACD (RR, 0.4) and This should settle, once and for all, the positive association
AD (RR, 0.3). Other studies have also suggested the benefit between smoking and AD.
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347 345

4.9.5. Exposure to toxins tive studies concluded that increased risk of cognitive de-
Evidence from CSHA established an association be- cline was reported in three, decreased in two, and un-
tween exposure to pesticides or fertilizers and VaD (OR, changed in one [46].
2.05) [25]. Exposure to defoliants and fumigants was asso-
4.10.4. Vaccinations
ciated with an increased risk of AD (RR, 4.35) in a longi-
Prior inoculations against poliomyelitis, diphtheria, or
tudinal study from Manitoba, Canada [36].
tetanus and influenza are all associated with a lower risk of
4.9.6. Head trauma AD [47]. In addition, inoculations of any kind appear to
Four population-based samples comprised the EURODEM reduce AD risk [36].
Study. In this study head trauma with unconsciousness was
4.11. Education
associated with an increased RR of both ACD (1.14) and AD
(1.02); however, neither of these reached statistical signifi- Longer periods of education have been associated with
cance [37]. In the CSHA, prior head injury with or without reduced risk of AD, and this relationship has been con-
loss of consciousness did not show any increased risk of AD firmed. When compared with those receiving less than 12
(OR, 0.87; 95% CI, 0.56 to 1.36) [31]. However, in a years of education, people engaged in more than 15 years of
long-term study of U.S. servicemen who had been injured education had a reduced risk of both ACD (RR, 0.64) and
during World War II, moderate head injury was associated AD (RR, 0.48) [48].
with an HR of 2.39 for ACD and 2.32 for AD [38]. For
severe injury the HR for ACD was 4.48 and 4.51 for AD. A 4.12. Occupation
conclusive association between head injury and subsequent Data from the Kungsholmen project in Sweden sug-
dementia remains to be established. gested that manual labor increased the risk of both ACD
4.10. Drug exposure (RR, 1.6) and AD (RR, 1.4), although the latter just missed
statistical significance [49]. Other studies have not con-
4.10.1. Lipid-lowering agents firmed this relationship.
Although a nested case-control study within the CSHA
suggested that in individuals younger than 80 years of age,
5. Conclusion
use of all types of lipid-lowering agents reduced the risk of
AD, with an OR of 0.26 at 5 years [39], other studies have This systematic review has explored the current state of
not confirmed this finding. In the Cache County Study, the evidence concerning risk factors for ACD, AD, and VaD,
use of statin-type drugs was associated with a nonsignificant which are presented in Table 1. Although for many of these
increased HR of 1.19 for both ACD (95% CI, 0.53 to 2.34) risk factors the evidence remains inconclusive or contradic-
and AD (95% CI, 0.53 to 2.96) at 3 years [40]. There were tory, for others an emerging picture of consensus is appear-
similar findings in an American community-based study in ing. It should be noted that with rare exceptions, there are no
which use of statin agents was associated with a nonsignif- data on the effects of multiple risk factors in the same
icant increased HR of 1.19 (95% CI, 0.83 to 1.75) for ACD individual. One exception is a Finnish study in which the
but a nonsignificant reduced HR of 0.82 (95% CI, 0.46 to combined risk of systolic hypertension (OR, 2.3) and hy-
1.46) for AD [41]. percholesterolemia (OR, 2.1) increases the OR for AD to
3.5 [7]. Research priorities should focus on remediable risk
4.10.2. Nonsteroidal anti-inflammatory drugs
factors that have the highest chance of being corrected.
Individual studies such as the Cache County popula-
These include such risk factors as hypertension, hypercho-
tion have concluded that exposure to nonsteroidal anti-
lesterolemia, inadequate physical and mental activity, and
inflammatory drugs (NSAIDs) is associated with reduced
dietary factors. Attempts to limit the noxious effects of
risk of AD (RR, 0.42) [42]. A systematic review of six
smoking and head injury are justifiable for their own sake
longitudinal studies in 2003 concluded that there was a
and might also have benefits in the cognitive domain.
nonsignificant reduction in RR of 0.84 (95% CI, 0.54
to1.05) for AD [43]. However, a more recent and compre-
hensive systematic review of 25 studies, both cohort and Author Disclosures
case-controlled, concluded that NSAID exposure was asso-
ciated with a significantly reduced risk of both prevalent Angeles Garcia has received support from Janssen-Ortho
(RR, 0.51) and incident (RR, 0.79) cases of ACD [44]. (Consultant, Advisor), Pfizer (Consultant, Advisor), Novartis
(Consultant, Advisor), and Lundbeck (Consultant, Advisor).
4.10.3. Benzodiazepines Chris MacKnight has received support from Alzheimer
In a French population-based study, exposure to benzo- Society of Nova Scotia (Board Member), Janssen-Ortho
diazepine drugs at any time was associated with an in- (Speaker, Trial Investigator), Pfizer Canada (Speaker, Trial
creased risk of ACD (OR, 1.7), and for former use the OR Investigator), Novartis (Speaker, Trial Investigator), Lund-
was 2.3 [45]. However, a systematic review of six prospec- beck Canada (Speaker, Trial Investigator), Voyager Phar-
346 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347

maceuticals (Trial Investigator), Myriad (Trial Investiga- [17] Jorm AF, Scott R, Cullen JS, MacKinnon AJ. Performance of the
tor), and Neurochem (Trial Investigator). Informant Questionnaire on Cognitive Decline in the Elderly
(IQCODE) as a screening test for dementia. Psychol Med 1991;21:
785–90.
[18] Henon H, Durieu I, Guerouaou D, Lebert F, Pasquier F, Leys D.
Acknowledgments
Poststroke dementia: incidence and relationship to prestroke cogni-
During preparation of this article, Christopher Patterson tive decline. Neurology 2001;57:1216 –22.
[19] Notkola I-L, Sulkava R, Pekkanen J, Erkinjuntti T, Ehnholm C,
received financial support from the Institute of Advanced
Kivinen P, et al. Serum total cholesterol, apolipoprotein E ␧4 allele,
Studies, University of Bologna, Bologna, Italy. and Alzheimer’s disease. Neuroepidemiology 1998;17:14 –20.
Chris MacKnight is supported from a CIHR New Inves- [20] Ravaglia G, Forti P, Maioli F, Martelli M, Servadei L, Brunetti N,
tigator Award. et al. Homocysteine and folate as risk factors for dementia and
Alzheimer disease. Am J Clin Nutr 2005;82:636 – 43.
[21] Kalmijn S, Mehta KM, Pols HAP, Hofman A, Drexhage HA, Breteler
References MMB. Subclinical hyperthyroidism and the risk of dementia: the
Rotterdam Study. Clin Epidemiol 2000;53:733–7.
[1] Black SE, Patterson C, Feightner J. Preventing dementia. Can J Neu- [22] Geerlings MI, Launer LJ, de Jong FH, Ruitenberg A, Stijnen T, van
rol Sci 2001;28(Suppl 1):S56 – 66. Swieten JC, et al. Endogenous estradiol and risk of dementia in
[2] Patterson C, Feightner JW, Garcia A, MacKnight CR. Primary pre- women and men: the Rotterdam Study. Ann Neurol 2003;53:607–15.
vention of dementia. Alzheimers Dement 2007;3:. [23] Moffat SD, Zonderman AB, Metter EJ, Kawas C, Blackman MR,
[3] Almeida OP, Hulse GK, Lawrence D, Flicker L. Smoking as a risk Harman SM, et al. Free testosterone and risk for Alzheimer disease in
factor for Alzheimer’s disease; contrasting evidence from a system- older men. Neurology 2004;62:188 –93.
atic review of case-control and cohort studies. Addiction 2002;1997: [24] Fuhrer R, Cufouil C, Dartigues JF, for the Paquid Study. Exploring
15–28. sex differences in the relationship between depressive symptoms and
[4] Qui C, von Strauss E, Fastbom J, Winblad B, Fratiglioni L. Low dementia incidence: prospective results from the PAQUID Study.
blood pressure and risk of dementia in the Kungsholmen Project: a J Am Geriatr Soc 2003;51:1055– 63.
6-year follow-up study. Arch Neurol 2003;60:223– 8. [25] Hébert R, Lindsay J, Verreault R, Rockwood K, Hill G, Dubois M-F.
[5] Guo C, Viitanen M, Winblad B, Fratiglioni L. Low blood pressure Vascular dementia: incidence and risk factors in the Canadian Study
and incidence of dementia in a very old sample: dependent on initial of Health and Aging. Stroke 2000;31:1487–93.
cognition. J Am Geriatr Soc 1999;47:723– 6.
[26] Jorm AF. History of depression as a risk factor for dementia: an
[6] Ruitenberg A, Skoog I, Ott A, Aevarsson O, Witteman JCM, Lernfelt
updated review. Aust N Z J Psychiatry 2001;35:776 – 81.
B, et al. Blood pressure and risk of dementia: results from the
[27] Kalmijn S, Launer LJ, Ott A, Witteman JCM, Hofman A, Breteler
Rotterdam Study and the Tothenburg H-70 Study. Dement Geriatr
MMB. Dietary fat intake and the risk of incident dementia in the
Cogn Disord 2001;12:33–9.
[7] Kivipelto M, Helkala E-L, Laakso MP, Hänninen T, Hallikainen M, Rotterdam study. Ann Neurol 1997;42:776 – 82.
Alhainen K, et al. Midlife vascular risk factors and Alzheimer’s [28] Larrieu S, Letenneur L, Helmer C, Dartigues JF, Barberger-Gateau P.
disease in later life: longitudinal, population based study. BMJ 2001; Nutritional factors and risk of incident dementia in the PAQUID
322:1447–51. longitudinal cohort. J Nutr Health Aging 2004;8:150 – 4.
[8] Qui C, von Strauss E, Winblad B, Fratiglioni L. Decline in blood [29] Helmer C, Peuchant E, Letenneur L, Bourdel-Marchasson I, Larrieu
pressure over time and risk of dementia: a longitudinal study from the S, Dartiques JF, et al. Association between antioxidant nutritional
Kungsholmen project. Stroke 2004;35:1810 –5. indicators and the incidence of dementia: results from the PAQUID
[9] Qui C, Winblad B, Wiitanen M, Fratiglioni L. Pulse pressure and risk of prospective cohort study. Eur J Clin Nutr 2003;57:1555– 61.
Alzheimer disease in persons aged 75 years and older: a community- [30] Orgogozo J-M, Dartigues J-F, Lafont S, Letenneur L, Commenges D,
based, longitudinal study. Stroke 2003;34:594 –9. Salamon R, et al. Wine consumption and dementia in the elderly: a
[10] MacKnight C, Rockwood K, Awalt E, McDowell I. Diabetes mellitus prospective community study in the Bordeaux area. Rev Neurol
and the risk of dementia, Alzheimer’s disease and vascular cognitive 1997;153:3, 185–92.
impairment in the Canadian Study of Health and Aging. Dement [31] Lindsay J, Laurin D, Verreault R, Hébert R, Helliwell B, Hill GB,
Geriatr Cogn Disord 2002;14:77– 83. et al. Risk factors for Alzheimer’s disease: a prospective analysis
[11] Schnaider Beeri M, Goldbourt U, Silverman JM, Noy S, Schmeidler from the Canadian Study of Health and Aging. Am J Epidemiol
J, Ravona-Springer R, et al. Diabetes mellitus in midlife and the risk 2002;156:445–53.
of dementia three decades later. Neurology 2004;63:1902–7. [32] Laurin D, Verreault R, Lindsay J, MacPherson K, Rockwood K.
[12] Xu WL, Qiu CX, Wahlin A, Winblad B, Fratiglioni L. Diabetes
Physical activity and risk of cognitive impairment and dementia in
mellitus and risk of dementia in the Kungsholmen project: a 6-year
elderly persons. Arch Neurol 2001;58:498 –504.
follow-up study. Neurology 2004;63:1181– 6.
[33] Podewills LJ, Guallar E, Kuller LH, Fried LP, Lopez OL, Carlson M,
[13] Yaffe K, Blackwell T, Kanaya AM, Davidowitz N, Barrett-Connor E,
et al. Physical activity, APOE genotype and dementia risk: findings
Krueger K. Diabetes, impaired fasting glucose, and development of
cognitive impairment in older women. Neurology 2004;63:658 – 63. from the Cardiovascular Health Cognition Study. Am J Epidemiol
[14] Ivan CS, Seshadri S, Beiser A, Au R, Kase CS, Kelly-Hayes M, et al. 2005;161:639 –51.
Dementia after stroke: The Framingham Study. Stroke 2004;35:1264 – 8. [34] Wang H-X, Karp A, Winblad B, Fratiglioni L. Late-life engagement
[15] Honig LS, Tang M-X, Albert S, Costa R, Luchsinger J, Manley J, et in social and leisure activities is associated with a decreased risk of
al. Stroke and the risk of Alzheimer disease. Arch Neurol 2003;60: dementia: a longitudinal study from the Kungsholmen project. Am J
1707–12. Epidemiol 2002;155:1081–7.
[16] Vermeer SE, Prins ND, den Heijer T, Hofman A, Koudstaal PJ, [35] Scarmeas N, Levy G, Tang M-X, Manly J, Stern Y. Influence of
Breteler MMB. Silent brain infarcts and the risk of dementia and leisure activity on the incidence of Alzheimer’s disease. Neurology
cognitive decline. N Engl J Med 2003;348:1215–22. 2001;57:2236 – 42.
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 341–347 347

[36] Tyas LS, Manfreda J, Strain LA, Montgomery PR. Risk factors for [49] Qiu C, Karp A, von Strauss E, Winblad B, Fratiglioni L, Bellander T.
Alzheimer’s disease: a population-based, longitudinal study in Mani- Lifetime principal occupation and risk of Alzheimer’s disease in the
toba, Canada. Int J Epidemiol 2001;30:590 –7. Kungsholmen Project. Am J Ind Med 2003;43:204 –11.
[37] Launer LJ, Andersen K, Dewey ME, Letenneur L, Ott A, Amaducci
LA, et al. Rates and risk factors for dementia and Alzheimer’s
disease: results from EURODEM pooled analyses. Neurology 1999;
52:78 – 84. Appendix 1: Search strategies
[38] Plassman BL, Havlik RJ, Steffens DC, Helms MJ, Newman TN,
Drosdick D, et al. Documented head injury in early adulthood and
1966 to December 2005
risk of Alzheimer’s disease and other dementias. Neurology 2000;55:
1158 – 66. RF & Dementia or AD AND
[39] Rockwood K, Kirkland S, Hogan DB, MacKnight C, Merry H,
Verreault R, et al. Use of lipid-lowering agents, indication bias, and ((Relative [title/abstract] AND risk* [title/abstract]) OR
the risk of dementia in community-dwelling elderly people. Arch (relative risk [Text Word]) OR risks [Text word] OR Cohort
Neurol 2002;59:223–7.
[40] Zandi PP, Sparks L, Khachaturian AS, Tschanz J, Norton M, Stein- studies [MeSH: no exp] OR (cohort [Title/Abstract] AND
berg M, et al. Do statins reduce risk of incident dementia and Alz- stud* [Title/Abstract]) Limits – Abstracts, Review, Human
heimer disease? Arch Gen Psychiatry 2005;62:217–24.
[41] Li G, Higdon R, Kukull W, Peskind E, Van Valen Moore K, 1966 to December 2005
Tsuang D, et al. Statin therapy and risk of dementia in the elderly:
a community-based prospective cohort study. Neurology 2004;63:
((“risk factors” [MeSH Terms] OR risk factors {Text
1624 – 8. Word]) AND (“dementia” [MeSH Terms] OR dementia
[42] Zandi PP, Anthony JC, Hayden KM, Mehta K, Mayer L, Breitner [Text Word]) OR (((“alzheimer disease” [TIAB] NOT Med-
JCS, et al. Reduced incidence of AD with NSAID but not H2 receptor line [SB]) OR “alzheimer disease” [MeSH Terms] OR alz-
antagonists: the Cache County Study. Neurology 2002;59:880 – 6.
[43] Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflamma-
heimer [Text Word]) AND s [All Fields] AND (“disease”
tory drugs on risk of Alzheimer’s disease: systematic review and [MeSH Terms] OR disease [Text Word])) NOT (“therapy”
meta-analysis of observational studies. BMJ 2003;327:128. [Subheading] OR (“therapeutics” [TIAB] NOT Medline
[44] de Craen AJM, Gussekloo J, Vrijsen B, Westendorp RGJ. Meta- [SB]) OR “Therapeutics” [MeSH Terms] OR therapy [Text
analysis of nonsteroidal antiinflmmatory drug use and risk of demen-
tia. Am J Epidemiol 2005;161:114 –20.
Word])) AND systematic [sb] AND ((“humans” [TIAB]
[45] Lagnaoui R, Bégaud B, Moore N, Chaslerie A, Fourrier A, Letenneur NOT Medline [SB]) or “humans” [MeSH Terms] OR Hu-
L, et al. Benzodiazepine use and risk of dementia: a nested case- man [Text Word])
control study. J Clin Epidemiol 2002;55:314 – 8.
[46] Verdoux H, Lagnaoui R, Begaud B. Is benzodiazepine use a risk 1996 to 2006
factor for cognitive decline and dementia? a literature review of
epidemiological studies. Psychol Med 2005;35:307–15. (Prevention of Alzheimer*) AND systematic [sb] OR
[47] Verreault R, Laurin D, Lindsay J, De Serres G. Past exposure to (prevention of dementia) AND systematic [sb] OR (“risk
vaccines and subsequent risk of Alzheimer’s disease. CMAJ 2001; factors AND (dementia OR alzheimer*)) AND systematic
165:1495– 8.
[48] Kukull WA, Higdon R, Bowen JD, McCormich WC, Teri L, Schel-
[sb] All fields, publication date from 1996 to 2006, core
lenberg GD, et al. Dementia and Alzheimer disease incidence: a clinical journals OR (”risk factors AND (dementia OR Alz-
prospective cohort study. Arch Neurol 2002;59:1737– 46. heimer*)) AND systematic [sb]
Alzheimer’s & Dementia 3 (2007) 348 –354

Primary prevention of dementia


Christopher Pattersona,*, John Feightnerb, Angeles Garciac, Chris MacKnightd
a
Division of Geriatric Medicine, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
b
Department of Family Medicine, Faculty of Medicine, University of Western Ontario, London, Ontario, Canada
c
Department of Medicine (Geriatrics), Queen’s University, Kingston, Ontario, Canada
d
Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada

Abstract The purpose of this article is to recommend strategies to practicing physicians for the prevention
of dementia in people without preexisting cognitive deficits.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Alzheimer’s disease; Vascular dementia; Primary prevention

1. Background 2. Methods
In preparation for this article, a systematic evidence Studies addressing prevention of dementia were identi-
review was undertaken to determine the risk factors for fied from three sources. First, from the original search for
all-cause dementia (ACD), Alzheimer’s disease (AD), or risk factors performed in December 2005 (total of 3,424
vascular dementia (VaD) [1]. This review involved litera- articles with abstracts); second, from a targeted search for
ture from 1996 until December 2005; 1996 was chosen as RCTs and systematic evidence reviews in February 2006
the initial date because literature up until 1998 had been (see Appendix 1 for search strategy); and third, articles
reviewed in preparation for an article by Black et al [2] on gleaned from bibliographies of retrieved articles and authors
“Preventing Dementia” for the 2nd Canadian Consensus files.
Conference on Dementia. The new review addresses poten- Quality assessment of RCTs was performed with criteria
tially modifiable risk factors. Genetic factors are considered of the Canadian Task Force on Preventive Health Care and
in another article in this series [3]. resulted in designation of good (all criteria fulfilled); fair
Risk factors that are potentially remediable are listed in (minor flaws only, no fatal flaw), and poor (fatal flaw or
Table 1 and form the background for this article on primary multiple minor flaws) [4]. For systematic reviews, criteria
prevention. The relative risk (RR) for ACD, AD, or VaD for described by Hunt and McKibbon [5] were used for quality
each potentially remediable risk factor is included in Table 1. assessment.
The table shows statistically significant RR values without Where conflicting evidence exists, priority is given to
95% confidence intervals, which are detailed in the risk studies (1) with higher levels of evidence, (2) with better
factor article [1]. The presence of a potentially remediable quality, and (3) most contemporary. For example, in the
risk factor does not imply that modification of that risk case of estrogens, where several systematic evidence re-
factor will influence the subsequent development of demen- views of epidemiologic and case-controlled trials suggested
tia. Only randomized controlled trials (RCTs) are consid- that estrogens were protective against dementia (Level 2
ered to provide strong evidence for efficacy of risk factor evidence), conclusions are superseded by a large RCT in-
modification (Tables 2 and 3). dicating an increased risk of dementia (Level 1 evidence)
[6]. We found very few RCTs of interventions to prevent
dementia on which to base firm conclusions, and maintain-
ing this rigorous standard of evidence leads to a large
*Corresponding author. Tel.: (905) 521-7931; Fax: (905) 521-7905. number of “C” grade recommendations, where there is in-
E-mail address: pattec@hhsc.ca sufficient or contradictory evidence of efficacy.

1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.005
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354 349

Table 1
Potentially modifiable risk factors for dementia: RR is shown except where alternative risk measures are used: hazards ratio (HR); odds ratio (OR)
Illnesses and Conditions ACD AD VaD

Hypertension 1.6 1.5 to 2.3 (OR) 2.05


Diabetes mellitus 2.83 2.03 2.6 (HR)
Stroke 2.4 1.83 Required for diagnosis
Silent infarctions on neuroimaging 2.17 to 2.26 (HR) 1.83
Reduced thyroid-stimulating hormone 3.5
Hypercholesterolemia 3.1 2.1 to 3.1
Hyperhomocysteinemia 2.08 2.11
Depression 1.87 4.2 (men) 2.41 (OR)
Lifestyle
High fat diet 2.4
High omega 3 fatty acid./fish in diet 0.4 0.3 0.45 (OR)
Moderate consumption of wine 0.19 (OR) to 0.56 0.53
Activity, highest level, physical 0.58 to 0.63 0.5 to 0.69 0.46 (OR)
Activity, higher level, mental 0.59
Smoking 2.2 1.99 to 2.3
Occupation (manual work) 1.6
Exposure to toxins 4.35 2.05 (OR)
Medications
3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) 0.33 to 1.19 (HR) 0.82 to 1.19 (HR)
All lipid-lowering agents 0.26 (OR)
NSAIDs 0.51 0.42
Lowest tertile serum vitamin E level 2.54 2.10
Benzodiazepines 1.7 to 2.3 (OR)
Vaccinations 0.4
Antihypertensives 0.93 0.89
Miscellaneous
Head injuries (serious) 2.39 to 4.48 (HR) 2.32 to 4.51 (HR)
Education (⬎15 y vs ⬍12 y) 0.64 0.48

Abbreviations: ACD, any cause dementia; AD, Alzheimer’s disease; VaD, vascular dementia.

3. Hypertension though it is tempting to assume that a reduced incidence of


The association between elevated systolic blood pressure stroke resulting from antihypertensive treatment will trans-
and stroke is well-established. However, the relationship late into a lower risk of dementia, this is not borne out by
between hypertension and dementia is somewhat less clear. evidence from all RCTs.
Both elevated systolic blood pressure and depressed dia- In 1998 the results of the Systolic Hypertension in Eu-
stolic blood pressure in middle age are positively correlated rope (SYST-Eur) trial were published [7]. Three thousand
with the later development of dementia [1]. In the years one hundred sixty-two patients older than the age of 60
preceding the onset of dementia, systolic blood pressure years with seated systolic blood pressures of 160 to 219 mm
might fall [1]. Furthermore, not all epidemiologic studies Hg and diastolic blood pressures below 95 mm Hg were
confirm the positive association between elevated systolic randomized to receive nitrendipine 10 to 40 mg/day with the
pressure and subsequent development of dementia. Al- addition of enalapril and/or hydrochlorothiazide titrated to

Table 2
Quality assessment for individual RCTs
Reference Adequacy of Comparison of Placebo Groups Treated All Subjects Dementia Outcome Analysis Quality
Randomization and Treatment Groups Equally Accounted for Assessed Blindly Appropriate
at Conclusion

Forette et al [7], 1998 Yes Equal Yes Yes Yes Yes Good
PROGRESS [8], 2003 Yes Equal Yes Yes Yes Yes Good
Shepherd et al [14], 2002 Yes Equal Yes Yes Yes Yes Good
Shumaker et al [6], 2004 Yes More in placebo group Yes Yes Yes Yes Good
aged ⬎75 y; more in
estrogen group were
hypertensive
350 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354

Table 3
Results of individual RCTs
Reference Population N Intervention Duration Results
(years)

Forette et al [7], 1998 Men and women ⬎60 y; seated 2418 1238 nitrendipine 10–40 mg/day ⫾ 2.0 Treated group had lower risk of
systolic blood pressure 160 enalapril 5–20 mg/day ⫾ ACD; 7.7–3.8/1000 patient-y;
to 219 mm Hg; diastolic hydrochlorothiazide 12.5–25 mg/ 50% reduction; P ⫽ .05
blood pressure ⬍95 mm Hg; day 1180 placebo
dementia excluded at
baseline
PROGRESS [8], 2003 Men and women with transient 6105 3051 perindopril ⫾ indapamide 3.9 12% lower risk of ACD (⫺8%
ischemic attack or stroke 3054 placebo to 28%); 34% lower risk of
within 5 y dementia with recurrent
stroke (3% to 55%)
Shepherd et al [14], 2002 Men and women aged 70 to 5804 2888 prescribed pravastatin 40 mg/ 3.2 No significant difference in
82 y with increased risk or day 2912 placebo cognitive function between
preexisting vascular disease treatment and placebo groups
Shumaker et al [6], 2004 Post menopausal women 65 to 2947 1464 estrogen alone 5.2 HR for probable ACD
75 y prior hysterectomy 1483 placebo 1.4 (0.83, 2.66)
Post menopausal women 65 to 4532 2229 estrogen & progestin 4.0 HR for probable ACD
75 y no prior hysterectomy 2.05 (1.21, 3.48)
Pooled 7479 Pooled 4.5 HR for probable ACD
1.76 (1.19, 2.60)

reduce the systolic blood pressure by at least 20 mm Hg to subsequent stroke), the influence on dementia reduction,
reach a value below 150 mm Hg or to matching placebo. although encouraging, is not definitive.
These individuals were free of dementia at baseline and
were subsequently monitored with the Mini-Mental State
4. Diabetes mellitus
Examination (MMSE), with further diagnostic tests to reach
a definitive diagnosis of dementia by using the Diagnostic There is evidence from epidemiologic studies that the
and Statistical Manual [of Mental Disorders], Third Edi- presence of diabetes is associated with subsequent cognitive
tion, Revised criteria if the score fell below 23. After a decline, specifically the development of ACD, AD, and
median follow-up of 2 years, the incidence of dementia was VaD [1]. Attempts to determine whether treatment of type 2
reduced by 50% from 7.7 to 3.8 cases per 1,000 patient diabetes mellitus affects the development of cognitive im-
years. The RR of dementia was 0.47 (95% confidence in- pairment or dementia have been addressed in a systematic
terval [CI], 0.28 to 0.78), and interestingly, most were review performed in the Cochrane Database [11]. Five
diagnosed with probable AD. In absolute terms, the number RCTs were identified in which different treatments for type
of older hypertensive patients needed to treat for 5 years to 2 diabetes were compared, and in which measures of cog-
prevent one case of dementia is approximately 53 [(NNT(5) ⫽ nitive function were made at entry and after treatment. In
53]. Five other RCTs of blood pressure lowering have exam- one trial the comparison of cognitive function was not
ined dementia as an outcome. Only one other, the blinded, in three studies cognitive function was not appro-
PROGRESS Study, showed a RR reduction for dementia of priately evaluated, and the fifth did not report on baseline
0.89 (95% CI, 0.74 to 1.07), which was not statistically cognitive function. The Cochrane analysis was that “there is
significant (P ⫽ .2) [8]. no convincing evidence relating type or intensity of diabetic
A meta-analysis of four trials (PROGRESS, SCOPE, treatment to the prevention or management of cognitive
SHEP, SySt-Eur) concluded that the RR reduction for demen- impairment in type 2 diabetes” [11].
tia was 0.80 (95% CI, 0.63 to 1.02), just missing statistical
significance [9]. It should be noted that the PROGRESS and
5. Stroke
HOPE trials enrolled patients with established cardiovascular
and cerebrovascular disease, so they cannot be considered as Stroke is associated with increased risk of ACD, AD, and
truly primary prevention studies. A further systematic evidence VaD. Although a comprehensive discussion of the literature
review protocol has been registered with the Cochrane Data- regarding stroke prevention is beyond the scope of this
base of Systematic Reviews by members of the Cochrane article, suffice it to say that there is an abundance of liter-
Dementia and Cognitive Improvement Group [10]. The date of ature addressing the topic of primary prevention of stroke.
completion of this review is uncertain. The consensus statement from the National Stroke Associ-
Although there are compelling reasons to treat systolic ation is a systematic review of fair quality [12]. These
hypertension in older individuals (eg, reducing the risk of guidelines offer recommendations for treatment of hyper-
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354 351

tension; acetylsalicylic acid (ASA) and statins after myo- stroke (OR, 0.86; 95% CI, 0.50 to 1.49). Only the combined
cardial infarction; warfarin or ASA for nonvalvular atrial outcome of stroke, myocardial infarction, or vascular death
fibrillation; consideration of carotid endarterectomy for was significantly reduced (OR, 0.71; 95% CI, 0.51 to 0.97)
asymptomatic stenosis greater than 60%; and attention to [16]. Although it is possible that antithrombotic treatment
lifestyle factors (diet, smoking cessation, regular exercise, for atrial fibrillation might result in a lower risk of dementia,
moderate alcohol use, and a healthy diet). Notwithstanding it remains unproven.
the efficacy of these interventions to reduce the risk of
subsequent stroke, a known risk factor for dementia, it 9. Lifestyle factors
remains to be seen whether such measures will influence the
incidence of dementia. 9.1. Diet
Both total fat and reduced levels of omega 3 fatty acids
6. Hyperlipidemia have been linked to increased risk of dementia in epidemi-
ologic studies [1]. Two systematic evidence reviews have
Several epidemiologic studies have established a rela-
addressed the issues of omega 3 fatty acids for the preven-
tionship between hyperlipidemia and subsequent develop-
tion of dementia [17,18]. Both concluded that there were no
ment of dementia. A systematic evidence review of the topic
RCTs on which to base a recommendation. Similarly, no
of statins for the prevention of AD was performed for the
RCTs were identified by the search strategy used here for other
Cochrane Database of Systematic Reviews [13]. The au-
dietary advice in relation to the prevention of dementia.
thors concluded that in the absence of RCTs there was “no
good evidence to recommend statins for reducing the risk of 9.2. Alcohol
AD”. Subsequent to this review, an RCT (PROSPER) was
published in 2002 [14]. In this study, 5,804 men and women Epidemiologic evidence has established that a moderate
with a history or risk factors for vascular disease were consumption of wine (250 to 500 mL/day) reduces the risk
randomized to receive either pravastatin 40 mg per day or of dementia [1]. However, the literature search did not
placebo. After follow-up of 3.2 years, various vascular end identify any RCTs addressing the topic of any type of
points were considered, but there was no significant effect alcohol for the prevention of dementia. Advice about alco-
on cognitive function or disability. However, this was not a hol consumption should be tempered by the known risks of
primary prevention trial. Not withstanding the epidemio- excessive use in terms of neurologic and other organ dam-
logic evidence, there is at present no clear rationale for the age as well as increased risk of injury from intoxication.
prescription of statin drugs to lower the risk of AD. 9.3. Activity, physical or mental
Although higher levels of physical activity have been
7. Hyperhomocysteinemia linked to lower subsequent rates of dementia, the search
Epidemiologic evidence links the presence of hyperho- failed to identify any RCTs of exercise with dementia as
mocysteinemia to an increased risk of ACD and AD. The an outcome. Similarly, no RCTs of mental activity were
literature search failed to identify any randomized con- identified.
trolled treatment trials aimed at lowering serum homocys- 9.4. Tobacco
teine levels with an outcome of dementia or AD.
Although case-controlled studies had suggested that to-
bacco smoking reduced the risk of dementia, longitudinal
8. Atrial fibrillation
cohort studies have identified a significant increased risk of
Atrial fibrillation is recognized as a risk factor for stroke ACD and AD for tobacco smokers [1]. Even in the absence
and has been identified in epidemiologic studies as a risk of RCTs, this epidemiologic evidence further adds to the
factor for dementia. Although numerous studies indicate many other reasons to discourage this habit.
that treatment with anticoagulants or ASA reduces the risk
9.5. Specific medications
of stroke, the literature search did not identify any studies
with dementia as an outcome. 9.5.1. Nonsteroidal anti-inflammatory drugs
In a meta-analysis of 16 RCTs, Hart et al [15] concluded Several recent systematic evidence reviews have ad-
that adjusted dose warfarin reduced the risk of stroke by dressed the topic of nonsteroidal anti-inflammatory drugs
62% (range, 48% to 72%), whereas ASA (6 trials) reduced (NSAIDs) on subsequent risk of AD and other dementias
the risk by 22% (range, 2% to 38%). Another meta-analysis [19,20]. Although two of these [19,20] concluded that those
of three trials of ASA for nonvalvular atrial fibrillation consuming NSAIDs had a lower risk of ACD and AD, the
concluded that the risk of all stroke was reduced by 30% third [21] did not. There have been no prospective RCTs of
(odds ratio [OR], 0.7; 95% CI, 0.47 to 1.07); ischemic NSAIDs in this context. Not withstanding the epidemio-
stroke (OR, 0.70; 95% CI, 0.46 to 1.07), and disabling fatal logic evidence, there is insufficient evidence at present to
352 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354

recommend for or against prescribing any NSAID for the boarding) including legislation on wearing helmets should
specific intention of reducing the risk of ACD or AD. be considered.

9.5.2. Vitamin supplementation 10.2. Education


A large prospective observational study has suggested The relationship between education and subsequent de-
that combined use of vitamin E (400 units daily) and vita- velopment of dementia might be explained by innate ability
min C (500 mg daily) for at least 3 years was associated (those possessing more innate intelligence might progress
with a reduction in the incidence of AD [23]. Other studies further in formal education and might be less likely to
have shown no such reduction, and high doses of vitamin E develop dementia), or that the acquisition of more formal
(ⱖ400 units daily) have been associated with increased education somehow promotes improved resilience to factors
mortality from all causes. A recent systematic evidence that predispose to dementia, or that some other factor might
review concluded that in the absence of prospective RCTs, have a common mechanism affecting the ability to pursue
there is no justification for consuming antioxidant vitamins formal education yet reducing the risk of dementia. Regard-
C and E for the intention of reducing the subsequent risk of less of the mechanism and in spite of the appeal of advocacy
AD [22]. for increasing standards of education, there is no evidence
that this strategy would lower the risk of dementia.
9.5.3. Estrogens
Systematic reviews of case-control and epidemiologic 10.3. Occupational exposure to toxins
evidence have suggested that consumption of estrogens
Longitudinal cohort studies have established a relation-
alone or in conjunction with progestins by perimenopausal
ship between exposure to environmental toxins and subse-
or postmenopausal women has been associated with a re-
quent development of dementia, specifically pesticides and
duced risk of AD and ACD. However, the situation changed
fumigants [1]. Although the absence of RCTs prevents a
dramatically with publication of the results from the Women’s
firm recommendation, it would seem prudent to minimize
Health Initiative Memory Study, a large RCT of estrogen and
exposure to such chemicals by the appropriate use of pro-
progestin replacement in more than 7,000 women aged 65 to
tective clothing and respiratory protection.
79 years [6]. Women assigned to estrogens alone had an
overall hazard ratio (HR) for probable dementia of 1.49
(95% CI, 0.83 to 2.66) compared with placebo, whereas 11. Discussion
those assigned to estrogen plus progestin showed a HR of
2.05 (95% CI, 1.21 to 3.48). Importantly, after participants A large amount of epidemiologic evidence is accumulat-
with baseline modified MMSE (3MS) scores at or below the ing for many risk factors for the subsequent development of
education-adjusted screening cut point were excluded, the ACD, AD, and VaD. However, when these risk factors are
HR was 1.77 (95% CI, 0.74 to 4.23; P ⫽ .20) in the estrogen critically evaluated in the context of studies aimed at reduc-
alone group and 2.19 (95% CI, 1.25 to 3.84) in the pooled ing the incidence of dementia, the quantity of Level 1
group [6]. The daily doses used were 0.625 mg of conju- (evidence from at least one well-designed RCT) is pitifully
gated equine estrogen and 2.5 mg of medroxyprogesterone small. Very few recommendations can be based on highest
acetate. In absolute terms, treatment of about 111 women quality evidence. Furthermore, it is unlikely that RCTs will
for 5 years would result in one additional case of dementia. ever be conducted on many of the risk factors identified in
Thus, there is no justification for prescribing estrogens or this review; thus recommendations must necessarily be
estrogen plus progestin to postmenopausal women with the based on less stringent types of evidence. The research
intention of reducing the risk of dementia. community is encouraged to focus on studies to determine
whether physical or mental activity can reduce the risk of
subsequent dementia; whether treatments aimed at hyper-
lipidemia, hyperhomocysteinemia, and glycemic control are
10. Miscellaneous factors
beneficial; and whether any type of NSAID, combination of
10.1. Head injury vitamins, alternatives to estrogens (eg, selective estrogen
receptor modifiers), or lipid-lowering agents might be ef-
Although there has been some controversy from the fective in primary prevention of dementia.
literature concerning the contribution of head injury to sub-
sequent development of dementia, it is now considered
well-established that serious head injury with loss of con- 12. Recommendations for the primary prevention of
sciousness increases the risk of subsequent AD [1]. dementia (ACD, AD, VaD)
Although there have been no RCTs on prevention of
dementia by reduction of head injuries, prudence would 1. There is good evidence to treat systolic hyperten-
dictate that measures to reduce serious head injuries (eg, sion (⬎160 mm Hg) in older individuals. In ad-
wearing of helmets during contact sports, bicycling, skate- dition to reducing the risk of stroke, the incidence
C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354 353

of dementia might be reduced. The target blood patients about the potential advantages of increased
pressure should be 140 mm Hg or less (Grade A, consumption of fish, reduced consumption of dietary
Level 1). fat, and moderate consumption of wine (Grade C,
2. Although ASA and statin medications after myocar- Level 2).
dial infarction, antithrombotic treatment for nonval-
vular atrial fibrillation, and correction of carotid ar- Author Disclosures
tery stenosis ⬎60% have been shown to reduce the
risk of stroke, there is insufficient evidence to rec- Angeles Garcia has received support from Janssen-Ortho
ommend for or against these measures for the spe- (Consultant, Advisor), Pfizer (Consultant, Advisor), Novar-
cific purpose of primary prevention of dementia tis (Consultant, Advisor), and Lundbeck (Consultant, Advi-
(Grade C, Level 1). sor).
3. Although there are many reasons for treating type 2 Chris MacKnight has received support from Alzheimer
diabetes, hyperlipidemia, and hyperhomocysteine- Society of Nova Scotia (Board Member), Janssen Ortho Inc.
mia, there is insufficient evidence to recommend (Speaker, Trial Investigator), Pfizer Canada (Speaker, Trial
treatment of these conditions for the specific pur- Investigator), Novartis (Speaker, Trial Investigator), Lund-
pose of reducing the risk of dementia (Grade C, beck Canada (Speaker, Trial Investigator), Voyager Phar-
Level 2). maceuticals (Trial Investigator), Myriad (Trial Investiga-
4. There is insufficient evidence to recommend for or tor), and Neurochem (Trial Investigator).
against the prescription of NSAIDs for the sole purpose
of reducing the risk of dementia (Grade C, Level 2). Acknowledgments
5. There is good evidence to avoid the use of estrogens
alone or together with progestins for the sole pur- Chris MacKnight is supported from a CIHR New Inves-
pose of reducing the risk of dementia (Grade E, tigator Award.
Level 1). During preparation of this article, Christopher Patterson
6. Although there is insufficient evidence to make a received financial support from the Institute of Advanced
firm recommendation, physicians might advocate for Studies, University of Bologna, Bologna, Italy.
strategies, including legislation, to reduce the risk of
serious head injuries (Grade C, Level 2). References
7. Although there is insufficient evidence to make a
firm recommendation, physicians might advise their [1] Patterson C, Feightner JW, Garcia A, MacKnight CR. General risk
patients about, and advocate for, appropriate pro- factors for dementia. a systematic evidence review. Alzheimers De-
tective clothing during administration of pesti- ment 2007;3.
[2] Black SE, Patterson C, Feightner J. Preventing dementia. Can J Neu-
cides, fumigants, fertilizers, and defoliants (Grade rol Sci 2001;28(Suppl 1):S56 – 66.
C, Level 2). [3] Hsuing G-YR, Sadovnick AD. Genetics and dementia. risk factors,
8. There is insufficient evidence to recommend for or diagnosis and management. Alzheimers Dement 2007;3.
against supplementation with vitamins E or C for the [4] Woolf SH, Battista RN, Anderson GM, et al. Assessing the clinical
prevention of dementia (Grade C, Level 2). High effectiveness of preventive manoeuvres: analytic principles and sys-
tematic methods in reviewing evidence and developing clinical prac-
dose vitamin E (ⱖ400 units/day) is associated with tice recommendations. J Clin Epidemiol 1990;43:891–905.
excess mortality and should not be recommended [5] Hunt DL, McKibbon KA. Locating and appraising systematic re-
(Grade E, Level 1). views. In: Mulrow C, Cook D, eds. Systematic reviews: synthesis of
9. Although recommendations might be made on other best evidence for health care decisions. Philadelphia: American Col-
grounds (such as part of a healthy lifestyle), there is lege of Physicians; 1998. p. 13–22.
[6] Shumaker SA, Legault C, Kuller L, Rapp SR, Thal L, Lane DS, et al.
insufficient evidence to recommend for or against Conjugated equine estrogens and incidence of probable dementia and
higher levels of physical or mental activity for the mild cognitive impairment in postmenopausal women: Women’s
specific purpose of reducing the incidence of demen- Health Initiative Memory Study. JAMA 2004;291:2947–58.
tia (Grade C, Level 2). [7] Forette F, Seux M-L, Staessen JA, Thijs L, Birkenhäger WH, Babar-
10. Although there is insufficient evidence to make a skiene MR, et al. Prevention of dementia in randomized double-blind
placebo-controlled Systolic Hypertension in Europe (Syst-Eur) trial.
firm recommendation for the primary prevention of Lancet 1998;352:1347–51.
dementia, physicians might advocate for appropriate [8] The PROGRESS Collaborative Group. Effects of blood pressure
levels of education and strategies to retain students lowering with perindopril and indapamide therapy on dementia and
in appropriate learning environments (Grade C, cognitive decline in patients with cerebrovascular disease. Arch In-
Level 2). tern Med 2003;163:1069 –75.
[9] Feigin V, Ratnasabapathy Y, Anderson C. Does blood pressure low-
11. Although there is insufficient evidence to make a ering treatment prevent dementia or cognitive decline in patients with
firm recommendation for the primary prevention of cardiovascular and cerebrovascular disease? J Neurol Sci 2005;229 –
dementia, physicians might choose to advise their 230:151–5.
354 C. Patterson et al. / Alzheimer’s & Dementia 3 (2007) 348 –354

[10] McGuinness B, Todd S, Passmore P, Bullock R. The effects of blood [22] Boothby LA, Doering PL. Vitamin C and vitamin E for Alzheimer’s
pressure lowering on development of cognitive impairment and de- disease. Ann Pharmacother 2005;39:2073– 80.
mentia in patients without apparent prior cerebrovascular disease. [23] Zandi PP, Anthony JC, Khachaturian AS, Stone SV, Gustafson D,
Cochrane Database Syst Rev 2006;(1). Tschanz JT, et al. Reduced risk of Alzheimer disease in users of
[11] Areosa Sastre A, Grimley Evans J. Effect of the treatment of Type II antioxidant vitamin supplements: the Cache County Study. Arch
diabetes mellitus on the development of cognitive impairment and Neurol 2004;61:82– 8.
dementia. Cochrane Database Syst Rev 2006;1.
[12] Gorelick PB, Sacco RL, Smith DB, et al. Prevention of a first stroke:
a review of guidelines and a multidisciplinary consensus statement Appendix 1
from the National Stroke Association. JAMA 1999;281:1112–20.
[13] Scott HD, Laake K. Statins for the prevention of Alzheimer’s disease. Search strategies for systematic reviews of RCTs on
Cochrane Database Syst Rev 2006;1: accession number: 00075320- prevention of dementia or Alzheimer’s disease
100000000-02141. Medline: used clinical queries function of Medline to
[14] Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM, Cobbe search for systematic reviews (dementia AND prevent*)
SM, et al. Pravastatin in elderly individuals at risk of vascular disease
(PROSPER): a randomized controlled trial. Lancet 2002;360:1623–30.
AND systematic [sb] 126 articles; (Alzheimer* AND pre-
[15] Hart RG, Benavente O, McBride R, Pearce LA. Antithrombotic vent* AND systematic (sb) 62 articles; (Alzheimer* OR
therapy to prevent stroke in patients with atrial fibrillation: a meta- dementia) AND prevent* Field: All Fields, Limits: Publi-
analysis. Ann Intern Med 1999;131:492–501. cation Date from 1996 to 2006, Randomized Controlled
[16] Aguilar MI, Hart R. Oral anticoagulants for preventing stroke in Trial, Human 122 articles
patients with non-valvular atrial fibrillation and no previous history of
stroke or transient ischemic attacks. Cochrane Database Syst Rev
Also searched: google scholar: NHS database;
2006;1: accession number: 00075320-100000000-01342. Total, 310 articles
[17] Lim W, Gammack J, Van Niekerk J, Dangour A. Omega 3 fatty acid Articles were considered ineligible if (1) secondary pre-
for the prevention of dementia. Cochrane Database Syst Rev 2006: vention for those with dementia or AD; (2) human immu-
CD005379. nodeficiency virus–related dementia; (3) articles with neu-
[18] MacLean CH, Issa AM, Newberry SJ, Mojica WA, Morton SC,
rochemical focus; (4) articles not containing original or new
Garland RH, et al. Effects of omega-3 fatty acids on cognitive
function with aging, dementia, and neurological diseases. Evidence data.
Report/Technology Assessment; 2005; number 114:13– 6. For targeted search of individual preventive interven-
[19] Etminan M, Gill S, Samii A. Effect of non-steroidal anti-inflammatory tions, the following search strategy was used:
drugs on risk of Alzheimer’s disease: systematic review and meta-
analysis of observational studies. BMJ 2003;327:128. Cochrane Database of Systematic Reviews (up to
[20] Szekely CA, Thorne JE, Zandi PP, Ek M, Messias E, Breitner JC, 2006);
et al. Nonsteroidal anti-inflammatory drugs for the prevention of ACP Journal Club (1991 to February 2006);
Alzheimer’s disease: a systematic review. Neuroepidemiology
Database of Abstracts of Review of Effects (DARE)
2004;23:159 – 69.
[21] de Craen AJM, Gussekloo J, Vrijsen B, Westendorp RGJ. Meta- (up to 2006);
analysis of nonsteroidal antiinflmmatory drug use and risk of demen- Cochrane Central Register of Controlled Trials
tia. Am J Epidemiol 2005;161:114 –20. (CCRCT) (up to 2006)
Alzheimer’s & Dementia 3 (2007) 355–384

Management of mild to moderate Alzheimer’s disease and dementia


David B. Hogana,*, Peter Baileyb, Anne Carswellc, Barry Clarked, Carole Cohene,
Dorothy Forbesf, Malcolm Man-Son-Hingg, Krista Lanctôth, Debra Morgani, Lilian Thorpej
a
Departments of Medicine and Clinical Neuroscience, University of Calgary; Calgary, Alberta, Canada
b
Department of Medicine (Neurology), Dalhousie University, Halifax, Nova Scotia, Canada
c
School of Occupational Therapy, Dalhousie University, Halifax, Nova Scotia, Canada
d
Department of Family Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
e
Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
f
School of Nursing, Faculty of Health Sciences, University of Western Ontario, London, Ontario, Canada
g
Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
h
Departments of Psychiatry and Pharmacology, University of Toronto, Toronto, Ontario, Canada
i
Institute of Agricultural Rural and Environmental Health, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
j
Department of Psychiatry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

Abstract The authors were charged with making a series of evidence-based recommendations that would
provide concrete advice on all aspects of the management of mild to moderate stages of dementia
and Alzheimer’s disease. The recommendations were primarily targeted to primary care physicians
practicing in Canada. The assigned topic area did not include either the assessment of a patient with
suspected dementia or the prevention of Alzheimer’s disease and other dementias. An extensive
examination of the available literature was conducted. Explicit criteria for grading the strength of
recommendations and the level of evidence supporting them were used. The 28 evidence-based
recommendations agreed on are presented in this article.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Alzheimer’s disease; Vascular dementia; Mild-to Moderate; Disease management

1. Introduction State Examination (MMSE) score between 10 –11 and


24 –26 (inclusive). This would translate to a Global De-
This article deals with the management of Alzheimer’s
terioration Scale stage of 4-6 and/or a Clinical Dementia
disease (AD) and other forms of dementia. The working
Rating score of 1–2.
group was asked to produce a summary of the available
In this document we will not deal with the prevention of
evidence on this topic. On the basis of this review we were
dementia and AD. Also, other than for AD we will make
to develop a series of recommendations that would provide
few recommendations for the management of specific types
concrete advice on all aspects of therapy along with sug-
of dementia (eg, dementia with Lewy bodies [DLB]). We
gestions for future developments. The primary target audi-
will not address the issue of the cost-effectiveness of the
ence for these recommendations would be primary care
various interventions described for patients with mild to
physicians.
moderate AD and dementia. A fiercely debated issue at this
The definition of mild to moderate AD and dementia
time is whether any of the available medications for mild to
was left up to the authors of the articles reviewed. Typ-
moderate AD are cost-effective. Our assigned topic area
ically they defined it as a patient meeting criteria for a
overlapped with the severe dementia working group be-
diagnosis of AD and/or dementia who had a Mini-Mental
cause a number of the studies we examined dealt with
moderate to severe stages of the condition. We would advise
*Corresponding author. Tel.: 403-220-4578; Fax: 403-283-6151. the reader to review the background article of this working
E-mail address: dhogan@ucalgary.ca group as well.
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.006
356 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

2. Methods research literature (eg, meta-analyses, systematic reviews,


consensus statements, clinical practice guidelines), there
Our working group was charged with the responsibility would be some areas in which they would have to perform
of addressing the management of mild to moderate AD and a primary review of the pertinent literature. They were also
dementia. The chair (D.H.) and the initial members of the asked to focus their energies on what they thought were the
working group (P.B., C.C., M.H., L.T.) were selected by the key areas within their subsections and to draft recommen-
Steering Committee of the Third Canadian Consensus Con- dations that would be both important and feasible for a
ference on Diagnosis and Treatment of Dementia (3rd primary care physician.
CCCDTD). In a series of teleconferences the assigned task The draft recommendations developed by working group
was divided into a number of subsections, with delegation members and a first draft of the background article were
of lead responsibility to members of the working group. To distributed by the Chair to all working group members for
ensure that we had the range of required talent and exper- review, discussion, and modification. After this process the
tise, additional members of the working group were re- background article with recommendations was submitted to
cruited (A.C., B.C., D.F., and K.L.). The involved subsection the Steering Committee for posting on the Web page of
leads identified, discussed, and resolved in mutually acceptable the 3rd CCCDTD. Feedback received was discussed by the
manner areas of potential overlap. During our teleconferences working group, and final modifications were made to the
we clarified how we would approach the task and dealt col- recommendations before their presentation at the consensus
lectively with any concerns expressed by working group mem- meeting on March 10, 2006. All of the recommendations
bers. Minutes were kept of these teleconferences and circulated presented in this article achieved consensus (80% plus ap-
to all members of the working group and the Chair of the 3rd proval by participants of the 3rd CCCDTD).
CCCDTD Steering Committee. The quality of the literature (levels of evidence) was
The initial literature searches were conducted by an in- graded by using the following system adapted from Cana-
formation specialist hired by the 3rd CCCDTD. PubMed dian Task Force on Preventive Health Care [1]:
and Embase databases were used. The strategy and major
keywords used in the searches were “dementia” OR “Alz- I. Evidence from at least one properly randomized
heimer’s disease” AND “mild” OR “moderate” AND “ther- controlled trial (RCT).
apy” OR “treatment.” Secondary search terms included II-1. Evidence from well-designed controlled trials with-
(listed alphabetically) “affective disorder,” “agitation,” “an- out randomization.
tidepressants,” “anti-inflammatory drugs,” “antioxidants,” II-2. Evidence from well-designed cohort or case-
“anxiety,” “anxiolytics” OR “tranquilizers,” “behaviour,” control analytic studies, preferably from more than
“cholinesterase inhibitors,” “care-giver,” “counseling,” “de- one center or research group.
pression,” “disinhibition,” “discontinue,” “education,” “en- II-3. Evidence from comparisons between times or
vironment,” “ginkgo,” “hormones,” “hypnotics” OR “sleep places with or without the intervention. Dramatic
medications,” “maintain,” “memantine,” “metabolic en- results in uncontrolled experiments are included in
hancers,” “neurotrophic agents,” “nootropics,” “rehabilita- this category.
tion,” “selective serotonin reuptake inhibitors” OR “sleep.” III. Opinions of respected authorities, on the basis of
The search was limited to articles written in English, dealing clinical experience, descriptive studies, or reports
with human research, and published since January 1, 1996. of expert committees.
A total of 1,615 articles were identified. Six hundred The strength of the recommendations (Grade of Recom-
sixty-one of the articles were eliminated after examination mendation) was graded by using the following system
of the title. The titles, authors, and abstracts of the remain- adapted from the Canadian Task Force on Preventive Health
ing 954 were distributed to all working group members. Care [1,2]:
Each was responsible for selecting articles for detailed re-
view by their subsection, abstracting data from the selected A: There is good evidence to support this maneuver.
articles, synthesizing the available information, and devel- B: There is fair evidence to support this maneuver.
C: The existing published evidence is either conflicting
oping draft recommendations. Full texts of articles selected
or insufficient and does not allow one to recommend
by working group members were provided by the informa-
for or against this maneuver; however, a recommen-
tion specialist. On the request of working group members,
dation might be made on other grounds.
additional searches were conducted by the information spe-
D: There is fair evidence to recommend against this
cialist. The search strategies for them were developed in
maneuver.
consultation with the individuals making the request. Work-
E: There is good evidence to recommend against this
ing group members were also encouraged to use their own
maneuver.
files and to search the reference lists of selected articles for
additional relevant articles. Working group members were Our recommendations were based on the best available
told that although they could use articles that summarize the evidence. We preferentially used rigorously done system-
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 357

atic reviews of the current literature. When a primary ex- 3.4. Recommendation 3
amination of the literature was done, we sought to base our
conclusions on statistically and clinically significant find- The care and management of patients with dementia
ings from high quality RCTs. from specific cultural groups should take into account the
The conclusions of the 1998 Canadian Consensus risk of isolation, the importance of culturally appropriate
Conference on Dementia (CCCD) that were considered rel- services, and issues that arise in providing caregiver support
evant to our assigned area and were still supported by the (Grade B, Level III).
members of the working group are also included in this The presence of a preexisting dementia is the risk factor
document [1]. most strongly associated with the development of delirium
in older hospitalized patients [3]. A multicomponent inter-
vention to prevent delirium (ie, orienting communication,
therapeutic activities, sleep enhancement strategies, exer-
3. Results: Management of mild to moderate
cise and mobilization, provision of vision and hearing aids,
Alzheimer’s disease and dementia
oral repletion of dehydration) has been shown to decrease
3.1. General measures the likelihood of delirium developing in older hospitalized
patients at increased risk [4]. Once it occurs, the manage-
A number of the conclusions reached at the CCCD [1] ment of delirium in a patient with dementia remains empir-
were believed to be both relevant to our assigned topic area ical, with no evidence from recent studies to support
and received the support of the members of the working changes from current practices [5].
group after minor modifications. Three are presented below, Comorbidities are both common and costly in patients
and others will be appear later in the document. with AD and dementia [6,7]. Appropriate therapy of their
comorbidities is an important component of the care pro-
3.2. Recommendation 1
vided to these patients. There is evidence that patients with
Most patients with dementia can be assessed and man- dementia are less likely to be offered recommended therapy
aged adequately by their primary care physicians. However, for other conditions [8]. Also, poor control of comorbidities
to assist them in meeting the needs of patients and their might accelerate the rate of progression in AD. With dia-
caregivers, it is recommended that (1) all patients with betes as an example, hyperglycemia itself can induce cog-
dementia and their families who consent be referred to the nitive changes by disrupting glucose metabolism with the
local chapter of the Alzheimer Society (eg, First Link pro- formation of abnormal glycosylation products and the de-
gram where available); and (2) primary care physicians velopment of microvascular changes [9]. Hyperinsulinemia
should be aware of the resources available for the care of is a risk factor for accelerated cognitive decline [10]. Insulin
those with dementia in their community (eg, support groups, increases the release of beta-amyloid and interferes with its
adult day programs) and to make appropriate referrals to degradation by competing for the insulin-degrading enzyme
them (Grade B, Level III). that metabolizes both insulin and beta-amyloid in the central
nervous system [11]. Optimal diabetic management might
3.3. Recommendation 2 slow down the rate of further decline in patients with ex-
The referral/consultation process is essential to the de- isting AD, but this requires confirmation [12]. Community-
livery of high quality health care. In the care of a patient based studies have shown that the presence of comorbidities
with mild to moderate dementia, reasons to consider referral predicts a higher mortality rate in patients with AD [13–15].
to a geriatrician, geriatric psychiatrist, neurologist, or other The presence of AD and dementia will affect the manage-
health care professional (eg, neuropsychologist, nurse, ment of other chronic conditions. A unique feature of the
nurse practitioner, occupational therapist, physical therapist, care of a demented patient is reduced reliance on patient
psychologist, social worker) with the appropriate knowl- self-care and a concomitant increase in the effort to provide
edge and expertise in dementia care would include (1) caregiver support and education.
continuing uncertainty about the diagnosis after initial as-
3.5. Recommendation 4: Recommendations with regards
sessment and follow-up; (2) request by the patient or the
to the general medical care of a patient with mild to
family for another opinion; (3) presence of significant de-
moderate dementia
pression, especially if there is no response to treatment; (4)
treatment problems or failure with specific medications for
AD; (5) need for additional help in patient management (eg, A. Patients with mild to moderate dementia when hos-
behavioral problems, functional impairments) or caregiver pitalized should be identified as being at increased
support; (6) genetic counseling when indicated; and (7) if risk for delirium. They should be offered multicom-
the patient and/or family express interest in either diagnostic ponent interventions including orienting communica-
or therapeutic research studies that are being carried out by tion, therapeutic activities, sleep enhancement strat-
the recipient of the consult request (Grade B, Level III). egies, exercise and mobilization, provision of vision
358 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

and hearing aids, and/or oral repletion of dehydration with medication management, including poor adher-
to decrease their risk of developing delirium (Grade ence, should be done on all patients with mild to
B, Level II-1). moderate dementia. If problems are detected, in par-
B. Comorbidities of patients with mild to moderate AD ticular with adherence, the use of compliance aids or
should be appropriately managed (Grade B, Level the assumption of medication management by an-
III). other party will be necessary. The effectiveness of
C. The management of other chronic medical conditions any alterations in medication management will have
might have to be modified in the setting of a demen- to be assessed (Grade B, Level III).
tia. In general there should be less reliance on patient B. Even when the patient is safely self-managing their
self-care and a concomitant increase in the role medications, there should be planning for the in-
played by caregivers (Grade B, Level III). volvement of a third party in the management of
medications for all patients with a progressive de-
Ensuring medication adherence can be a significant chal-
mentia because this will eventually become necessary
lenge in the care of an individual with dementia because
in nearly all (Grade B, Level III).
cognitive factors limit the ability of patients to self-medicate
C. The use of medications with anticholinergic effects
[16]. Caregivers might have to become involved in medi-
should be minimized in persons with AD (Grade D,
cation management. The use of compliance aids would be
Level III).
another option. For example, computer telephony systems
might improve medication adherence [17]. The presence of dementia does not in itself mean that
Studies have shown that older adults with probable de- patients lack capacity to make decisions about themselves
mentia are more likely to be taking anticholinergics than and their estate. AD and other neurodegenerative dementias
matched controls [18]. A partial listing of medications with are progressive conditions, however, that at some point will
anticholinergic activity is given in Table 1. Use of medica- likely rob patients of their mental capacity to consent to
tions with anticholinergic effects can worsen the cognitive treatment, consent to participate in a research study, look
status of individuals with AD and dementia [19 –23]. Older after their estate, and/or make decisions about other aspects
persons taking anticholinergic medications can manifest of their life. Questions about capacity are likely to occur
significant deficits in cognitive functioning and be classified during the mild and moderate stages of AD.
as having mild cognitive impairment [24]. Although some
drugs (eg, amitriptyline, benztropine) are well-known for 3.7. Recommendation 6: Ethicolegal recommendations
their anticholinergic effects, numerous other medications
possess mild anticholinergic properties. By themselves A. Although each case should be considered individu-
these latter agents are unlikely to lead to significant clinical ally, in general the diagnosis of dementia should be
symptoms, but the additive effects of multiple anticholin- disclosed to the patient and family. This process
ergic medications taken simultaneously might result in ad- should include a discussion of prognosis, diagnostic
verse effects. Another concern with their use is that they uncertainty, advance planning, driving issues, treat-
might blunt the effects of cholinesterase inhibitors because ment options, support groups, and future plans
anticholinergics directly oppose the therapeutic effect of (Grade B, Level III).
these medications. The concurrent use of anticholinergics B. Primary care physicians should be aware of the per-
and cholinesterase inhibitors is reportedly common [25]. A tinent laws in their jurisdiction about informed con-
small cohort study found that concomitant therapy with sent, the assessment of capacity, the identification of
anticholinergics was associated with worse outcomes in a a surrogate decision maker, and the responsibilities of
group of demented individuals being treated with donepezil physicians in these matters (Grade B, Level III).
[26]. Another small study looked at cognitive and behav- C. While patients with AD retain capacity, they should
ioral status both on and off incontinence medications that be encouraged to update their will and to enact both
had anticholinergic effects. The subjects exhibited better an advance directive and an enduring power of attor-
performance on specific measures of cognition and behavior ney (Grade B, Level III).
when off these medications. A significant, inverse relation-
ship was found between mental status and anticholinergic 4. Nonpharmacologic interventions for cognitive and
level [27]. functional limitations
3.6. Recommendation 5: Recommendations about the
4.1. Cognitive training/cognitive rehabilitation
use of medications in the setting of a mild to
moderate dementia Cognitive training is defined as guided practice on a set of
standard tasks designed to reflect specific cognitive function
A. Determination of how medications are being con- such as memory or attention. Cognitive rehabilitation is an
sumed and identification of any problems/concerns individualized approach to helping people with cognitive im-
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 359

Table 1 Table 1
Select medications with anticholinergic activity [18,280,281] Continued
Antiarrhythmic Glycopyrrolate
Disopyramide Hyoscine butylbromide
Antidiarrheal Hyoscyamine
Diphenoxylate/atropine Methscopolamine bromide
Antiemetics/antivertigo Oxybutynin
Cyclizine Pinaverium bromide
Dimenhydrinate Propantheline
Meclizine Tolterodine
Scopolamine Muscle relaxants
Trimethobenzamide Cyclobenzaprine
Antihistamines, either single or combination products containing Orphenadrine
Azatadine Opioid
Brompheniramine Meperidine
Carbinoxamine Tricyclic antidepressants
Chlorpheniramine Amitriptyline
Clemastine Amoxapine
Cyproheptadine Clomipramine
Dexbrompheniramine Doxepin
Dexchlorpheniramine Imipramine
Dimenhydrinate Nortriptyline
Diphenhydramine Protriptyline
Doxylamine Trimipramine
Hydroxyzine
Phenindamine
Promethazine
Trimeprazine pairment during which the person and family member deter-
Triprolidine mine personally relevant goals and then devise strategies for
Antiparkinsonian
addressing them to improve function in everyday contexts.
Benztropine
Biperiden Three small-sample RCTs examined the effect of cognitive
Ethopropazine training on memory tasks and functional performance in per-
Orphenadrine sons with dementia also taking cholinesterase inhibitors [28 –
Procyclidine 30]. The results suggest that performance on instrumental ac-
Trihexyphenidyl
tivities of daily living (IADL) were modestly enhanced.
Antipsychotics
Chloropromazine Overall there were no significant differences in functional
Clozapine performance between the groups at the end of the study period
Flupenthixol or on follow-up. The modest improvements seen on specific
Fluphenazine memory tasks were not sustained or generalizable to other
Loxapine
tasks.
Mesoridazine
Methotrimeprazine The only systematic review of cognitive training con-
Olanzapine cluded that because of a limited number of small-sample
Pericyazine RCTs and their equivocal results, it was not possible to draw
Pimozide any firm conclusions about the effectiveness of these inter-
Prochlorperazine
ventions on cognitive skills [31]. The data suggest that there
Promazine
Promethazine was a training effect on the specific skills being trained
Thioflupromazine (splinter skills) but little or no generalization to other cog-
Thioproperazine nitive abilities or to functional performance.
Thioridazine There was some evidence that an errorless learning par-
Thiothixene
adigm and/or consistent practice of usual daily activities
Zuclopenthioxol
Bronchodilators (procedural memory training) worked in a cognitive reha-
Atropine bilitation program to improve functional performance [32–
Ipratropium 35]. There were gains in functional performance that were
Gastrointestinal/genitourinary antispasmodics, either single or maintained during a period of 2 years. However, none of the
combination products containing
studies were RCTs.
Belladonna alkaloids
Clidinium bromide 4.2. Environment
Dicyclomine
Dicycloverine There is one high quality RCT, some pilot projects, and
Flavoxate
two systematic reviews of the literature examining the im-
360 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

pact of environmental interventions on functional perfor- 4.5. Other therapies


mance. The RCT examining the effectiveness of a home-
One small RCT (14 subjects) reported that transcutane-
environmental intervention was conducted to examine,
ous electrical nerve simulation (TENS) appeared to have a
among other things, its effect on the daily functioning of
statistically beneficial effect on ADL that was present 6
persons with dementia living at home. The program con-
weeks after intervention [44]. A systematic review of TENS
sisted of individualized environmental modifications (con- suggests that it produced short-term improvements in spe-
sistency, structured) and aides (bath seats, visual cues). The cific neuropsychological tests, but that the effectiveness of
results indicated a modest but statistically significant effect this intervention was stage-dependent, did not appear to
on IADL 3 and 6months after intervention [36], with trends impact functional performance, and was not maintained
still present at 12 months [37]. beyond 3 weeks [45]. A systematic review of psychosocial
Two systematic reviews of environmental impact on interventions for persons with mild or moderate dementia
functional performance were undertaken. Day et al [38] [46] reported that reality orientation was not effective in
reported that discrete individualized design features such as improving functional performance.
organization of space, simplicity, and structure that pro-
moted orientation, problem-solving, memory, and mobility 4.6. Recommendation 7: Recommendations for
had positive impacts on functional performance. A review nonpharmacologic interventions for the management of
of six RCTs that met inclusion criteria [39] suggested that the cognitive and functional limitations arising from mild
environmental modification in the form of assistive devices, to moderate AD
aides, and adaptations together with individualized occupa-
The available research on nonpharmacologic interven-
tional therapy impacted positively on functional perfor-
tions for functional performance in persons with mild or
mance of persons with cognitive impairment still living at moderate dementia is limited. There are few well-designed
home. studies on which to base any firm conclusions. There is
promise for individualized exercise programs, individual-
4.3. Exercise ized environmental modifications, and individualized prac-
An RCT of 153 persons [40] examined whether a tice of ADL and IADL tasks. Future research, however, is
3-month home-based exercise program together with required to provide the evidence needed to reach solid
conclusions about the effectiveness of these interventions.
strategies for caregivers to manage behavior improved
functional independence in those with mild to moderate
A. There is insufficient research evidence to come to any
AD. The results indicated that the exercise group had firm conclusions about the effectiveness of cognitive
significant increases on measures of physical functioning training/cognitive rehabilitation in improving and/or
and ADL. This trend persisted at 1 year after interven- maintaining cognitive and/or functional performance
tion. A systematic review of the literature supports the in persons with mild or moderate dementia (Grade C,
notion that repetitive, consistent training of tasks by Level 1).
using procedural memory improves motor tasks and func- B. Further research is required to be able to conclude
tional performance [41]. A meta-analysis of 2,020 sub- that cognitive training/cognitive rehabilitation is ef-
jects in 30 trials to determine whether exercises are fective in improving cognitive and/or functional per-
beneficial for people with mild to moderate dementia formance in persons with mild or moderate dementia
suggested that exercise programs increased strength, fit- (Grade B, Level II-1).
ness, functional performance (ie, ADL, IADL), cognitive C. Although there is some indication of a beneficial
function, and positive behavior [42]. impact on IADL and ADL, there is insufficient evi-
dence to make firm conclusions about the effective-
4.4. Occupational therapy ness of environmental interventions in promoting
functional performance in persons with mild or mod-
One experimental study compared a structured occupa- erate dementia (Grade C, Level 1).
tional therapy program of caregiver strategies, environmen- D. There is good evidence to indicate that individualized
tal modifications, and community-based assistance to care- exercise programs have an impact on functional per-
givers receiving a written report [43]. They found that for formance in persons with mild or moderate dementia
persons with moderate dementia, self-care improvements (Grade A, Level 1).
noted after intervention were maintained at 3 months. How- E. For other nonpharmacologic therapeutic interventions
ever, a systematic review [39] concluded that there is in- there is insufficient evidence to allow any conclusions
sufficient evidence about the effectiveness of individualized being made about their efficacy in improving or
occupational therapy programs, and further research was maintaining functional performance in persons with
required. mild or moderate dementia (Grade C, Level 1).
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 361

5. Pharmacotherapy for cognitive and comprehension of spoken language, recall of test instructions,
functional impairments word finding, following commands, naming objects, construc-
tion, ideational praxis, orientation, word recall, and word rec-
5.1. Retained general recommendations for the ognition) and is scored out of 70 (higher scores indicate greater
pharmacotherapy of dementia initially proposed by the impairment). MMSE scores are also often reported as a sec-
CCCD [1] (with minor amendments) ondary cognitive outcome. Activities of daily living have been
5.1.1. Recommendation 8 evaluated with a variety of instruments such as the Progressive
Primary care physicians should be able to administer and Deterioration Scale (PDS), the Disability Assessment for De-
interpret brief measures of functional activities and cogni- mentia (DAD), and the Bristol Activities of Daily Living Scale
tive abilities or refer to health care professionals with the (BADLS). Global assessment has usually been done with the
required knowledge and expertise (Grade B, Level III). Clinician’s Interview-Based Impression of Change with care-
giver input (CIBIC-Plus). Patients are assessed at baseline. At
5.1.2. Recommendation 9 subsequent assessments they are graded on a 1 to 7 scale
After treatment has been started, patients should be re- relative to this baseline assessment, with one indicating very
assessed regularly by the appropriate health care profes- much improved, four no change, and seven very much worse.
sional involved in their care (Grade B, Level III). The RCTs of these agents have shown consistent, albeit mod-
5.1.3. Recommendation 10 est, benefits of treatment in cognition, activities of daily living,
Records should be kept such that stabilization, improve- and global clinical state. Systematic reviews including those
ment, or persisting deterioration in treated patients will be with severe dementia find similar results to those including
determinable (Grade B, Level III). only trials of those with mild to moderate impairment. The
methodologic limitations of the published studies (eg, report-
5.1.4. Recommendation 11 ing more than one outcome without statistical correction for
In monitoring the response to therapy of patients with multiple comparisons, absence of final outcome measures on
dementia, the input of caregivers (where available) should subjects who had withdrawn) were specifically noted in a
be sought. They can provide information on the patient’s systematic review of the cholinesterase inhibitors. Because of
cognition, behavior, and social and daily functioning (Grade the modest benefits seen and the methodologic limitations of
B, Level III). the studies, the authors questioned the utility of these agents
[57]. Other commentators have concluded, however, that
5.1.5. Recommendation 12
the likely effect of the methodologic concerns does not
If the attending primary care physician is unable to
invalidate the findings of the studies reviewed and that
perform the assessments required to gauge response to ther-
these agents are modestly efficacious for the treatment of
apy, referral to another health care professional with knowl-
mild to moderate AD.
edge and expertise in dementia care (eg, other physician,
Comparison of the relative efficacy and tolerability of the
nurse, occupational therapist) or a service (eg, memory
cholinesterase inhibitors across clinical trials is not appropriate
clinic) who is willing to perform such assessments is ad-
because of differences in the baseline characteristics of the
vised (Grade B, Level III).
subjects, the efficacy assessments done, and how subjects were
5.1.6. Recommendation 13 assessed for adverse effects. The available trials that directly
Primary care physicians should be able to communicate compared one cholinesterase inhibitor to another have meth-
appropriate information concerning dementia, including re- odologic limitations and/or show no significant differences in
alistic treatment expectations, to their patients and their their primary outcome measures [47,58,59].
families (Grade B, Level III). The most common side effects encountered with the
Most clinicians view the cholinesterase inhibitors as cholinesterase inhibitors in the RCTs of these agents were
first-line treatment for mild to moderate stages of AD [47]. gastrointestinal (eg, anorexia, nausea, vomiting, diarrhea)
There are three cholinesterase inhibitors available in Can- [47–50,60]. They are more likely to occur at the commence-
ada: donepezil, galantamine, and rivastigmine. Although ment of therapy or when the dose of the agent is increased.
rivastigmine and galantamine have additional modes of ac- These side effects are dose-related and tend to be transient.
tion, they all inhibit the breakdown of acetylcholine by Gastrointestinal side effects in the RCTs were more com-
blocking the enzyme acetylcholinesterase. mon with rivastigmine. Slower titration and ensuring riv-
Cochrane reviews of each of the agents are available [48 – astigmine is taken with food appear to decrease the risk of
50] as well as other meta-analyses [51–53] and qualitative gastrointestinal side effects. Weight loss did occur during
systematic reviews [54 –57]. Cognitive, functional, and global the RCTs of all three agents, but a follow-up of patients with
outcomes have been measured in these trials. The primary AD treated with cholinesterase inhibitors found that there
cognitive test in most studies has been the cognitive section of was not an increased risk for long-term weight loss (com-
the Alzheimer’s Disease Assessment Scale (ADAS-Cog). This pared with patients with AD not receiving a cholinesterase
instrument consists of 11 individual items (spoken language, inhibitor) and might in fact be a protective factor [61].
362 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

A variety of other adverse effects can occur. Dizziness B. Although all three cholinesterase inhibitors available
has been reported with all three agents. If disabling, dose in Canada have efficacy for mild to moderate AD,
reduction would be a reasonable initial approach. Syncope, equivalency has not been established in direct com-
while rare, has been associated with the use of these agents. parisons. Selection of which agent to be used will be
In a small descriptive study, noninvasive evaluation suc- based on adverse effect profile, ease of use, familiar-
cessfully identified the probable cause of syncope in most ity, and beliefs about the importance of the differ-
patients with AD treated with donepezil [62]. Therapeutic ences between the agents in their pharmacokinetics
options would include stopping the agent or pacemaker and other mechanisms of action (Grade B, Level I).
implantation [63]. Donepezil has been associated with sleep C. All physicians prescribing these agents should be
disturbances, vivid dreams/nightmares, and hypnopompic aware of the contraindications and precautions with
hallucinations [60]. The Cochrane meta-analysis of donepe- the use of cholinesterase inhibitors (Grade B, Level
zil confirmed a dose-related increased odds ratio for insom- III).
nia [47]. Rivastigmine and galantamine are less likely to D. If adverse effects occur with a cholinesterase inhib-
cause sleep disturbances. Management options for this itor, the agent should either be discontinued (if the
problem would include changing the timing and/or dose of side effects are judged to be disabling and/or danger-
donepezil or switching to another cholinesterase inhibitor. ous), or the dose of the agent should be decreased,
A prescribing cascade involves the misinterpretation of with an option to retry the higher dose after 2 to 4
an adverse reaction to one drug followed by the prescription weeks if the lower dose is tolerated (if the side effects
of potentially inappropriate second drug to deal with this are judged to be minor in severity) (Grade B, Level
adverse effect. The use of cholinesterase inhibitors is asso- III).
ciated with an increased risk of receiving an anticholinergic E. If nausea and/or vomiting occur with the use of a
drug to manage urinary incontinence [64]. The use of an cholinesterase inhibitor, review how the medication
anticholinergic drug in this setting might represent a clini- is being taken (eg, dose, frequency, with or without
cally important prescribing cascade. Another potential ex- food, evidence of an unintentional overdose) and con-
ample of this would be the higher use of hypnotics in sider modifying the prescription (eg, lower dose),
patients with AD treated with donepezil [65]. responsibility for administration (eg, caregiver taking
A number of studies have shown that it is possible to over from the patient), the directions given to the
switch from one cholinesterase inhibitor to another [66 –72]. patient (eg, with food), or stopping the agent. Al-
Generally in these studies patients abruptly discontinued the though antiemetics can be used for nausea and/or
first cholinesterase inhibitor and started taking the second vomiting, a number of them (eg, dimenhydrinate,
agent the following day at the usual starting dose followed prochlorperazine) have anticholinergic properties that
by up-titration at the usual rate for the new agent. Unfortu- can lead to adverse cognitive effects (Grade B, Level
nately, these studies do not tell us when we should switch. III).
Commonly cited reasons for switching include unsatisfac- F. Clinicians should consider the possible contributing
tory response to the first agent, intolerable side effects to the role of cholinesterase inhibitors in new-onset or wors-
first agent, and request of the patient and/or caregiver. ening medical presentations and the potential risk of
Switching from donepezil to memantine was well-tolerated co-prescribing cholinesterase inhibitors and other
in a study sponsored by Lundbeck, whether it was done drugs to patients with dementia (Grade B, Level II-2).
abruptly (donepezil discontinued abruptly with memantine G. Patients can be switched from one cholinesterase
up-titrated to 20 mg/d during a period of 3 weeks) or inhibitor to another. A decision to make a switch is
gradually (donepezil dropped from 10 mg/d to 5 mg/d for 2 based on the judgment of the prescribing physician
weeks before stopping with memantine up-titrated to 20 and the patient (or their proxy) about the relative
mg/d during a period of 3 weeks) [73]. Please note that benefits and risks of making a change in the patient’s
switching from one cholinesterase inhibitor to another cho- pharmacotherapy (Grade B, Level III).
linesterase inhibitor or memantine can lead to deterioration H. Patients can be switched from a cholinesterase inhib-
in the status of a patient. Patients and families should be itor to memantine (note: see recommendation 15B).
informed of this possibility before a switch is made. The decision of when to make a switch is based on
the judgment of the prescribing physician and the
5.2. Recommendation 14: Recommendations regarding the patient (or their proxy) (Grade B, Level III).
use of cholinesterase inhibitors
Memantine is a low to moderate affinity, uncompetitive
A. All three cholinesterase inhibitors available in Canada antagonist to glutamate N-methyl-D-aspartate (NMDA) re-
are modestly efficacious for mild to moderate AD. They ceptors and might prevent excitatory neurotoxicity in de-
are all viable treatment options for most patients with mentia. Published studies show a small beneficial effect of
mild to moderate AD (Grade A, Level I). memantine in moderate to severe AD [74]. It has not been
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 363

shown to be effective for mild stages of AD. Although the F. The comorbidities of the patient make continued use
one published study showed efficacy on cognitive, behav- of the agent either unacceptably risky or futile (eg,
ioral, and global outcomes, two additional unpublished terminally ill); or
studies of memantine failed to show statistically significant G. The patient’s dementia progresses to a stage where
benefits for patients with mild AD [74,75]. there is no significant benefit from continued therapy
The NMDA receptor antagonist memantine and the cho- (Grade B, Level III).
linesterase inhibitors have different mechanisms of action.
It seems reasonable to assume that an additive effect might 5.5. Recommendation 17
be achieved from combination therapy. One RCT did show
additional benefit when memantine was added to chronic After stopping therapy for AD, patients should be care-
donepezil therapy in patients with moderate to severe AD fully monitored, and if there is evidence of a significant
[76]. An unpublished study of memantine failed to show decline in their cognitive status, functional abilities, or the
any statistically significant benefit when it was given to development/worsening of behavioral challenges, consider-
participants with mild to moderate AD who were on a stable ation should be given to reinstating the therapy (Grade B,
dose of a cholinesterase inhibitor [75]. Level III).
Both basic and clinical studies have examined whether
5.3. Recommendation 15: Recommendations regarding the antioxidants might be beneficial for individuals with AD
use of memantine [78,79]. It remains unclear whether reactive oxidative spe-
cies are a cause or are a consequence of AD pathology. A
A. Memantine is an option for patients with moderate to number of studies have examined the natural antioxidants
severe stages of AD (Grade B, Level I). Its use in found in foods, vitamins E and C, and carotenes. In vitro
mild stages of AD is not recommended (Grade D, studies have shown that vitamin E can decrease both
Level I). amyloid-induced lipid peroxidation and oxidative stress
B. Combination therapy of a cholinesterase inhibitor and while suppressing the inflammatory signaling cascade
memantine is rational (because the medications have [79,80]. Vitamin C can block the creation of nitrosamines
different mechanisms of action), appears to be safe, through the reduction of nitrates and might also affect cat-
and might lead to additional benefits for patients with echolamine synthesis. Carotenes can modify lipid peroxida-
moderate to severe AD. This would be an option for tion [80]. Cohort studies examining the relationship be-
patients with AD of a moderate severity (Grade B, tween antioxidant intake and the likelihood of subsequent
Level I). dementia have shown equivocal results [81– 83]. Uncer-
tainty persists as to the optimal agent, timing, duration,
An area of clinical uncertainty is when to stop a cho-
dosage, and mode of intake [79]. There has been one high
linesterase inhibitor (or any other agent being given for
quality RCT that examined the utility of high dose vitamin
AD). There is general acceptance that these decisions
should be individualized and based on the balance between E (2,000 IU/d) in subjects with moderate AD [84]. Although
benefits and harm for the patient. This has become partic- this study suggested benefit, a recent RCT for subjects with
ularly contentious as a result of studies suggesting that mild cognitive impairment found none [85]. An older Co-
interrupting therapy for prolonged periods of time (eg, 6 chrane review concluded there was insufficient evidence of
weeks) can result in the loss of treatment benefits that efficacy of vitamin E in the treatment of AD to justify its use
cannot be recaptured [77]. [86]. Recent reports of a higher mortality rate among those
treated with high doses of vitamin E (400⫹ IU/d) supple-
5.4. Recommendation 16: Recommendations about when mentation have cast even further doubt on the advisability
medications for the treatment of cognitive and functional of using this antioxidant [87]. Treatment with idebenone, a
manifestations of AD should be discontinued synthetic analog of coenzyme Q10 that is an antioxidant, did
not slow cognitive decline in a 52-week RCT of 536 sub-
A. The patient and/or their proxy decision maker decide jects with AD [88].
to stop; A number of studies have examined the potential role of
B. The patient refuses to take the medication; vitamin supplementation. Homocysteine levels can increase
C. The patient is sufficiently nonadherent with the med- with a deficiency of any of vitamins B6, B12, and/or folic
ication that continued prescription of it would be acid [89]. In the Framingham study the incidence of both
useless, and it is not possible to establish a system for cerebrovascular disease and AD was increased if homocys-
the administration of the medication to rectify the teine levels were greater than 14 ␮mol/ L [90]. Other stud-
problem; ies, however, have failed to support the association between
D. There is no response to therapy after a reasonable higher homocysteine levels and AD [91,92]. Individuals
trial; with lower levels of folate and/or vitamin B12 have been
E. The patient experiences intolerable side effects; found to have a higher risk of developing dementia in some
364 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

[93] but not all studies [94]. Several trials of vitamin B12 for ase inhibition that might contribute to the potential benefits
AD have shown inconsistent results [95,96]. Cochrane re- of NSAIDs in the prevention or management of AD. Some
views for vitamins B1 (for AD), B6 (for cognition), B12 (for (ie, sulindac, indomethacin, flurbiprofen. ibuprofen) but not
cognition), and folic acid (for cognition and dementia) have all NSAIDs have been found to affect beta-amyloid depo-
been done. No conclusions could be drawn from the B1 sition and metabolism. It is possible that the negative
studies examined [97]. No methodically adequate B6 trials NSAID trials to date might have occurred because the
that involved people with dementia were found [98]. Two wrong NSAID was tested. For example, no RCTs have been
studies of people with dementia and low serum B12 levels completed that used ibuprofen [114]. A study of prednisone
were examined, but no statistically significant evidence of (20 mg for 1 month and then 10 mg for 1 year) in 132
cognitive benefits with B12 supplementation was found [99]. patients failed to show any significant benefit [115]. An
No benefits on any measure of cognition or mood from folic RCT of hydroxychloroquine was also negative [116].
acid with or without vitamin B12 supplementation were seen The large RCTs of inhibitors of the 3-hydroxy-3-
in patients with a dementia [100]. Although more studies are methylglutaryl coenzyme A (HMG-CoA) reductase enzyme
needed, the routine administration of any of these vitamins (ie, “statins”) have had primary cardiovascular outcomes,
to individuals with a dementia who do not have a docu- although some of the studies have included secondary cog-
mented deficiency state cannot be endorsed at the present nitive outcomes. The MRC/BHF Heart Protection Trial of
time. simvastatin found no beneficial impact with active treatment
Ginkgo biloba is an ancient Chinese herbal preparation on the likelihood of either cognitive decline or being diag-
that is commonly used for the treatment of cognitive im- nosed with a dementia [117]. The PROSPER trial of prav-
pairment and dementia. Ginkgo is one of the five top pre- astatin also could not detect any cognitive benefit with
scribed products in Germany, and in North America it is the active therapy [118]. On the other hand, a 12-month
top-selling herbal remedy [101]. A review of the published placebo-controlled pilot trial of atorvastatin 60 mg in 71
studies of ginkgo concluded there was a small but statisti- patients with mild to moderate AD showed statistically
cally significant benefit seen with ginkgo compared with the significant improvement on the ADAS-Cog at 6 months and
placebo arm. Although the typical benefit seen was consis- a positive trend at 12 months [119]. Additional trials are
tently less than that obtained with the cholinesterase inhib- ongoing.
itors, ginkgo was better tolerated [102]. A Cochrane review Estrogens have a number of potentially beneficial effects
of Ginkgo biloba for cognitive impairment and dementia for women with dementia. A Cochrane review examined the
concluded that the agent appeared both safe and promising. effect of hormone replacement therapy on cognition in
Concerns were expressed, however, about the methods used women with a dementia. Five double-blind RCTs that in-
in a number of the earlier trials. The more methodologically cluded 210 women were evaluated in detail. Short-lived and
sound trials showed inconsistent results [103]. Fifteen pub- clinically insignificant positive effects with conjugated
lished case reports have described a temporal association equine estrogens (CEEs) were found on the MMSE (CEE,
between use of ginkgo and a bleeding event [104]. In 13 of 0.625 mg/d only), Trail-Making Test-B (CEE, 0.625 mg/d
the case reports other identified risk factors for bleeding only), and digit span backwards (CEE, 1.25 mg/d only).
were present. Patients using ginkgo, particularly those with Cued delayed recall of a word list was positively affected after
known bleeding risks (eg, concurrent use of warfarin or 2 months of transdermal diestradiol. Control subjects did sig-
antithrombotics), should be counseled about a possible in- nificantly better on delayed recall (1 month), finger tapping (12
crease in bleeding risk. months), and on the Clinical Dementia Rating scale. The pos-
A number of lines of evidence suggest that there are itive effects seen were believed to be possibly from random
increased levels of inflammatory mediators (eg, interleukins effects caused by multiple analyses. After correction for mul-
1 and 6, tumor necrosis factor, complement) in the brains of tiple testing only the short-term effect of transdermal estrogen
those with AD [105,106]. Microglia cells are activated remained statistically significant [120].
around amyloid plaques [107]. There is epidemiologic evi- Androgens (the primary androgen is testosterone; it can
dence that patients on anti-inflammatory agents have a re- be metabolized to the more potent androgen, dihydrotestos-
duced incidence of AD [108,109]. The RCTs that use anti- terone) can influence brain function directly through inter-
inflammatory drugs (eg, naproxen) as therapy for AD, actions with androgen receptors or indirectly through estra-
however, have been unsuccessful to date [110]. Initial trials diol (testosterone is converted to estradiol by aromatase)
with nonsteroidal anti-inflammatory drugs (NSAIDs) were [121]. In vitro and animal studies indicate that androgen
plagued with high dropout rates [111]. A systematic review depletion is associated with higher brain levels of beta-
concluded that indomethacin cannot be recommended for amyloid, hyperphosphorylation of tau protein, and de-
the treatment of mild to moderate AD [112]. Selective creased neuronal survival after exposure to a toxin [121–
cyclooxygenase 2 inhibitors were better tolerated, but the 123]. Studies of healthy older men suggest that therapy with
trials of both rofecoxib and celecoxib were negative testosterone has a weak and inconsistent association with
[111,113]. There are mechanisms other than cyclooxygen- better visuospatial and memory scores on testing [122].
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 365

Some but not all studies have shown an association between the treatment of dementia. Further methodologically
reduced testosterone levels and a diagnosis of AD [124]. sound trials are required (Grade C, Level I).
Two small intervention studies that included subjects with E. The use of an anti-inflammatory drug is not recom-
AD have been done. In one study 10 hypogonadal nursing mended for the treatment of the cognitive, functional,
home patients with AD were randomized to either intramus- or behavioral manifestations of a dementia (Grade D,
cular testosterone enanthate 200 mg every 2 weeks or pla- Level I).
cebo [125]. Unblinded assessments at 3, 6, and 9 months F. The use of a HMG-CoA reductase enzyme inhibitor is
showed improvements on the ADAS-Cog, MMSE, and the not recommended for the treatment of the cognitive,
Clock Drawing Test. One patient became aggressive and functional, or behavioral manifestations of a dementia
developed hypersexual behaviour. No other problems were (Grade D, Level III).
noted. The second study was a randomized, double-blind, G. Hormone replacement therapy (estrogens combined
placebo-controlled 6-week trial that examined the effects of with a progestagen) or estrogen replacement therapy
weekly intramuscular injections of 100 mg of testosterone (estrogen alone) is not recommended for the cognitive
enanthate on subjects with mild cognitive impairment or impairments of women with AD (Grade D, Level I).
AD (a total of 15 subjects with AD were enrolled) [126]. H. There is insufficient evidence to recommend the use
Improvements in spatial memory/ability and verbal memory of androgens (eg, testosterone) to treat AD in men
were seen with testosterone therapy. No adverse effects (Grade C, Level I).
were encountered. Both groups of researchers believed that I. There is negative, inconclusive, or conflicting evi-
additional studies were required. dence for a number of other agents proposed as
A large number of other agents with diverse mechanisms potential therapies for the cognitive and behavioral
of action have been tested as potential therapies for AD. A manifestations of AD. Their use cannot be recom-
partial listing (in alphabetical order) of these agents would mended at this time (Grade C or D, Levels I to III,
include acetyl-L-carnitine; active or passive beta-amyloid varies between agents).
immunization; Alzhemed; aniracetam; BMY21,502; be-
sipiridine; cerebrolysin; clioquinol; cytidinediphosphocho-
line (CDP-choline); D-cycloserine; DGAVP; dehydroepi- 6. Nonpharmacologic and pharmacologic therapy of
androsterone; doxycycline and rifampin; erythropoietin; behavioral and mood disturbances
extract of Melissa officinalis; garlic; gamma-secretase in-
hibitor; growth hormone releasing hormone; huperzine A; Neuropsychiatric symptoms of dementia, also known as
hydergine; ispronicline; lethicin; lithium carbonate; melato- behavioral and psychological symptoms of dementia
nin; milacemide; neramexane; nicergoline; nicotine; nimo- (BPSD), occur in the majority of people with dementia over
dipine; paclitaxel; phosphatidyl serine; physostigmine; the course of the disease [127]. Disease severity affects the
piracetam; propentofylline; rosiglitazone; selegiline; prevalence of psychiatric symptoms. Major depression oc-
velnacrine; and vinpocetine. The studies of these agents curs more often in mild to moderate cognitive impairment,
have been either negative or inconclusive. None have whereas most other symptoms are considered more com-
been approved for the treatment of AD in Canada. mon with greater dementia severity [128,129]. However,
noncognitive behavioral symptoms do not correlate well
5.6. Recommendation 18: Recommendations with regard with each other. In some longitudinal studies they have not
to supplements, herbal preparations, and other shown progressive worsening with time but have appeared
medications for the cognitive and functional episodically [130]. In mild AD, some symptoms are com-
manifestations of AD and dementia mon. For example, Lopez et al [129] found that of those
with mild AD, 60% had anxiety, 55% had lack of energy,
50.5% had anhedonia, 49% had agitation, 39% had irrita-
A. High-dose (ie, 400⫹ IU/day) vitamin E supplemen- bility, and 25.5% had delusions or hallucinations. In early
tation is not recommended for the treatment of AD dementia psychotic symptoms (especially visual hallucina-
(Grade E, Level I). tions) are relatively uncommon with AD, but they occur
B. The use of the synthetic antioxidant idebenone is not more frequently with DLB.
recommended for the treatment of AD (Grade E, The typical rating scales used for psychiatric disorders
Level I). become progressively more difficult to use as the severity of
C. The administration of vitamin B1, B6, B12, and/or dementia increases. For example, whereas in mild AD in-
folic acid supplements to persons with AD who are struments such as the Geriatric Depression Scale [131]
not deficient in these vitamins is not recommended might still be valid in screening for depression, by the later
(Grade D, Level III). stages psychometric factors such as internal consistency
D. There is insufficient evidence to allow for a recommen- worsen [132], necessitating the use of alternate instruments.
dation either for or against the use of ginkgo biloba in Instruments available for rating behavioral and mood symp-
366 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

toms in more advanced dementia are addressed in more pressants might also be appropriate for first-line treatment of
detail in the section on severe dementia. depression [143]. Although data are sparse, electroconvulsive
treatment can be useful for patients with depression who have
6.1. Recommendation 19 not responded to antidepressants or who cannot tolerate
pharmacotherapy [144].
Assessment of patients with mild to moderate AD should
include measures of behavior and other neuropsychiatric 6.3. Recommendation 21: Recommendations with regard
symptoms (Grade B, Level III). to the management of depressive symptoms in the setting
of mild to moderate dementia
6.2. Recommendation 20
A. Because depressive syndromes are frequent in pa-
The management of BPSD should include a careful doc-
tients with dementia, physicians should consider di-
umentation of behaviors and identification of target symp-
agnosing depression when patients present with the
toms, a search for potential triggers or precipitants, record-
subacute development (eg, weeks, rather than months
ing of the consequences of the behavior, an evaluation to
or years) of symptoms characteristic of depression
rule out treatable or contributory causes, and consideration
such as behavioral symptoms, weight and sleep
of the safety of the patient, their caregiver, and others in
changes, sadness, crying, suicidal statements, or ex-
their environment (Grade B, Level III).
cessive guilt (Grade B, Level III).
Few data are available for the nonpharmacologic treat-
B. Depressive symptoms that are not part of a major
ment of depression in dementia. One controlled study [133]
affective disorder, severe dysthmia, or severe emo-
suggests that behavioral treatments, including those empha-
tional lability should initially be treated nonpharma-
sizing pleasant events (for the patient) and those emphasiz-
cologically (Grade B, Level III).
ing caregiver problem solving, might decrease depressive
C. If the patient had an inadequate response to the non-
symptoms in this group. A Cochrane review [134] exam-
pharmacologic interventions or has a major affective
ined high quality trials of antidepressants for depressive
disorder, severe dysthymia, or severe emotional la-
disorders in patients with dementia. Eight studies met their
bility, a trial of an antidepressant should be consid-
stringent inclusion criteria. The mean MMSE score before
ered (Grade B. Level III).
treatment in the studies ranged between 15 and 23. The
D. If an antidepressant is prescribed to a person with
authors concluded there was only weak support for the
AD, the preferred choice would be an agent with
efficacy of antidepressants in treating depression in patients
minimal anticholinergic activity, such as an SSRI
with dementia, but they noted that only two studies used
(Grade B, Level III).
selective serotonin reuptake inhibitors (SSRIs) and sample
sizes were small. The prevalence of sleep disturbance and complaints is
Although the treatment of depression in demented people high in older persons including those with dementia. Nor-
might not improve cognition [135], there is good evidence mal age-associated changes in sleep include less deep sleep
that it might improve the quality of life of patients and their and less rapid eye movement (REM) sleep and worse sleep
caregivers [136]. Therefore, most review articles [137] efficiency (defined as the amount of time sleeping over the
strongly recommend treatment of depression in people with amount of time spent in bed). The major sleep difficulty
AD. Antidepressants with significant anticholinergic activ- encountered in older individuals is a decreased ability to
ity (eg, tertiary amine tricyclics such as amitriptyline, imip- maintain sleep. A variety of factors such as medical and
ramine, trimipramine, and doxepin) are likely to worsen psychiatric illnesses, changes in the timing and consolida-
cognition [138] and should generally be avoided. The SSRIs tion of sleep (from changes in the endogenous circadian
generally have less anticholinergic activity (note: paroxetine rhythm), medications, the presence of other sleep disorders
has significantly more anticholinergic activity than other (eg, periodic limb movements in sleep [PLMS], REM sleep
SSRIs) [139], although they do have other side effects that behavior disorder [RBD], sleep-disordered breathing), en-
are of concern in frail older people (ie, gastrointestinal vironmental factors (eg, noise and light), and poor sleep
symptoms, weight loss, sleep problems, and hyponatremia) habits can all contribute to a decline in the quantity and/or
[140]. They have been associated with falls and hip frac- quality of sleep [145]. Sleep disturbances can affect up to
tures [141], possibly related to mechanisms that differ from 44% of community-dwelling persons with dementia [146].
those causing falls in tricyclics [142]. The choice of a spe- For family caregivers, nocturnal disturbances such as being
cific antidepressant therefore has to consider both its particular awakened at night by care recipients wandering or getting
advantages and potential adverse effects, individualizing the out of bed repeatedly are disturbing aspects of care and a
choice for each patient. Once a decision has been made to use major risk factor for institutionalization [147].
an antidepressant, of the classes of agents currently available, McCurry et al [148,149] evaluated the effectiveness of
SSRIs would be appropriate for first-line treatment of depres- the Nighttime Insomnia Treatment and Education for Alz-
sion in patients with AD. Other classes of nontricyclic antide- heimer Disease (NITE-AD) program in improving sleep.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 367

Caregivers in the treatment group received specific recom- nesses (including pain), psychiatric illnesses (espe-
mendations about setting up and implementing a sleep pro- cially depression), potentially contributing medica-
gram for their care recipient and training in behavior man- tions, environmental factors, and/or poor sleep habits
agement skills. Care recipients were also instructed to walk (eg, daytime naps) that might be adversely affecting
daily and increase daytime light exposure with the use of a sleep. Any identified secondary cause should be man-
light box. Control subjects received general dementia edu- aged (Grade B, Level III).
cation and caregiver support. After 2 months of treatment, B. The presence of an RBD in the setting of a dementia
care recipients exhibited significantly greater reductions in would be suggestive of DLB and related conditions.
number of nighttime awakenings, total time awake at night, Treatment options would include clonazepam (Grade
and depression. At 6-month follow-up, treatment gains were B, Level II-2).
maintained, and additional significant improvements in du- C. Nonpharmacologic approaches to sleep disturbances
ration of night awakenings emerged. Clinicians who recom- can be effective for patients with AD, but a combi-
mend these interventions should be aware that many nation of these approaches will likely be required
caregivers need active assistance with setting up and imple- (Grade B, Level I).
menting a sleep program. Simply providing caregivers with D. When considered clinically necessary, pharmaco-
education is often insufficient. A strength of this study is the logic interventions for insomnia, including short- to
combination of approaches (sleep program, walking, and intermediate-acting benzodiazepines and related
bright light) that individually might influence behavioral agents, can be used at the lowest effective doses and
problems. for the shortest possible time (Grade B, Level III).
Although the treatment of insomnia in the older patients
with the newer sedative hypnotics has been shown to be Several systematic reviews of approaches that address be-
effective and safe [145], a recent and very comprehensive havioral problems associated with dementia also have been
meta-analysis has found that the magnitude of effect was conducted [158 –161], with the most comprehensive review
small, and that the increased risk of adverse effects was of conducted by Livingston et al [162]. Specific types of psycho-
concern [150]. These adverse effects include ataxia, mem- social education for caregivers about managing behavioral and
ory loss, and falls, as well as impairment in driving ability. psychological symptoms were effective treatments whose ben-
There is also a risk of dependency and accidental overuse. efits lasted for months. Music therapy, Snoezelen, and possibly
There are no randomized clinical trials of sedative hypnotic sensory stimulation were beneficial during the treatment ses-
medications for sleep disturbance in AD [151], but these sion but had no long-term effects [162].
agents are widely clinically used. Their short-term use can Several Cochrane Reviews have examined the effect of a
be justified in situations in which sleep disturbance is par- variety of interventions in managing the symptoms of de-
ticularly problematic. The pharmacotherapy of specific mentia. They concluded that although some of the interven-
sleep disturbances such as PLMS and restless legs syn- tions studied look promising, they remain unproven. All of
drome are supported by a greater evidence base and might the following Cochrane Reviews included subjects with
include other pharmacologic interventions such as dopa- moderate to severe dementia and who resided in long-term
mine agonists. care facilities. A review of reminiscence therapy included
RBD is manifested by vivid, often frightening dreams four trials that revealed significant improvement in cogni-
during REM sleep but without atonia. Patients “act out their tion and mood 4 to 6 weeks after the treatment [163]. A
dreams” with vocalization, flailing of their limbs, and/or review of validation therapy included three studies that
moving around the bed. The history of RBD is obtained demonstrated no significant differences between validation
from the patient’s bed partner. RBD is frequently associated and social contact or between validation and usual therapy
with synucleinopathies including Parkinson’s disease and [164]. Five trials were included in a review on bright light
DLB. It is considered a suggestive feature of DLB [152]. therapy [165]. This review revealed insufficient evidence of
REM sleep reportedly has a cholinergic basis [153]. Al- the effectiveness of bright light in managing sleep, behav-
though this is an area requiring further study, treatment ior, cognitive, or mood disturbances associated with demen-
options for RBD in the setting of DLB would include tia. A review to assess whether music therapy can diminish
a clonazepam, cholinesterase inhibitor, melatonin, and behavioral and cognitive problems or improve social and
quetiapine [152,154 –157]. emotional functioning included five trials. However, there
was insufficient evidence to draw any useful conclusions
6.4. Recommendation 22: Recommendations with [166]. A review that assessed the efficacy of aroma therapy
regard to sleep problems in the setting of a mild to as an intervention for persons with dementia concluded that
moderate dementia the one small trial provided insufficient evidence [167]. A
review of Snoezelen (multi-sensory stimulation) included
A. Patients with AD experiencing sleep problems should two trials that demonstrated some short-term benefits in
first undergo a careful assessment for medical ill- promoting adaptive behaviors during and immediately after
368 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

participation in the sessions. However, the carryover and were randomized to either placebo or 10 mg of donepezil
longer-term effects of Snoezelen were not evident [168]. A and assessed at further intervals. Patients randomized to
review of the effectiveness of a range of massage and touch donepezil after the open-label segment had significantly
therapies offered to persons with dementia is currently be- more improvement on the NPI as well as on the NPI-distress
ing conducted [169]. measure compared with the placebo group. Discordant re-
The lack of demonstrated effectiveness in all of these sults were obtained in the AD2000 study [181]. In this
reviews does not mean that the interventions are ineffective, randomized, placebo-controlled, double-blind trial the ef-
but rather that there is a lack of sufficient evidence to fects of donepezil on 565 community-residing patients with
demonstrate effectiveness. More experimental studies with mild to moderate AD were studied. The NPI was used. The
larger samples and improved quality are needed to further study design was complicated, and as a result, its outcomes
examine the efficacy of these interventions with different are difficult to evaluate. Although the authors concluded
types and severity of dementia. Combining subjects with a that there was no significant improvement on any of their
variety of dementias and at various stages of their illness cognitive, functional, or psychiatric measures, it is unclear
does not contribute to our understanding of the efficacy of what conclusions can be drawn from this study. A Cochrane
these interventions. review [49] of donepezil for AD evaluated the available
Most trials of cholinesterase inhibitors have been done data on the effects of treatment on behavior (as measured by
on those with a mild to moderate severity of their dementia. the NPI). The authors concluded that there was a significant
Many of these trials have included caregiver-rated scales benefit seen with donepezil. Compared with the placebo
that measure behavioral and psychiatric symptoms such as arm, there was a mean difference of 6.20 at 6 weeks and 3.2
the Neuropsychiatric Inventory (NPI) [170], the Cornell at 24 weeks favoring active therapy on the intent to treat
Depression Rating Scale [171], the Behavioral Rating Scale (ITT)–last observation carried forward (LOCF) analysis. An
for Dementia [172], the noncognitive subscale of the ADAS article by Seltzer et al [182] was not included in the Co-
[173], the Behavioral Pathology in Alzheimer’s Disease chrane review because they used an apathy scale [183] as
Rating Scale [174], the Gottries-Brane-Steen scale (GBS) their behavioral measure. This trial was a double-blind,
[175], and the Cohen-Mansfield Agitation Inventory [176]. 24-week, placebo-controlled study of donepezil in 153 pa-
Trinh et al [177] performed a meta-analysis that exam- tients with early AD. The active treatment group scored
ined the efficacy of cholinesterase inhibitors in the treatment better on the apathy scale, although this was not statistically
of the neuropsychiatric symptoms and the functional im- significant.
pairments that arise with AD. They reviewed 29 parallel Rösler al [184] used the CIBIC-Plus to measure the
group or crossover randomized, double-blind, placebo- effects of rivastigmine treatment compared with placebo in
controlled trials of patients with mild to moderate probable 98 patients with mild to moderate AD. This was a 26-week
AD. Patients randomized to cholinesterase inhibitors im- randomized, placebo-controlled trial followed by an open-
proved on average 1.72 points on the NPI and 0.03 points on label extension period. A variety of behavioral and psychi-
the ADAS-noncog compared with those on placebo. There atric measures derived from the CIBIC-Plus showed sig-
were no significant differences between the various cho- nificant improvements with rivastigmine compared with
linesterase inhibitors. The authors concluded that there was placebo. Finkel [185] published a meta-analysis of three
a modest improvement of neuropsychiatric symptoms with 6-month, placebo-controlled trials of rivastigmine in mild to
the use of cholinesterase inhibitors. moderate AD. He found that patients on rivastigmine with
Cummings et al [178] used a caregiver mailout survey to neuropsychiatric symptoms at baseline exhibited significant
compare 84 patients taking donepezil with 248 patients not improvements with paranoid and delusional thoughts as
on it. They found that those taking donepezil were signifi- well as in aggression. Patients who did not have significant
cantly less likely to be threatening, destroy property, and neuropsychiatric symptoms at study entry were less likely to
talk loudly. Although the treatment groups were not ran- see their emergence if treated with rivastigimine, but this
domly assigned, supportive evidence for donepezil having a was not statistically significant for hallucinations.
positive effect on difficult behaviors was the decreased rate Herrmann et al [186] performed a post hoc analysis of
of use of sedatives in those on the agent. Winblad et al [179] pooled data from three large trials (2,033 subjects) of ga-
studied 286 patients with mild to moderate AD by using the lantamine treatment for mild to moderate AD. They found
GBS scale (which includes various behavioral and emo- that compared with those on placebo, treatment with galan-
tional items) and the NPI. There were nonsignificant im- tamine was associated with a better total NPI score and
provements on the emotional/behavioral items of the GBS better scores on specific NPI items that included agitation/
and on the NPI seen in those patients receiving donepezil aggression, anxiety, disinhibition, and aberrant motor be-
compared with those allocated to the placebo group. havior. Similar beneficial results were found in a priori
Holmes et al [180] examined the efficacy of donepezil in the defined symptom clusters 1 (delusions, hallucinations), 3
treatment of neuropsychiatric symptoms in AD by using the (disinhibition, elation, aberrant motor behavior), and 4 (hal-
NPI. After an initial open trial of donepezil, 134 patients lucinations, anxiety, apathy, aberrant motor behavior). The
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 369

authors believed that the cluster of hallucinations, anxiety, 6.5. Recommendation 23: Recommendations with regard
apathy, and aberrant motor behaviors might represent a to the management of BPSD in the setting of a mild to
specific group of cholinergic-responsive behavioral symp- moderate dementia
toms. A Cochrane review of galantamine for AD and mild
cognitive impairment examined 10 trials with a total of A. Nonpharmacologic treatment of BPSD should be
6,805 subjects [50]. Treatment effects on the NPI were considered first. Nonpharmacologic interventions are
reported at 3 months in one trial and at 6 months in three often used in combination with pharmacotherapy
trials. There was no statistically significant treatment effect (Grade C, Level I).
at 3 months for the 24 to 32 mg per day dosage (OC and B. Although there is insufficient evidence regarding the
ITT), but at 6 months there was a significant treatment effect effectiveness of the interventions to strongly advo-
(OC and ITT) for 16 mg per day. cate for their routine use in the management of
Areosa Sastre et al [187] performed a Cochrane review BPSD, some persons with dementia might benefit
of the use of memantine for dementia. They identified only from the following: music, Snoezelen (multi-sensory
one reported trial for mild to moderate AD [188]. In this stimulation), bright light therapy, reminiscence ther-
trial, NPI scores at 24 weeks were significantly better (3.5 apy, validation therapy, aroma therapy, and massage
points) if the patients received memantine compared with and touch therapy (Grade C, Level II-3).
those allocated to the placebo arm. However, there were two C. Pharmacotherapy for BPSD should be initiated only
completed but unpublished studies that examined subjects after consideration, and usually a trial where appro-
with mild to moderate AD. Their specific behavioral results priate, of nonpharmacologic interventions (Grade B,
Level III).
are not available for detailed evaluation. This makes it
D. The presence of visual hallucinations in the setting of
difficult to make any conclusions at this time.
mild dementia would suggest that the patient has
RCT data on the effects of cholinesterase inhibitors and
DLB. Patients with DLB are abnormally sensitive to
memantine on the BPSD come principally from secondary
antipsychotics. If pharmacotherapy is required for
outcome measures of trials primarily designed to measure the visual hallucinations, a cholinesterase inhibitor
cognitive and global outcomes in subjects with a low fre- should be tried first if possible. If acute symptom
quency and severity of BPSD. With few exceptions, these control is required or the cholinesterase inhibitor is
studies were not designed to look at BPSD as the primary ineffective, a cautious trial of an atypical antipsy-
outcome. The available data have to be interpreted with this chotic (eg, very low dose quetiapine) can be at-
in mind. The benefits seen in the studies done to date might tempted (Grade B, Level II-2).
not apply to patient populations with more severe baseline E. Medications for BPSD should normally be initiated at
neuropsychiatric abnormalities. a low starting dose and then subsequently titrated
Antipsychotics, antidepressants, anticonvulsants, and carefully on the basis of the patient’s response and the
other medications have also been used for the treatment of presence of adverse effects (Grade B, Level III).
neuropsychiatric symptoms of dementia. However, most of F. There should be periodic attempts to taper and with-
the trials of these agents have been conducted in more draw medications after a period of 3 months of be-
severely impaired populations (see section on severe AD for havioral stability (Grade B, Level III).
information and recommendations on specific agents). G. Patients who have mild to moderate AD and neuro-
Patients with psychotic features and a mild dementia psychiatric symptoms can be considered for a trial of
might be prescribed an antipsychotic. Patients with DLB a cholinesterase inhibitor and/or memantine for these
exhibit an abnormal sensitivity to antipsychotics, and they symptoms (Grade B, Level III).
should be avoided if possible [152]. The presence of visual H. Treatment of BPSD with cholinesterase inhibitors or
hallucinations and visuospatial/constructional dysfunction memantine should persist until clinical benefits can
(eg., problems copying the intersecting pentagons on the no longer be demonstrated (Grade B, Level III).
MMSE) early in the course of the dementia would be Behavioral disturbances have been reported to occur in
suggestive of DLB [189]. In this autopsy-confirmed study, 63% of community-dwelling persons with dementia [190].
the positive predictive value for DLB was 83% for visual These disturbances increase distress for those with demen-
hallucinations, whereas the lack of visuospatial impairment tia, increase the strain for caregivers, and might be poten-
had the best negative predictive value (90%). Treatment tially dangerous for the care recipient, caregivers, and oth-
options for visual hallucinations in the setting of DLB ers. Those with behavioral disturbances enter long-term
would include cholinesterase inhibitors and/or a cautious care facilities nearly 2 years earlier than those without
trial of an atypical neuroleptic [152]. Of the available atyp- [191]. Dementia-related behavioral disturbances that most
ical neuroleptics, quetiapine would appear to be an attrac- frequently occur in the home setting are wandering, general
tive choice, but controlled trials are needed [152,157]. restlessness, agitation, and uncooperativeness [192].
370 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

In 1994, the Canadian Study on Health and Aging medical care [202,203]. The study investigated whether
(CSHA) [193] reported that half of those diagnosed with increased access to community care and case management
dementia live in the community and are cared for by family led to a reduction in health care use and expenditures. Only
and friends who must deal with the long-term and disabling a tendency toward reduced health care expenditures was
behavioral problems associated with the care recipient’s observed in the treatment group. Case management did not
dementia. In 2000, the proportion of persons with dementia result in increased access to health care or to the prevention
who live at home had risen to 80% [194]. Although com- of conditions that might increase expenditures. Perhaps case
munity services have been shown to be useful in prolonging manager involvement produces short-term increases in ex-
independence and quality of life of those with dementia, the penditures, and a longer time period (greater than 3 years) is
CSHA revealed that community services are underutilized. required to demonstrate the benefits. The demonstration
Sixty-nine percent of spouse caregivers and 46% of care- study was also not designed to promote collaboration be-
givers who were sons or daughters used no services, and tween the case manager and other health care practitioners
only 3% to 5% of caregivers used three or more services in identifying and managing high-risk people. Further re-
[195]. Only 38% of Canadians with dementia received search is necessary to demonstrate and evaluate the role
home care services in 2000/2001 [196]. Several barriers to of case managers in managing behavioral problems of
using home care services have been identified: cultural and community-dwelling persons with dementia.
ethnic factors, a reluctance to use formal services until Adult day care programs provide supervised, structured
absolutely necessary, a perception that caregiving is a fa- activities for the care recipients during the day, enabling
milial responsibility, acknowledging their family member their caregivers to rest or tend to other responsibilities.
has dementia and they are unable to manage, lack of aware- Several studies have evaluated the effect on behavior prob-
ness, acceptability and accessibility of services (eg, distance lems of persons with dementia. One study demonstrated that
especially in rural areas, costs, inadequate training of ser- behavioral difficulties such as wandering, agitation, and
vice providers), and challenges in service delivery (care anger were not found to predict use of adult day care
often needed 24 hours a day) [197–199]. programs [204]. Several studies revealed that combined
Although community-based programs have primarily fo- family support (patient and family supported by one pro-
cused on caregivers [200], in this section we will highlight fessional staff member) while participating in an adult day
community-based programs and interventions that aim to care program was more effective in decreasing behavior
manage the behavioral disturbances associated with mild to problems (including inactivity and nonsocial behaviour),
moderate dementia. Interventions that are of benefit to care- improving mood, improving caregiver’s level of confidence,
givers are described elsewhere. Included studies in this and delayed nursing home placement when compared with
review had to meet the following criteria: (1) the majority of those who received psychogeriatric day care only. The sup-
subjects were diagnosed with AD or related dementia, (2) port offered to caregivers included informational, practical,
the majority of subjects were classified as having mild to emotional, and social support [205–207].
moderate dementia (defined as MMSE score of 肁10 or a Support groups for caregivers are widespread in health
Global Deterioration Scale score of 3 to 5 [201], and (3) the and voluntary organizations. However, the majority of ex-
sample size was at least 10. The term caregivers refers to perimental studies have been unable to demonstrate any
unpaid caregivers who are usually family or friends of the significant effect in decreasing caregiver burden and stress
care recipient and who provide support and assistance in the or on patient behavioral and psychological symptoms [208 –
activities of daily living and instrumental activities. 210]. Hébert et al [211] addressed some of the limitations of
Case management involves an assessment of client and these previous studies by offering the group program during
caregiver needs and the development, implementation, and a longer period (15 two-hour weekly sessions), incorporat-
monitoring of a care plan that can maintain a client safely in ing a specific theoretical framework, and focusing on the
the community. The care plan typically involves the ar- management of behavioral problems and the reactions they
rangement and coordination of a number of in-home and created. This experimental study demonstrated a 14% de-
community-based services such as housekeeping, personal crease in reactions to the behavioral problems of the care
care, and respite services to supplement care already re- receivers compared with a 5% decrease in the control group.
ceived from family and friends. Case management activity The frequency of behavioral problems also decreased. Fo-
has been shown to be reactive and focused on dealing with cusing on immediate, tangible outcomes that are expected to
the consequences of the behavioral problems rather than change as a result of the intervention (such as behavioral
addressing the management of the behavior [192]. A dem- problems) rather than more long-term, global outcomes
onstration project randomly assigned individuals with de- (such as preventing institutionalization and enhancing well-
mentia and their caregivers to a treatment group that re- being) are important lessons learned from this study.
ceived community-care services (eg, homemaking, personal More expensive home-based programs have also dem-
care, companion services, and adult day care) and case onstrated a significant effect in managing the disturbing
management or to a control group that received regular behaviors associated with dementia. In a Finnish study
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 371

Eloniemi-Sulkava et al [212] compared a 2-year program C. In-home systematic, comprehensive support by a


that consisted of systematic, comprehensive support by a health care provider with advanced training in de-
dementia family care coordinator (nurse) with conventional mentia care during an extended period (ie, couple of
care. The coordinator addressed health problems of the care years) (Grade B, Level I).
recipient and caregiver, behavioral and psychological symp- D. In-home psychoeducational intervention that teaches
toms of the care recipient including restlessness, anger, caregivers how to manage behavioral problems
delusions, aggression, and depression, and caregiver stress (Grade B, Level I).
and burden. Educational courses were offered annually. The
support of the coordinator deferred placement in long-term 7. Driving
institutionalized care, especially for persons with severe
dementia. The authors recommend that the intervention be In North America, driving a motor vehicle is an impor-
targeted especially at persons with behavioral problems that tant aspect of modern culture that has almost become an
threaten their ability to continue to remain in their home. activity of daily living. The ability to drive is often needed
A less intensive psychoeducational intervention, based to maintain independent mobility [220]. It is also an expres-
on the Progressively Lowered Stress Threshold Model sion of autonomy and independence and contributes to
[213], was offered in home during two 2-hour sessions to many important aspects of a person’s quality of life, includ-
teach caregivers how to manage behavioral problems (eg, ing maintenance of family/social ties and participation in
structured routine, environment modifications, decreased recreational activities. For all individuals, including those
environmental stimuli) [214]. This study was conducted in with physical, mental, or functional disabilities, the ability
five states in the United States. The intervention had a to drive permits the continuation of independent living.
positive impact during a period of 12 months on the fre- Loss of driver licensure for an older person can have a
quency of and response to problem behaviors among spou- direct health impact, with increases in depressive symptom-
sal caregivers, and nonspousal caregivers reported a reduc- atology [221]. Increased loneliness, social isolation, and
tion in the frequency of memory/behavioral problems. An stress on family and friends have been linked to the loss of
experimental study conducted in Taiwan demonstrated that the ability to drive in older persons [222–225], with this
a two-session in-home caregiver training program and tele- impact tending to be greater for those in rural areas [226].
phone consultations every 2 weeks for 3 months decreased Even for those in urban areas, public transportation systems
physically nonaggressive behavior, verbally aggressive and do not adequately replace the mobility and freedom of
nonaggressive behavior (but not physical aggression), and operating one’s own motor vehicle [227]. Older drivers
improved caregivers’ self-efficacy for managing these be- often feel angry and frustrated with those who deem them
haviors. The subjects’ level of severity of dementia is not unfit to drive, straining personal and professional relation-
reported [215]. Other similar behavioral intervention studies ships [228]. However, as much as it is desirable to promote
[216] have not been able to demonstrate a significant effect personal independence, the safety implications of driving
in managing disturbing behaviors, possibly as a result of from an individual and societal perspective require careful
their small sample sizes and short duration of the programs. consideration.
The remaining retrieved community-based studies did Given that many forms of dementia are progressive in
not meet our inclusion criteria because the sample sizes nature, and unlike other medical conditions, persons with
were less than 10 [217,218], or the subjects’ MMSE scores dementia often lack the insight to curtail their driving ex-
were less than 10 [219]. posure in the face of an increased crash risk, some authors
have advocated for the suspension of driving privileges in
6.6. Recommendation 24: Recommendations concerning all persons who are diagnosed with dementia [229]. How-
community-based programs for the management of ever, some studies have shown that some older persons in
behavioral disturbances the early stage of dementia are able to safely operate a
motor vehicle [230,231]. Research and public policy have
For the following community-based programs for the been primarily focused on cognitive and visuoperceptual
management of behavioral disturbances, there is limited deficits as they relate to driving [232–235]. Cognitive def-
high quality evidence regarding effectiveness. The recom- icits affecting driving include memory impairment, poor
mendations are based on one to two RCTs for each program. sequencing skills, impaired insight and judgment, apraxia,
and slowed processing time [236 –243]. Visuoperceptual
A. Adult day care (greater involvement of the caregiver deficits are an important subset of cognitive skills directly
might decrease problem behaviors in the care recip- related to driving ability [244 –246].
ient) (Grade B, Level II-2).
7.1. Dementia and driving risk
B. Support groups that focus on the management of
behavioral problems and extend for a period of sev- Table 2 summarizes controlled studies that have exam-
eral months (Grade B, Level I). ined driving risk in patients with dementia. A total of 25
372 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

Table 2
Summary of controlled studies determining motor vehicle crash risk in persons with dementia [229 –232,282–302]
Study (Author, Date) Methodology Outcome Measure Used Main Findings

Waller et al [300], Case control: 99 AD patients; 495 unmatched State driving record No difference in crash rates
1993 controls
Cooper et al [284], Case control: 165 dementia patients; 165 age-, State driving record Dementia patients 2⫻ more likely to
1993 sex matched have had a crash
Tuokko et al [298], Case control: 249 AD patients; 249 age-, sex State driving record AD patients 2.5⫻ more likely to have
1995 matched controls had a crash
Trobe et al [230], 1996 Case control: 143 AD patients; 715 age-, sex State driving record No difference in crash rates
matched controls
Carr et al [231], 2000 Case control: 63 AD patients; 58 unmatched State driving record No difference in crash rates
controls
Drachman et al [287], Case control: 130 dementia patients; 112 Caregiver reported crashes Dementia patients 2.5⫻ more likely to
1993 unmatched controls have had a crash
Friedland et al [229], Case control: 30 AD patients; 20 age-matched Caregiver reported crash past 5 y AD patients 8⫻ more likely to have had
1988 controls a crash
Zuin et al [302], 2002 Case control: 56 dementia patients; 31 Caregiver reported crashes More frequent crashes in dementia
unmatched controls patients (P ⫽ .12)
Ott et al [294], 2003 Case control: 27 mild dementia patients; 40 Caregiver reported driving ability More frequent crashes in dementia
unmatched controls patients
Hunt [291], 1989 Case control: 12 questionable dementia patients; On road performance More mild dementia patients failed test
14 mild dementia patients 13 age matched (P ⬍ .05)
controls
Hunt [292], 1993 Case control: 12 very mild dementia patients; On road performance 5/13 mild failed test; all controls and
13 mild dementia patients; 13 unmatched very mild passed
controls
Rebok et al [295], Case control: 10 AD patients; 12 unmatched On road performance Worse performance in AD patients
1994 controls
Fitten et al [232], 1995 Case control: 25 mild dementia patients; 24 On road performance Worse performance in dementia patients
age-matched controls
Cushman [286], 1996 Case control: 32 early AD patients; 91 On road performance More AD patients failed test
unmatched controls
Hunt et al [293], 1997 Case control: 36 very mild AD patients; 29 On road performance Worse performance in AD patients
mild AD patients; 58 unmatched controls
Wald [299], 1998 Case control: 112 dementia patients; 50 On road performance Worse performance in dementia patients
unmatched controls
Duchek et al [288], Case control: 49 very mild AD patients; 29 On road performance Worse performance in AD patients
1998 mild AD patients: 58 unmatched controls
Bieliauskas et al [282], Case control: 9 AD patients; 9 age matched On road performance More driving errors in AD patients
1998 controls
Duchek et al [289], Case control: 21 very mild AD patients; 29 On road performance More rapid decline in driving skills in
2003 mild AD patients; 58 unmatched controls dementia patients
Whelihan [301], 2005 Case control: 23 mild dementia patients; 23 On road performance Worse performance in dementia patients
age-matched controls
Harvey [290], 1995 Case control: 13 dementia patients; 125 Driving simulator performance Worse performance in dementia patients
unmatched controls
Rizzo et al [296], 1997 Case control: 21 AD patients; 18 unmatched Driving simulator performance Higher crash rate in AD patients
controls (P ⫽ .02)
Cox et al [285], 1998 Case control: 29 AD patients; 21 age-matched Driving simulator performance Worse performance in AD patients
controls
Rizzo et al [297], 2001 Case control: 18 AD patients; 12 unmatched Driving simulator performance Higher crash rate in AD patients
controls (P ⬍ .05)
Carr et al [283], 1998 Case control: 70 dementia patients; 667 Traffic sign recognition test Worse performance in dementia patients
unmatched controls

studies were identified, all using a case-control design. In- one with the ability to recognize traffic signs. Similar results
creased driving risk for patients with dementia was found in were found for studies that examined patients with AD only.
two of five studies with state driving records as their out- All studies that used a form of driver performance (on-road
come, four of four with caregiver report of collisions/ or driving simulator performance) as their outcome measure
driving ability, ten of ten with on-road driving performance, found that drivers with dementia performed worse than
four of four with driving simulator performance, and one of control subjects. However, when using state crash records,
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 373

the most objective and arguably the most relevant measure believe have medical conditions that might/will impact on
of driving risk, increased driving risk for patients with their ability to drive to their respective Ministries of Trans-
dementia was not consistently found. Collision rates as portation. Given that not all drivers with dementia (espe-
recorded on state driving records might be a relatively cially those with mild dementia) have higher crash rates
insensitive measure of driving risk and thus have limited than drivers without dementia, a diagnosis of dementia is
ability to detect differences in outcomes between drivers not sufficient in itself to lead to automatic revocation of the
with and without dementia. However, crash rates might be driver’s licenses. Rather, as recommended by a number of
the most important outcome measures because they are the organizations and groups [249,250], determination of the
events of most concern to society, placing individual drivers functional driving abilities at the individual level is the
at most risk. It is possible that as the driving skills of fairest and most appropriate method of assessing fitness to
patients with dementia deteriorate, patients, their families, drive in persons with mild dementia.
and health professionals undertake measures to restrict driv- Given that many causes of dementia are progressive in
ing exposure, thus reducing crash risk. No studies were nature, most persons with dementia will eventually need to
found examining the driving performance of persons with give up driving. To lessen the impact of transition to non-
mild cognitive impairment. driving, planning for this inevitability should take place as
soon as the diagnosis of dementia is made. This planning
7.2. Magnitude of driving risk could include the development of alternative transportation
It is difficult to precisely estimate the magnitude of options and participation in driver cessation support groups.
driving risk for persons with dementia. Of the two of five For those with a progressive dementia initially deemed safe
studies that found a positive association between crashes to drive, progressive deterioration in driving skills can be
recorded on the state driving record and dementia, persons expected. Studies show that persons with mild dementia
with dementia had a 2 to 2.5 times increased risk. However, who are initially deemed safe to drive are often found to be
factors such as severity of dementia and level of driving unsafe 6 to 12 months later [251,252].
exposure were not factored into these estimates. The latter is
important because persons with dementia drive fewer miles 7.5. Compensation methods
per year compared with age- and sex-matched controls. Potential methods of compensating for the decreased
When tested on road and in driving simulators, it is very driver performance and increased crash risk in drivers with
clear that persons with dementia are poorer drivers com- dementia include the following.
pared with those with normal cognition. This poorer perfor-
mance might not translate into consistently higher collision
7.5.1. Retraining/education programs
rates per miles driven because of self, family, or authority
No studies were found assessing the efficacy of
imposed driving restriction.
retraining/education programs on improving the driving
7.3. Cognitive functions and driving performance in persons with dementia. Because persons
with dementia have underlying progressive memory and
Numerous studies have examined the association of spe- cognitive deficits and often difficulties with insight and
cific neuropsychological tests and driving risk [247]. Brief judgment, attempts to upgrade their driving skills is not a
tests of general cognitive functioning such as the MMSE reasonable option.
[248] have shown an inconsistent relationship with driving
risk. Similar results were also found for studies specifically 7.5.2. Use of co-pilots
examining tests of attention and concentration, visuospatial No studies were found assessing the efficacy of having
skills, memory, executive functioning, and language. In other persons accompany drivers with dementia with re-
each of these domains, there were a few studies finding spect to reducing their driving risk. Because many crashes
positive associations between specific tests and driving risk, occur in a split second without time to give instructions to
but these results have not been confirmed in other studies. drivers, this method of compensation would seem to be
There is no single brief cognitive test that has sufficient ineffective.
validity, reliability, sensitivity, and/or specificity to be con-
sidered a robust tool in identifying older drivers with de- 7.5.3. Use of on-board navigation and crash
mentia who are cognitively unfit to drive or need further warning systems
testing. Further research is necessary to determine whether No studies were found that assessed the efficacy of these
a combination of brief tests can meet this goal. systems on improving the driving risk in persons with de-
mentia. Because the information processing of most persons
7.4. Assessment and follow-up
with dementia is impaired, these technologies are unlikely
In many provinces and territories, physicians and other to compensate for the driving deficiencies that drivers with
clinicians are legally mandated to report drivers whom they dementia demonstrate.
374 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

7.5.4. Restricted licensing characterized the negative consequences of dementia care-


Although some studies have shown that, in general, re- giving or caregiver burden. Depression and anxiety are of
stricted licenses reduce crash rates, no studies were found particular concern. All individuals caring for someone with
that specifically examined whether the granting of restricted a chronic illness have been found to have increased rates of
or conditional licensing to drivers with dementia reduces psychiatric morbidity, but those caring for someone with
their driving risk. Although many drivers with dementia dementia are even more at risk, with rates reportedly as high
perform adequately in routine predicable situations, they do as 50% [253].
not perform as well in situations that are less predictable, The primary theoretical model explaining the specific
precisely the time when many crashes occur. Therefore, it is effects of this burden is the stress/health model that explains
unlikely that the use of restricted/conditional licenses sig- how the stress of caregiving is translated into psychiatric
nificantly reduces crash risk in persons with dementia. and physical morbidity [254]. Factors associated with sig-
nificant caregiver burden and/or stress have been well-
7.6. Recommendation 25: Recommendations with regard described. These include factors related to the dementia
to driving a motor vehicle and individuals with a mild to patient and those related to the caregiver themselves. Prob-
moderate dementia lem behaviors of the dementia patient such as depression
and aggression are the most important predictors of care-
A. Clinicians should counsel persons with a progressive giver burden. Less predictive of burden is the amount of
dementia (and their families) that giving up driving will assistance with ADL provided to the patient by the care-
be an inevitable consequence of their disease. Strategies giver. Important caregiver factors that have been found to be
to ease this transition should occur early in the clinical associated with caregiver burden include female gender,
course of the disease (Grade B, Level II-2). low income, low life satisfaction, poor self-esteem and
B. No single brief cognitive test (eg, MMSE) or combi- self-assessed competence, and lack of social support. Lim-
nation of brief cognitive tests has sufficient sensitiv- ited research has been done to understand the positive as-
ity or specificity to be used as a sole determinant of pects of caregiving. These might be of particular importance
driving ability. Abnormalities on cognitive tests such because caregivers who could identify at least one positive
as the MMSE, clock drawing, and Trails B should aspect of caregiving are less likely to report burden or
result in further in-depth testing of driving ability depression [255]. A thorough understanding of the negative
(Grade B, Level III). and positive aspects of caregiving is important not only to
C. Driving is contraindicated in persons who, for cog- reduce caregiver burden but also to reduce potentially neg-
nitive reasons, have an inability to independently ative outcomes for individuals with dementia. For example,
perform multiple instrumental activities of daily liv- caregiving burden has been shown to be a risk factor for
ing (eg, medication management, banking, shopping, nursing home placement [256].
telephone use, cooking) or any of the basic activities Many intervention studies have been designed to reduce
of daily living (eg, toileting, dressing) (Grade B, caregiver burden and delay the negative outcomes of burden
Level III). such as nursing home placement. These interventions target
D. The driving ability of persons with earlier stages of patient and caregiver factors that have been associated with
dementia should be tested on an individual basis burden. These interventions include those designed to re-
(Grade B, Level III). duce the behavioral and psychological problems of the de-
E. A health professional– based comprehensive off-road mentia patient or the amount of assistance needed by the
and on-road driving evaluation is the fairest method demented patient. They also include strategies aimed pri-
of individual testing (Grade B, Level III). marily at the caregiver to increase their knowledge about the
F. In places where comprehensive off-road and on-road disease, provide additional resources and supports, reduce
driving evaluations are not available, clinicians must anxiety and depression, or alter caregiver behavior.
rely on their own judgment (Grade B, Level III). Interventions have been delivered in a variety of modal-
G. For persons deemed safe to drive, reassessment of ities either singly or in combination. Intervention formats
their ability to drive should take place every 6 to 12 include those that are group based, individually based, tech-
months or sooner if indicated (Grade B, Level III). nology based (telephone or computer), or service configu-
H. Compensatory strategies are not appropriate for those rations [257]. Broadly speaking, the interventions focus on
deemed unsafe to drive (Grade B, Level III). education, psychotherapy, or provision of services. Educa-
tion can include providing information about dementia or
8. Support of caregivers skills training to change the caregivers’ interaction with the
dementia patient. Psychotherapy can include psychological
In Canada about half of the individuals with dementia support for the caregiver, assistance to develop their social
reside in the community, and more than 90% are cared for support network, and self-care strategies. Services might
by family and friends [193,195]. Previous research has include access to respite care, day care, or a case manager.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 375

The most clinically relevant effects of these interventions American Academy of Neurology (AAN) published guide-
have been reductions in rates of caregiver depression and lines in 2001 that included two recommendations of rele-
delays in nursing home placement, although some might vance for family caregivers [261]. One recommendation
argue that prolonging community care might not always be was that short-term programs directed toward educating
a desirable outcome. Other positive effects include care- caregivers about AD be offered to improve caregiver satis-
giver satisfaction with services, improved self-appraisal of faction. In addition, the AAN recommended that intensive
caregiver coping skills and knowledge, and improved rela- long-term education and support services (when available)
tionships with the care recipients. Common features of be offered to caregivers to delay time to nursing home
successful interventions include (1) a continuing relation- placement. Listed as practice options (evidence supported
ship between the caregiver and helper over time, (2) a by expert opinion, case series, and studies with historical
variety of flexible interventions offered that can meet the controls only) were use of computer networks to provide
varied needs of caregivers, (3) psycho-education or skills education and support to caregivers, telephone support pro-
training that teaches caregivers to change their interactions grams, and respite services including adult day care.
with patients with dementia, and (4) involvement of the The American Association of Geriatric Psychiatry issued
caregiver and patient in the intervention. Interventions that a statement on family caregivers of dementia patients in
appear unsuccessful include short educational programs that 2001 [262]. It emphasized the importance of caregivers in
only enhance knowledge about dementia, support groups the treatment of patients with dementia and recommended
alone, and service configurations such as case management that caregiver counseling be a reimbursable covered service.
or brief interventions that do not include long-term contact The American Medical Association has also encouraged a
with the caregiver [161,254,256 –258]. A Cochrane review physician/caregiver/patient partnership in dementia care.
on respite care for people with dementia and their caregivers They have advocated that physicians monitor caregiver
found no evidence of either benefit or adverse effects from functioning and have developed Web-based materials and
the use of respite services. The authors believed their results information to assist physicians in addressing caregiver
should be treated with caution because they might reflect the needs [263]. A caregiver self-assessment tool is available to
dearth of high quality studies rather than any true lack of assist in identifying caregivers at risk of adverse health
benefit [259]. outcomes. Caregivers who report high levels of distress in
There is often a major discrepancy between the elaborate completing this questionnaire are encouraged to get a
programs that the literature tells us are effective and what is check-up from their physician, obtain respite from caregiv-
locally available. Although these latter services might not ing, and consider joining a support group. Physicians caring
decrease caregiver burden, they can improve caregiver sat- for these caregivers are encouraged to discuss the need for
isfaction. Long-term involvement with these programs counseling and other interventions and to refer them to
tends to be more beneficial. available community services.
The studies examined were often limited by small sam- Financial hardships can be encountered in caring for a
ple sizes, short follow-up times, and the many caregiver and person with dementia at home. Government plans might
patient outcomes that were studied. The heterogeneity of the provide a degree of financial relief. For example, in Canada
caregiver experience is clearly a factor limiting the effec- most patients with a dementia would be eligible for a dis-
tiveness of these interventions or the generalizability of ability tax credit. Physicians and other qualified practitio-
many studies. Targeting caregivers who meet specific cri- ners can alert caregivers of this potential tax benefit and can
teria such as scoring above a certain level for measures of help them in making an application by filling out Part B of
depression might improve outcomes in the future. Schultz the Disability Tax Credit Certificate (Form T2201, available
and Martire [254] recommended the assessment of care- at www.cra.gc.ca/forms). For details of this program please
giver risk in five domains. Interventions can then be targeted refer to Information Concerning People With Disabilities
to those areas identified as problematic for individual care- (guide RC4064) or visit the Canada Revenue Agency Web
givers. The risk areas are safety, self-care and preventative site (www.cra.gc.ca/disability).
health behaviors, caregiver support (informational, instru-
mental, and emotional), depression and distress, and prob- 8.1. Recommendation 26: Recommendations with regard
lem behaviors of care recipients. to caregivers
A number of the RCTs of drug therapy for mild to
moderate AD have included caregiver burden as a second-
ary outcome measure. A recent systematic review of the A. The clinician should acknowledge the important role
effect of cholinesterase inhibitors on burden and active time played by the caregiver in dementia care. The clini-
use of caregivers of persons with AD concluded that they cian should work with caregivers and families on an
had a small beneficial effect on both [260]. ongoing basis and schedule regular appointments for
There have been a few guidelines and position state- patients and caregivers together and alone (Grade B,
ments on dementia care published since the CCCD. The Level III).
376 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

B. The clinician should inquire about caregiver informa- of this occurring does appear to be modifiable [269 –271].
tion and support needs, provide education to patients Guideline adherence is poor without specific interventions.
and families about dementia, and assist in recruiting Common obstacles identified by Canadian physicians
other family members and formal community ser- to meeting the health care needs of their community
vices to share the caregiving role. If available, refer include inadequate time, inadequate remuneration, and
patients to specialized dementia services (eg, Alzhei- lack of accessible community resources [272]. We were
mer Society, community-based dementia programs, struck by the difference between what the literature tells
memory clinics) that offer comprehensive treatment us works and what is available in our communities. The
programs including caregiver support, education, and availability of required community-based resources will be
training (Grade A, Level 1). critical in improving the care provided to those with demen-
C. The clinician should inquire about caregiver health tia and their families. There is some evidence that using
(both physical and psychiatric), offer treatment for
in-home help services earlier might delay institutionaliza-
these problems (including individual psychotherapy
tion [273].
or medications as indicated), and refer to appropriate
The favored model for chronic disease management is
specialists (Grade B, Level III).
the delivery of services by multidisciplinary teams who
D. The clinician should enquire about problem behav-
collaboratively educate, counsel, and empower patients with
iors of the dementia patient and the effect these
behaviours are having on the caregiver. If these are self-care techniques to manage their chronic diseases. Sup-
causing significant caregiver distress, refer the care- ported by customized treatment plans and the multidisci-
giver and patient to specialized dementia services plinary team, patients are charged with undertaking neces-
that can offer treatment to the patient and assist the sary lifestyle and behavioral changes to manage their
caregiver in modifying their interactions with the condition responsibly. Chronic disease management is de-
patient (Grade A, Level 1). pendent on the promotion of patient self-management and
E. Pharmacotherapy for AD can decrease caregiver bur- clinician adherence to evidence-based guidelines [274].
den and the time required of caregivers to support the This approach would require modification if used for de-
care recipient. It should be considered as a means to mentia. There would have to be reduced reliance on patient
help support caregivers (Grade B, Level I). self-care and a concomitant increased effort on caregiver
F. Future studies of medications for the treatment of AD support and education. A chronic disease management ap-
and dementia should examine the impact of these proach, however, could address the need for comprehen-
agents on caregiver burden and the time required to sive, evidence-based management of the person with de-
support the care recipient. There is a need to ensure mentia during the course of their illness. This could permit
consistency in the measurement of these outcomes the efficient use of the entire continuum of resources in-
(Grade B, Level III). cluding community-based and facility-based continuing
care [275]. Studies suggest that a more systematic approach
9. Training needs and system issues to the management of dementia can improve satisfaction
levels and enhance adherence to guidelines [276]. Whether
To implement the recommendations for the management of this approach for dementia improves patient outcomes,
the mild and moderate stages of AD, we need to ensure that however, remains unknown at the present time.
clinicians are adequately trained, needed resources and ser-
Shared care models are being explored as a way to deal
vices are available, and that we explore different models of
with chronic medical conditions. It can be defined as shared
care. Please note that the literature referenced below originates
responsibility, enhanced information exchange, continuing
almost entirely outside Canada, and caution must be exercised
medical education, and explicit clinical guidelines between
in extrapolating the findings reported to our country.
Dementia presents unique challenges to the clinician specialty services and primary care practitioners [277]. Al-
[264]. Many health care providers remain uninformed about though it is being explored as a way to provide dementia
AD [265,266]. Specific educational needs among primary care, obstacles to effective collaboration include therapeutic
care physicians would include knowledge about local diag- nihilism, risk reduction or avoidance, concerns about com-
nostic and support services, development of assessment and petency, and limited access to required resources [278].
communication skills, management of behavioral problems, Another barrier to shared care models for dementia is that a
and the coordination of support services [267]. A particular number of primary care physicians believe that dementia
attitudinal barrier is the skepticism of many primary care care should be dealt with by specialists and not themselves
practitioners about the effectiveness of interventions for [267]. Reimbursement issues are another barrier to the de-
AD [268]. livery of high quality dementia care by physicians [279].
Studies on the uptake of dementia guidelines by physi- The complex, time-consuming aspects of dementia care are
cians have generally been disappointing, but the likelihood not adequately reimbursed by fee-for-service payments.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 377

9.1. Recommendation 27: Recommendations with regards Acknowledgments


to education
The authors gratefully acknowledge numerous sources of
support and funding.
A. All clinicians caring for patients with mild to moderate
David Hogan is supported by the Brenda Strafford Foun-
AD have to acquire the core knowledge and skills
dation Chair in Geriatric Medicine, University of Calgary.
required to manage this condition (Grade B, Level III).
Dorothy Forbes receives salary support as a CIHR New
B. A multifaceted educational program should be imple-
Investigator.
mented to promote adoption of the recommendations of
Krista Lanctôt was supported by an Ontario Mental
the 3rd CCCDTD by practitioners (Grade B, Level I).
Health Foundation New Investigator Fellowship.
9.2. Recommendation 28: Recommendations with regards Debra Morgan receives support from a CIHR New
to the organization and funding of care for those Emerging Team grant Strategies to Improve the Care of
with a dementia Persons with Dementia in Rural and Remote Areas funded
by CIHR (Institute of Aging, Institute of Health Services
A. Every community should examine the services locally and Policy Research, Rural and Northern Health Initiative)
available for the management of those with a dementia, and partners (Saskatchewan Health Research Foundation,
assess their adequacy, and implement plans to deal with Alzheimer Society of Saskatchewan, and University of
identified deficiencies (Grade C, Level III). Saskatchewan).
B. There is a need to modify the prevailing model of
chronic disease management (ie, less reliance on pro-
motion of patient self-management coupled with References
greater caregiver involvement) for dementia. The ef-
[1] Patterson CJS, Gauthier S, Bergman H, Cohen CA, Feightner JW,
ficacy and efficiency of modified chronic disease
Feldman H, et al. The recognition, assessment and management of
management for dementia should be explored (Grade dementing disorders: conclusions from the Canadian Consensus
C, Level III). Conference on Dementia. CMAJ 1999;160(Suppl):S1–15.
C. Shared care models for the management of patients [2] Canadian Task Force on Preventive Health Care. New grades for
with mild to moderate AD and dementia should be recommendations from the Canadian Task Force on Preventive
Health Care. CMAJ 2003;169:207– 8.
developed and evaluated. This will require the accep-
[3] Elie M, Cole MG, Primeau FJ, Bellavance F. Delirium risk factors
tance of joint responsibility on the part of primary in elderly hospitalized patients. J Gen Intern Med 1998;13:204 –12.
care practitioners and specialty services in delivering [4] Inouye SK, Bogardus ST, Charpentier PA, Leo-Summers L, Acam-
care to patients with dementia (Grade C, Level III). pora D, Holford TR, et al. A multicomponent intervention to prevent
D. Dementia care must be adequately funded and reim- delirium in hospitalized patients. N Engl J Med 1999;340:669 –76.
[5] Britton A, Russell R. Multidisciplinary team interventions for de-
bursed. Inadequate remuneration should not be a bar-
lirium in patients with chronic cognitive impairment. Cochrane
rier to the delivery of good dementia care (Grade C, Database Syst Rev 2004:CD000395.
Level III). [6] Hill JW, Futterman R, Duttagupta S, Mastey V, Lloyd JR, Fillit H.
Alzheimer’s disease and related dementias increase costs of co-
morbidities in managed Medicare. Neurology 2002;58:62–70.
Author Disclosures [7] Schubert CC, Boustani M, Callahan CM, Perkins AJ, Carney CP,
Fox C, et al. Comorbidity profile of dementia patients in primary
David Hogan has been a site principle investigator in care: are they sicker? J Am Geriatr Soc 2006;54:104 –9.
studies supported by Janssen Ortho, Neurochem, Novartis [8] Winkelmayer WC, Fischer MA, Schneeweiss S, Wang PS, Levin R,
and Pfizer Canada within the last five years and given Avorn J. Underuse of ACE inhibitors and angiotensin II receptor
blockers in elderly patients with diabetes. Am J Kidney Dis 2005;
presentations sponsored by Janssen Ortho, Merck Frosst,
46:1080 –7.
Novartis and Pfizer within the last five years. [9] Biessels GJ, van der Heide LP, Kamal A, Bleys RLAW, Gispen
Peter Bailey has had support from pharmaceutical firms WH. Ageing and diabetes: implications for brain function. Eur
(as a Speaker) and the C5R (President). J Pharmacol 2002;441:1–14.
Krista Lanctôt has had support from Pfizer Canada (Con- [10] Kalmijn S, Feskens EJM, Launer LJ, Stijnen T, Kromhout D. Glu-
cose intolerance, hyperinsulinaemia and cognitive function in a
sultant, Speaker, Research Support), Abbott Laboratories
general population of elderly men. Diabetologia 1995;38:1096 –102.
(consultant, research support), Janssen Ortho Inc. (Consul- [11] Cook DG, Leverenz JB, McMillan PJ, Kulstad JJ, Ericksen S, Roth
tant, Research Support), and Lundbeck Canada (Research RA, et al. Reduced hippocampal insulin-degrading enzyme in late-
Support). onset Alzheimer’s disease is associated with the apolipoprotein
Lilian Thorpe has received support for research, advisory E-epsilon4 allele. Am J Pathol 2003;162:313–9.
[12] Messier C, Gagnon M. Glucose regulation and cognitive functions:
boards and/or presentations from the following: Astra Zen-
relation to Alzheimer’s disease and diabetes. Behav Brain Res 1996;
eca, Bristol-Meyers Squibb, Eli Lilly, Glaxo SmithKline, 75:1–11.
Janssen Ortho, Lundbeck, Novartis, Organon, Pfizer and [13] Martin DC, Miller JK, Kapoor W, Arena VC, Boller F. A controlled
Wyeth. study of survival with dementia. Arch Neurol 1987;44:1122– 6.
378 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

[14] Jagger C, Clarke M, Stone A. Predictors of survival with Alzhei- [33] Clare L, Roth I, Wilson B, Carter G, Hodges J. Relearning face-
mer’s disease: a community-based study. Psychol Med 1995;25: name associations in early Alzheimer’s disease. Neuropsychology
171–7. 2002;16:538 – 47.
[15] Agüero-Torres H, Fratglioni L, Guo Z, Viitanen M, Winblad B. [34] Farina E, Fioravanti R, Chiavari L, Imbornone E, Alberoni M,
Prognostic factors in very old demented adults: a seven-year Pomati G, et al. Comparing two programs of cognitive training in
follow-up from a population-based survey in Stockholm. J Am Alzheimer’s diseases: a pilot study. Acta Neurologica Scandinavica
Geriatr Soc 1998;46:444 –52. 2002;105:365–71.
[16] Pereles L, Romonko L, Murzyn T, Hogan D, Silvius J, Stokes E, [35] Clare L, Wilson B, Carter G, Hodges J. Cognitive rehabilitation as
et al. Evaluation of a self-medication program. J Am Geriatr Soc a component of early intervention in Alzheimer’s disease: a single
1996;44:161–5. case study. Aging Ment Health 2003;7:15–21.
[17] Biem HJ, Turnell RW, D’Arcy C. Computer telephony: automated [36] Gitlin L, Corcoran M, Winter L, Boyce A, Hauck W. A randomized,
calls for medical care. Clin Invest Med 2003;26:259 – 68. controlled trial of a home environmental intervention: effect of
[18] Roe CM, Anderson MJ, Spivack B. Use of anticholinergic medica- efficacy and upset in caregivers and on daily function of persons
tions by older adults with dementia. J Am Geriatr Soc 2002;50: with dementia. Gerontologist 2001;41:4 –14.
836 – 42. [37] Gitlin LN, Hauck WW, Dennis MP, Winter L. Maintenance of
[19] Sunderland T, Tariot PN, Newhouse PA. Differential responsivity of effects of the home environment skill-building program for family
mood, behavior, and cognition to cholinergic agents in elderly neu- caregivers and individuals with Alzheimer disease and related dis-
ropsychiatric populations. Brain Res 1988;472:371– 89. orders. J Gerontol Series A Biol Sci Med Sci 2005;60:368 –74.
[20] Thienhaus OJ, Allen A, Bennett JA, et al. Anticholinergic serum [38] Day K, Carreon D, Stump C. The therapeutic design of environ-
levels and cognitive performance. Eur Arch Psychiatry Clin Neuro- ments for people with dementia: a review of the empirical research.
sci 1990;240:28 –33. Gerontologist 2000;40:397– 413.
[21] Kitano S, Morimoto S, Fukuo, et al. Circulating suppressing factor [39] Steyltjens EMJ, Dekker J, Bouter LM, Jellma S, Bakker EB, van den
for the muscarinic acetylcholine receptor in patients with senile Ende CHM. Occupational therapy for community dwelling elderly
dementia of the Alzheimer type. Gerontology 1992;39(Suppl 1): people: as systematic review. Age Aging 2004;33:453– 60.
24 – 8. [40] Teri L, Gibbons LE, McCurry SM, Logsdon RG, Buchner DM,
[22] Mulsant BH, Gharabawi GM, Bossie CA, Mao L, Martinez RA,
Barlow WE, et al. Exercise plus behavioural management in patients
Tune LA, et al. Correlates of anticholinergic activity in patients with
with Alzheimer disease: a randomized controlled trial. JAMA 2003;
dementia and psychosis treated with risperidone or olanzapine.
290:2015–22.
J Clin Psychiatry 2004;65:1708 –14.
[41] Patterson JT, Wessel J. Strategies for retraining functional move-
[23] Chew ML, Mulsant BH, Pollock BG. Serum anticholinergic activity
ment in persons with Alzheimer disease: a review. Physiotherapy
and cognition in patients with moderate-to-severe dementia. Am J
Canada 2002;54:274 – 80.
Geriatr Psychiatry 2005;13:535– 8.
[42] Heyn P, Abreu BC, Ottenbacher JJ. The effects of exercise training
[24] Ancelin ML, Artero S, Porter F, Dupuy A-M, Touchon J, Ritchie K.
on elderly persons with cognitive impairment and dementia: a meta-
Non-degenerative mild cognitive impairment in elderly people and
analysis. Arch Phys Med Rehabil 2004;85:1694 –704.
use of anticholinergic drugs: longitudinal cohort study. BMJ 2006;
[43] Dooley NR, Hinojosa J. Improving quality of life for persons with
332:455–9.
Alzheimer disease and their family caregivers: brief occupational
[25] Carnahan RM, Lund BC, Perry PJ, Chrischilles EA. The concurrent
therapy intervention. Am J Occup Ther 2004;58:561–9.
use of anticholinergics and cholinesterase inhibitors: rare event or
[44] Scherder E, Bouma A, Steen AM. Effects of isolated transcutaneous
common practice? J Am Geriatr Soc 2004;52:2082–7.
[26] Lu CJ, Tune LE. Chronic exposure to anticholinergic medications electrical nerve stimulation on memory and affective behaviour in
adversely affects the course of Alzheimer disease. Am J Geriatr patients with probable Alzheimer’s disease. Biol Psychiatry 1998;
Psychiatry 2003;11:458 – 61. 43:417–24.
[27] Jewart RD, Green J, Lu CJ, Cellar J, Tune LE. Cognitive, behav- [45] Cameron M, Lonergan E, Lee H. Transcutaneous electrical nerve
ioral, and physiological changes in Alzheimer disease patients as a stimulation (TENS) for dementia. Cochrane Database Syst Rev
function of incontinence medications. Am J Geriatr Psychiatry 2005; 2003:CD004032.
13:324 – 8. [46] Bates J, Boote J, Beverley C. Psychosocial interventions for people
[28] Cahn-Weiner DA, Malloy PF, Rebok GW, Ott BR. Results of a with milder dementing illness: a systematic review. J Adv Nurs
randomized placebo-controlled study of memory training for mildly 2004;45:644 –58.
impaired Alzheimer’s disease patients. Applied Neuropsychology [47] Birks J. Cholinesterase inhibitors for Alzheimer’s disease. Cochrane
2003;10:215–23. Database Syst Rev 2006:CD005593.
[29] Lowenstein DA, Acevedo A, Czaja SJ, Duara R. Cognitive rehabil- [48] Birks J, Grimley Evans J, Iakovidou V, Tsolaki M. Rivastigmine for
itation of mildly impaired Alzheimer Disease patients on cholines- Alzheimer’s disease. Cochrane Database Syst Rev 2000:CD001191.
terase inhibitors. Am J Geriatr Psychiatry 2004;12:395– 402. [49] Birks J, Harvey R. Donepezil for dementia due to Alzheimer’s
[30] Bottino CM, Carvalho IA, Alvarez AM, Avila R, Zukauskas PR, disease. Cochrane Database Syst Rev 2006:CD001190.
Bustamante SE, et al. Cognitive rehabilitation combined with drug [50] Loy C, Schneider L. Galantamine for Alzheimer’s disease and
treatment in Alzheimer’s disease patients: a pilot study. Clin Rehabil mild cognitive impairment. Cochrane Database Syst Rev 2006:
2005;19:861–9. CD001747.
[31] Clare L, Woods RT, Moniz Cook ED, Orrell M, Spector A. Cog- [51] Lanctôt KL, Herrmann N, Yau KK, Khan LR, Liu BA, LouLou
nitive rehabilitation and cognitive training for early-stage Alzhei- MM, et al. Efficacy and safety of cholinesterase inhibitors in Alz-
mer’s disease and vascular dementia. Cochrane Database Syst Rev heimer’s disease: a meta-analysis. CMAJ 2003;169:557– 64.
2003:CD003260. [52] Ritchie CW, Ames D, Clayton T, Lai R. Meta-analysis of random-
[32] DeVreese LP, Neri M, Fiorvante M, Belloi L, Zanetti O. Memory ized trials of the efficacy and safety of donepezil, galantamine, and
rehabilitation in Alzheimer’s disease: a review of progress. Int J rivastigmine for the treatment of Alzheimer disease. Am J Geriatr
Geriatr Psychiatry 2001;16:794 – 809. Psychiatry 2004;12:358 – 69.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 379

[53] Rockwood K. Size of the treatment effect on cognition of cholines- [72] Dantoine T, Auriacombe S, Sarazin M, Becker H, Pere J-J,
terase inhibition in Alzheimer’s disease. J Neurol Neurosurg Psy- Bourdeix I. Rivastigmine monotherapy and combination therapy
chiatry 2004;75:677– 85. with memantine in patients with moderately severe Alzheimer’s
[54] Clegg A, Bryant J, Nicholson T, McIntyre L, De Broe S, Gerard K, disease who failed to benefit from previous cholinesterase inhib-
et al. Clinical and cost-effectiveness of donepezil, rivastigmine, and itor treatment. Int J Clin Pract 2006;60:110 – 8.
galantamine for Alzheimer’s disease: a systematic review. Int J [73] Waldemar G, Kørner A, Hyvärinen M, Wetterberg P, Lehto H,
Technol Assess Health Care 2002;18:497–507. Josiassen MK. Tolerability of switching from donepezil to meman-
[55] Wolfson C, Oremus M, Shukla V, Momoli F, Demers L, Perrault A, tine treatment in patients with moderate to severe Alzheimer’s
et al. Donepezil and rivastigmine in the treatment of Alzheimer’s disease. Available at: http://www.lundbeck.com/investor/Pipeline/
disease: a best-evidence synthesis of the published data on their ClinicalData/TOLERABILITY%20OF%20SWITCHING%20FROM%
efficacy and cost-effectiveness. Clin Ther 2002;24:862– 86. 20DONEPEZIL%20TO%20MEMANTINE%20TREATMENT%20
[56] Thompson S, Lanctôt KL, Herrmann N. The benefits and risks IN%20PATIENTS%20WITH%20MODERATE%20TO%20SEVERE
associated with cholinesterase inhibitor therapy in Alzheimer’s dis- %20ALZHEIMER02B9S%20DISEASE%20Dajs%20%
ease. Expert Opin Drug Saf 2004;3:425– 40. 20%20ICMPE%202005.pdf. Accessed July 2, 2007.
[57] Kaduszkiewicz H, Zimmermann T, Beck-Bornholdt HP, van den [74] Areosa Satre A, Sherriff F, McShane R. Memantine for dementia.
Bussche H. Cholinesterase inhibitors for patients with Alzheimer’s Cochrane Database Syst Rev 2005:CD003154.
disease: systematic review of randomised clinical trials. BMJ 2005; [75] Alzheimer’s Association fact sheet on “Memantine (Namenda)”.
331:321–7. Updated January 10, 2006.
[58] Hogan DB, Goldlist B, Naglie G, Patterson C. Comparison studies [76] Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S,
of cholinesterase inhibitors for Alzheimer’s disease. Lancet Neurol Gergel I, et al. Memantine treatment in patients with moderate to
2004;3:622– 6. severe Alzheimer disease already receiving donepezil: a randomized
[59] Bullock R, Touchon J, Bergman H, Gambina G, He Y, Rapatz G, controlled trial. JAMA 2004;291:317 Accessed 2 July, 2007.24.
et al. Rivastigmine and donepezil treatment in moderate to moder- [77] Doody RS, Geldmacher DS, Gordon B, Perdomo CA, Pratt RD.
ately-severe Alzheimer’s disease over a 2-year period. Curr Med Open-label, multicenter, phase 3 extension study of the safety and
Res Opin 2005;21:1317–27. efficacy of donepezil in patients with Alzheimer’s disease. Arch
[60] Hogan DB, Patterson C. Progress in clinical neurosciences: treat-
Neurol 2001;58:427–3.
ment of Alzheimer’s disease and other dementias—review and com-
[78] Markesbery WR. Oxidative stress hypothesis in Alzheimer’s dis-
parison of the cholinesterase inhibitors. Can J Neurol Sci 2002;29:
ease. Free Radic Biol Med 1997;23:134 – 47.
306 –14.
[79] Luchsinger JA, Mayeux R. Dietary factors and Alzheimer’s disease.
[61] Gillette-Guyonnet S, Cortes F, Cantet C, Vellas B. Long-term cho-
Lancet Neurol 2004;3:579 – 87.
linergic treatment is not associated with greater risk of weight loss
[80] Butterfield DA, Castegna A, Drake J, Scapagnini G, Calabrese V.
during Alzheimer’s disease: data from the French REAL.FR cohort.
Vitamin E and neurodegenerative disorders associated with oxida-
J Nutr Health Aging 2005;9:69 –73.
tive stress. Nutr Neurosci 2002;5:229 –39.
[62] Bordier P, Lanusse S, Garrigue S, Reynard C, Robert F, Gencel L,
[81] Masaki KH, Losonczy KG, Izmirlian G, Foley DJ, Ross GW, Petro-
et al. Causes of syncope in patients with Alzheimer’s disease treated
vitch H, et al. Association of vitamin E and C supplement use with
with donepezil. Drugs Aging 2005;22:687–94.
cognitive function and dementia in elderly men. Neurology 2000;
[63] Bordier P, Garrigue S, Barold SS, Bressolles N, Lanusse S, Clement
54:1265–72.
J. Significance of syncope in patients with Alzheimer’s disease
[82] Engelhart MJ, Geerlings MI, Ruitenberg A, Van Swieten JC,
treated with cholinesterase inhibitors. Europace 2003;5:429 –31.
[64] Gill SS, Mamdani M, Naglie G, Streiner DL, Bronskill SE, Kopp A, Hofman A, Witteman JCM, et al. Dietary intake of antioxidants and
et al. A prescribing cascade involving cholinesterase inhibitors and risk of Alzheimer disease. JAMA 2002;287:3223–9.
anticholinergic drugs. Arch Intern Med 2005;165:808 –13. [83] Grodstein F, Chen J, Willett WC. High-dose antioxidant supplements
[65] Stahl SM, Markowitz JS, Gutterman EM, Papadopoulos G. Co-use and cognitive function in community-dwelling elderly women. Am J
of donepezil and hypnotics among Alzheimer’s disease patients Clin Nutr 2003;77:975– 84.
living in the community. J Clin Psychiatry 2003;64:466 –72. [84] Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grund-
[66] Auriacombe S, Pere JJ, Loria-Kanza Y, Vellas B. Efficacy and man M, et al. A controlled trial of selegiline, alpha-tocopherol, or
safety of rivastigmine in patients with Alzheimer’s disease who both as treatment for Alzheimer’s disease. N Engl J Med 1997;336:
failed to benefit from treatment with donepezil. Curr Med Res Opin 1216 –22.
2002;18:129 –38. [85] Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R,
[67] Emre M: Switching cholinesterase inhibitors in patients with Alz- Ferris S, et al. Vitamin E and donepezil for the treatment of mild
heimer’s disease. Int J Clin Pract Suppl 2002;127:64 –72. cognitive impairment. N Engl J Med 2005;352:2379 – 88.
[68] Gauthier S, Emre M, Farlow MR, Bullock R, Grossberg GT, Potkin [86] Tabet N, Birks J, Grimley Evans J, Orrel M, Spector A. Vitamin E
SG. Strategies for continued successful treatment of Alzheimer’s for Alzheimer’s disease. Cochrane Database Syst Rev 2000:
disease: switching cholinesterase inhibitors. Curr Med Res Opin CD002854.
2003;19:707–14. [87] Miller ER, Pastor-Barriuso R, Dalai D, Riemersma RA, Appel LJ,
[69] Alwaahhabi FK. Successfully switching acetylcholinesterase inhib- Guallar E. Meta-analysis: high-dosage vitamin E supplementation
itor therapy in probable Lewy body dementia. J Psychopharmacol may increase all-cause mortality. Ann Intern Med 2005;142:37– 46.
2005;19:214 – 6. [88] Thal LJ, Grundman M, Berg J, Ernstrom K, Margolin R, Pfeiffer E,
[70] Sadowsky CH, Farlow MR, Atkinson L, Steadman J, Koumaras B, et al. Idebenone treatment fails to slow cognitive decline in Alzhei-
Chen M, et al. Switching from donepezil to rivastigmine is well mer’s disease. Neurology 2003;61:1498 –502.
tolerated: results of an open-label safety and tolerability study. Prim [89] Leboeuf R. Homocysteine and Alzheimer’s disease. J Am Diet
Care Companion J Clin Psychiatry 2005;7:43– 8. Assoc 2003;103:304 –7.
[71] Wilkinson DG, Howe I. Switching from donepezil to galantamine: a [90] Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH,
double-blind study of two wash-out periods. Int J Geriatr Psychiatry d’Agostino RB, et al. Plasma homocysteine as a risk factor for
2005;20:489 –91. dementia and Alzheimer’s disease. N Engl J Med 2002;346:476 – 83.
380 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

[91] Kalmijn S, Launer LJ, Lindemans J, Bots ML, Hofman A, Breteler [113] Firuzi O, Praticò D. Coxibs and Alzheimer’s disease: should they
MMB. Total homocysteine and cognitive decline in a community- stay or should they go? Ann Neurol 2006;59:219 –28.
based sample of elderly subjects: the Rotterdam study. Am J Epi- [114] Tabet N, Feldman H. Ibuprofen for Alzheimer’s disease. Cochrane
demiol 1999;50:283–9. Database Syst Rev 2003.CD004031.
[92] Luchsinger JA, Tang MX, Shea S, Miller J, Green R, Mayeux R. [115] Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Thomas RG,
Plasma homocysteine levels and risk of Alzheimer disease. Neurol- et al. A randomized controlled trial of prednisone in Alzheimer’s
ogy 2004;62:1972– 6. disease. Neurology 2000;54:588 –93.
[93] Wang HX, Wahlin A, Basun H, Fastbom J, Winblad B, Fratiglioni [116] van Gool WA, Weinstein HC, Scheltens PK, Walstra GJ. Effect of
L. Vitamin B12 and folate in relation to the development of Alz- hydroxychloroquine on progression of dementia in early Alzhei-
heimer’s disease. Neurology 1991;56:1188 –94. mer’s disease: an 18-month randomised, double-blind, placebo-
[94] Crystal HA, Ortof E, Frishman WH, Gruber A, Hershman D, Aron- controlled study. Lancet 2001;358:455– 60.
son M. Serum vitamin B12 levels and incidence of dementia in a [117] Heart Protection Study Collaborative Group. MRC/BHF Heart Pro-
healthy elderly population: a report from the Bronx longitudinal tection Study of cholesterol lowering with simvastatin in 20,536
aging study. J Am Geriatr Soc 1994;42:933– 6. high-risk individuals: a randomized placebo-controlled trial. Lancet
[95] Nilsson K, Gustafson L, Hultberg B. Improvement of cognitive 2002;360:7–12.
functions after cobalamin/folate supplementation in elderly patients [118] Shepherd J, Blauw GJ, Murphy MB, Bollen EL, Buckley BM,
with dementia and elevated plasma homocysteine. Int J Geriatr Cobbe SM, et al. Pravastatin in elderly individuals at risk of vascular
Psychiatry 2001;16:609 –14. disease (PROSPER): a randomised controlled trial. Lancet 2002;
[96] Kwok T, Tang C, Woo J, Lai WK, Law LK, Pang CP. Randomized 360:1623–30.
trial of the effect of supplementation on the cognitive function of [119] Sparks DL, Sabbagh MN, Connor DJ, Lopez J, Launer LJ, Browne
older people with subnormal cobalamin levels. Int J Geriatr Psychi- P, et al. Atorvastatin for the treatment of mild to moderate Alzhei-
atry 1998;13:611– 6. mer disease: preliminary results. Arch Neurol 2005;62:753–7.
[97] Rodriguez-Martin JL, Qizilbash N, Lopez-Arrieta JM. Thiamine for [120] Hogervorst E, Yaffe K, Richards M, Huppert F. Hormone replace-
Alzheimer’s disease. Cochrane Database Syst Rev 2001.CD001498. ment therapy to maintain cognitive function in women with demen-
[98] Malouf R, Grimley Evans J. Vitamin B6 for cognition. Cochrane tia. Cochrane Database Syst Rev 2002.CD003799.
Database Syst Rev 2003.CD004393. [121] Henderson VW, Hogervorst E. Testosterone and Alzheimer disease:
[99] Malouf R, Areosa Sastre A. Vitamin B12 for cognition. Cochrane is it men’s turn now? Neurology 2004;62:170 –1.
Database Syst Rev 2003.CD004326. [122] Almeida OP, Flicker L. Testosterone and dementia: too much ado
[100] Malouf R, Grimley Evans J, Aerosa Sastre A. Folic acid with or about too little data. J Br Menopause Soc 2003;9:107–10.
without vitamin B12 for cognition and dementia. Cochrane Database [123] Ramsden M, Shin TM, Pike CJ. Androgens modulate neuronal
Syst Rev 2003.CD004514. vulnerability to kainite lesion. Neuroscience 2003;122:573– 8.
[101] Watkins RW. Herbal therapeutics: the top 12 selling remedies. [124] Moffat SD, Zonderman AB, Metter EJ, Kawas C, Blackman MR,
Emerg Med 2002;34:12–9. Harman SM, Resnick SM. Free testosterone and risk for Alzheimer
[102] Kurz A, Van Baelen B. Ginkgo biloba compared with vholinesterase disease in older men. Neurology 2004;62:188 –93.
inhibitors in the treatment of dementia: a review based on meta- [125] Tan RS, Pu SJ. A pilot study on the effects of testosterone in
analyses by the Cochrane Collaboration. Dement Geriatr Cogn Dis- hypogonadal aging male patients with Alzheimer’s disease. Aging
ord 2006;18:217–25. Male 2003;6:13–7.
[103] Birks J, Gimley Evans J. Ginkgo biloba for cognitive impairment [126] Cherrier MM, Matsumoto AM, Amory JK, Asthana S, Bremner W,
and dementia. Cochrane Database Syst Rev 2002.CD003120. Peskind ER, et al. Testosterone improves spatial memory in men
[104] Bent S, Goldberg H, Padula A, Avins AL. Spontaneous bleeding with Alzheimer disease and mild cognitive impairment. Neurology
associated with ginkgo biloba: a case report and systematic review 2005;64:2063– 8.
of the literature: a case report and systematic review of the literature. [127] Lyketsos CG, Lopez O, Jones B, Fitzpatrick AL, Breitner J,
J Gen Intern Med 2005;20:657– 61. DeKosky S. Prevalence of neuropsychiatric symptoms in dementia
[105] Vandenabeele P, Fiers W. Is amyloidogenesis during Alzheimer’s and mild cognitive impairment: results from the cardiovascular
disease due to an IL-1/IL-6 mediated acute phase response in the health study. JAMA 2002;288:1475– 83.
brain? Immunology Today 1991;12:217–9. [128] Mega MS, Cummings JL, Fiorello T, Gornbein J. The spectrum of
[106] Aisen PS, Davis KL. Inflammatory mechanisms in Alzheimer’s behavioral changes in Alzheimer’s disease. Neurology 1996;46:
disease: implications for therapy. Am J Psychiatry 1994;151: 130 –5.
1105–13. [129] Lopez OL, Becker JT, Sweet RA, Klunk W, Kaufer DI, Saxton J,
[107] Giulian D. Microglia and the immune pathology of Alzheimer dis- et al. Psychiatric symptoms vary with the severity of dementia in
ease. Am J Hum Genet 1999;65:13– 8. probable Alzheimer’s disease. J Neuropsychiatry Clin Neurosci
[108] McGeer PL, Schulzer M, McGeer EG. Arthritis and anti- 2003;15:346 –53.
inflammatory agents as possible protective factors for Alzheimer’s [130] Marin DB, Green CR, Schmeidler J, Harvey PD, Lawlor BA, Ryan
disease: a review of 17 epidemiologic studies. Neurology 1996;47: TM, et al. Noncognitive disturbances in Alzheimer’s disease: fre-
425–32. quency, longitudinal course, and relationship to cognitive symp-
[109] Wolfson C, Perrault A, Moride Y, Esdaile JM, Abenhaim L, Momoli toms. J Am Geriatr Soc 1997;45:1331– 8.
F. A case-control analysis of nonsteroidal anti-inflammatory drugs [131] Yesavage JA, Brink TL, Rose TL, Lum O, Huang V, Adey M, et al.
and Alzheimer’s disease: are they protective? Neuroepidemiology Development and validation of a geriatric depression screening
2002;21:81– 6. scale: a preliminary report. J Psychiatr Res 1982– 83;17:37– 49.
[110] Launer LJ. Nonsteroidal anti-inflammatory drugs and Alzheimer [132] Muller-Thomsen T, Arlt S, Mann U, Mass R, Ganzer S. Detecting
disease: what next? JAMA 2003;289:2865–7. depression in Alzheimer’s disease: evaluation of four different
[111] Aisen PS. The potential of anti-inflammatory drugs for the treatment scales. Arch Clin Neuropsychol 2005;20:271– 6.
of Alzheimer’s disease. Lancet Neurol 2002;1:279 – 84. [133] Teri L, Logsdon RG, Uomoto J, McCurry SM. Behavioral treatment
[112] Tabet N, Feldman H. Indomethacin for Alzheimer’s disease. Co- of depression in dementia patients: a controlled clinical trial. J
chrane Database Syst Rev 2002.CD003673. Gerontol B Psychol Sci Soc Sci 1997;52:P159 – 66.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 381

[134] Bains J, Birks JS, Dening TR. The efficacy of antidepressants in the [155] Reading PJ, Luce AK, McKeith IG. Rivastigmine in the treatment of
treatment of depression in dementia. Cochrane Database Syst Rev parkinsonian psychosis and cognitive impairment: preliminary find-
2002.CD003944. ings from an open trial. Mov Disord 2001;16:1171– 4.
[135] Munro CA, Brandt J, Sheppard JM, Steele CD, Samus QM, [156] Boeve BF, Silber MH, Ferman TJ. REM sleep behavior disorder in
Steinberg M, et al. Cognitive response to pharmacological treatment Parkinson’s disease and dementia with Lewy bodies. J Geriatr Psy-
for depression in Alzheimer disease: secondary outcomes from the chiatry Neurol 2004;17:146 –57.
depression in Alzheimer’s disease study (DIADS). Am J Geriatr [157] Poewe W. Treatment of dementia with Lewy bodies and Parkinson’s
Psychiatry 2004;12:491– 8. disease dementia. Mov Disord 2005;20(Suppl 12):S77– 82.
[136] Moretti R, Torre P, Antonello RM, Cazzato G, Bava A. Depression [158] Forbes DA. Strategies for managing the behavioral symptomatology
and Alzheimer’s disease: symptom or comorbidity? Am J Alzhei- associated with dementia of the Alzheimer type: a systematic over-
mers Dis Other Demen 2002;17:338 – 44. view. Can J Nurs Res 1998;30:67– 86.
[137] Lyketsos CG, Olin J. Depression in Alzheimer’s disease: overview [159] Opie J, Rosewarne R, O’Connor DW. The efficacy of psychosocial
and treatment. Biol Psychiatry 2002;52:243–52. approaches to behaviour disorders in dementia: a systematic litera-
[138] Lechevallier-Michel N, Molimard M, Dartigues JF, Fabrigoule C, ture review. Aust N Z J Psychiatry 1999;33:789 –99.
Fourrier-Reglat A. Drugs with anticholinergic properties and cog- [160] McMinn B, Draper B. Vocally disruptive behaviour in dementia:
nitive performance in the elderly: results from the PAQUID Study. development of an evidence based practice guideline. Aging Ment
Br J Clin Pharmacol 2005;59:143–51. Health 2005;9:16 –24.
[139] Fujishiro J, Imanishi T, Onozawa K, Tsushima M. Comparison of [161] Verkaik R, van Weert JCM, Francke AL. The effects of psychoso-
the anticholinergic effects of the serotonergic antidepressants, par- cial methods on depressed, aggressive and apathetic behaviors of
oxetine, fluvoxamine and clomipramine. Eur J Pharmacol 2002;454: people with dementia: a systematic review. Int J Geriatr Psychiatry
183– 8. 2005;20:301– 4.
[140] Herrmann N. Use of SSRIs in the elderly: obvious benefits but [162] Livingston G, Johnston K, Katona C, Paton J, Lyketsos CG; Old
unappreciated risks. Can J Clin Pharmacol 2000;7:91–5. Age Task Force of the World Federation of Biological Psychiatry.
[141] Liu B, Anderson G, Mittmann N, To T, Axcell T, Shear N. Use of Systematic review of psychological approaches to the management
selective serotonin-reuptake inhibitors of tricyclic antidepressants of neuropsychiatric symptoms of dementia. Am J Psychiatry 2005;
and risk of hip fractures in elderly people. Lancet 1998;351:1303–7.
162:1996 –2021.
[142] Pacher P, Ungvari Z. Selective serotonin-reuptake inhibitor antide-
[163] Woods B, Spector A, Jones C, et al. Reminiscence therapy for
pressants increase the risk of falls and hip fractures in elderly people
dementia. Cochrane Database Syst Rev 2005:CD001120.
by inhibiting cardiovascular ion channels. Med Hypotheses 2001;
[164] Neal M, Briggs M. Validation therapy for dementia. Cochrane
57:469 –71.
Database Syst Rev 2003.CD001394.
[143] American Geriatrics Society and American Association for Geriatric
[165] Forbes D, Morgan DG, Bangma J, et al. Light therapy for managing
Psychiatry. Consensus statement on improving the quality of mental
sleep, behaviour, and mood disturbances in dementia. Cochrane
health care in U.S. nursing homes: management of depression and
Database Syst Rev 2004:CD003946.
behavioral symptoms associated with dementia. J Am Geriatr Soc
[166] Vink AC, Birks JS, Bruinsma MS, et al. Music therapy for people
2003;51:1287–98.
with dementia. Cochrane Database Syst Rev 2003:CD003477.
[144] van der Wurff FB, Stek ML, Hoogendijk WJG, Beekman ATF. The
[167] Thorgrimsen L, Spector A, Wiles A, et al. Aroma therapy for
efficacy and safety of ECT in depressed older adults: a literature
dementia. Cochrane Database Syst Rev 2003:CD003150.
review. In J Geriatr Psychiatry 2003;18:894 –904.
[168] Chung JCC, Lai CKY. Snoezelen for dementia. Cochrane Database
[145] Ancoli-Israel S, Ayalon L. Diagnosis and treatment of sleep disor-
ders in older adults. Am J Geriatr Psychiatry 2006;14:95–103. Syst Rev 2002.CD003152.
[146] Carpenter BD, Strauss ME, Patterson MB. Sleep disturbances in [169] Viggo Hansen N, Jørgensen T, Ørtenblad L. Massage and touch for
community-dwelling patients with Alzheimer’s disease. Clin Ger- dementia. Cochrane Database Syst Rev 2004.CD004989.
ontol 1995;16:35– 49. [170] Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi
[147] Gaugler JE, Edwards AB, Femia EE, et al: Predictors of institution- DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive
alization of cognitively impaired elders: family help and the timing assessment of psychopathology in dementia. Neurology 1994;44:
of placement. J Gerontol B Psychol Sci Soc Sci 2000;55B:P247–55. 2308 –14.
[148] McCurry SM, Gibbons LE, Logsdon RG, et al. Training caregivers [171] Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell
to change the sleep hygiene practices of patients with dementia: the Scale for Depression in Dementia. Biol Psychiatry 1988;23:271– 84.
NITE-AD project. J Am Geriatr Soc 2003;51:455– 60. [172] Tariot PN, Mack JL, Patterson MB, Edland SD, Weiner MF, Fil-
[149] McCurry SM, Gibbons LE, Logsdon RG, et al. Nighttime insomnia lenbaum G, et al: The Behavior Rating Scale for Dementia of the
treatment and education for Alzheimer’s disease: a randomized, Consortium to Establish a Registry for Alzheimer’s Disease: the
controlled trial. J Am Geriatr Soc 2005;53:793– 802. Behavioral Pathology Committee of the Consortium to Establish a
[150] Glass J, Lanctôt KL, Herrmann N, Sproule BA, Busto UE. Sedative Registry for Alzheimer’s Disease. Am J Psychiatry 1995;152:
hypnotics in older people with insomnia: meta-analysis of risks and 1349 –57.
benefits. BMJ 2005;331:1169. [173] Rosen WG, Mohs RC, Davis KL. A new rating scale for Alzhei-
[151] Bliwise DL. Sleep disorders in Alzheimer’s disease and other de- mer’s disease. Am J Psychiatry 1984;141:1356 – 64.
mentias. Clin Cornerstone 2004;6(Suppl 1A):S16 –28. [174] Reisberg B, Borenstein J, Salob SP, Ferris SH, Franssen E, Geor-
[152] McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman gotas A. Behavioral symptoms in Alzheimer’s disease: phenome-
H, et al. Diagnosis and management of dementia with Lewy bodies: nology and treatment. J Clin Psychiatry 1987;48(Suppl):9 –15.
third report of the DLB consortium. Neurology 2005;65:1863–72. [175] Gottfries CG, Brane G, Steen G. A new rating scale for dementia
[153] Steriade M. Basic mechanisms of sleep generation. Neurology 1992; syndromes. Gerontology 1982;28(Suppl 2):20 –31.
42(Suppl 6):9 –18. [176] Cohen-Mansfield J, Billig N. Agitated behaviors in the elderly. I. A
[154] Ringman JM, Simmons JH. Treatment of REM sleep behavior conceptual review. J Am Geriatr Soc 1986;34:711–21.
disorder with donepezil: a report of three cases. Neurology 2000; [177] Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase
55:870 –1. inhibitors in the treatment of neuropsychiatric symptoms and func-
382 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

tional impairment in Alzheimer disease: a meta-analysis. JAMA [196] Forbes DA, Debra D, Janzen BL. Rural and urban Canadians with
2003;289:210 – 6. dementia: use of health care services. Can J Aging 2006;25:321–30.
[178] Cummings JL, Donohue JA, Brooks RL. The relationship between [197] Edwards DF, Baum CM, Meisel M, et al. Home-based multidisci-
donepezil and behavioral disturbances in patients with Alzheimer’s plinary diagnosis and treatment of inner-city elders with dementia.
disease. Am J Geriatr Psychiatry 2000;8:134 – 40. Gerontologist 1999;39:483– 8.
[179] Winblad B, Engedal K, Soininen H, Verhey F, Waldemar G, Wimo [198] Morgan D, Semchuk K, Stewart N, et al. Rural families caring for a
A, et al. A 1-year, randomized, placebo-controlled study of done- relative with dementia: barriers to use of formal services. Soc Sci
pezil in patients with mild to moderate AD. Neurology 2001;57: Med 2002;55:51– 64.
489 –95. [199] Smale B, Dupuis S. A literature review. caregivers of persons with
[180] Holmes C, Wilkinson D, Dean C, Vethanayagam S, Olivieri S, dementia—roles, experiences, supports, and coping. Murray Alzhei-
Langley A, et al. The efficacy of donepezil in the treatment of mer Research and Education Program, University of Waterloo;
neuropsychiatric symptoms in Alzheimer disease. Neurology 2004; 2004.
63:214 –9. [200] Zarit SH, Gaugler JE, Jarrott SE. Useful services for families:
[181] Courtney C, Farrell D, Gray R, Hills R, Lynch L, Sellwood E, et al. research findings and directions. Int J Geriatr Psychiatry 1999;14:
Long-term donepezil treatment in 565 patients with Alzheimer’s 165–77.
disease (AD2000): randomised double-blind trial. Lancet 2004;363: [201] Gauthier S. Advances in the pharmacotherapy of Alzheimer’s dis-
2105–15. ease. CMAJ 2002;166:616 –23.
[182] Seltzer B, Zolnouni P, Nunez M, Goldman R, Kumar D, Ieni J, et al. [202] Newcomer R, Miller R, Clay T, et al. Effects of the Medicare
Efficacy of donepezil in early-stage Alzheimer disease: a random- Alzheimer’s disease demonstration on Medicare expenditures.
ized placebo-controlled trial. Arch Neurol 2004;61:1852– 6. Health Care Financ Rev 1999;20:45– 65.
[183] Starkstein SE, Mayberg HS, Preziosi TJ, Andrezejewski P, Lei- [203] Newcomer R, Spitalny M, et al. Effects of the Medicare Alzheimer’s
guarda R, Robinson RG. Reliability, validity, and clinical correlates Disease Demonstration on the use of community-based services.
of apathy in Parkinson’s disease. J Neuropsychiatry Clin Neurosci Health Serv Res 1999;34:645– 67.
1992;4:134 –9. [204] Douglass C, Visconti C. Factors influencing the use of adult day care
[184] Rösler M, Retz W, Retz-Junginger P, Dennler HJ. Effects of two- by individuals with Alzheimer’s disease: a multivariate examination
of the California Alzheimer’s Disease Diagnostic and Treatment
year treatment with the cholinesterase inhibitor rivastigmine on
Center Program. Home Health Care Serv Q 1998;17:53– 69.
behavioural symptoms in Alzheimer’s disease. Behav Neurol 1998;
[205] Droes RM, Breebaart E, Ettema T, et al. Effect of integrated family
11:211– 6.
support versus day care only on behavior and mood of patients with
[185] Finkel SI. Effects of rivastigmine on behavioral and psychological
dementia. Int Psychogeriatr 2000;12:99 –115.
symptoms of dementia in Alzheimer’s disease. Clin Ther 2004;26:
[206] Droes RM, Breebaart E, Meiland FJM, et al. Effect of Meeting
980 –90.
Centres Support Program on feelings of competence of family carers
[186] Herrmann N, Rabheru K, Wang J, Binder C. Galantamine treatment
and delay of institutionalization of people with dementia. Aging
of problematic behavior in Alzheimer disease: post-hoc analysis of
Ment Health 2004;8:201–11.
pooled data from three large trials. Am J Geriatr Psychiatry 2005;
[207] Droes RM, Meiland F, et al. Effect of combined support for people
13:527–34.
with dementia and carers versus regular day care on behaviour and
[187] Areosa SA, Sherriff F, McShane R. Memantine for dementia. Co-
mood of persons with dementia: results from a multi-centre imple-
chrane Database Syst Rev 2005.CD003154.
mentation study. Int J Geriatr Psychiatry 2004;19:673– 84.
[188] Peskind ER, Potkin SG, Pomara N, McDonald S, Xie Y, Gergel I.
[208] Berger G, Bernhardt T, Schramm U, et al. No effects of a combi-
Memantine monotherapy is effective and safe for the treatment of nation of caregivers support group and memory training/music ther-
mild to moderate Alzheimer’s disease. a randomized controlled trial. apy in dementia patients from a memory clinic population. Int J
American Association for Geriatric Psychiatry, 17th Annual Meet- Geriatr Psychiatry 2004;19:223–31.
ing, Baltimore, Maryland, February 21–24, 2004. [209] Gendron CE, Poitras LR, Dastoor DP, et al. Cognitive-behavior
[189] Tiraboschi P, Salmon DP, Hansen LA, Hofstetter RC, Thal LJ, group intervention for spousal caregivers: findings and clinical con-
Corey-Bloom J. What best differentiates Lewy body from Alzhei- siderations. Clin Gerontol 1996;17:3–19.
mer’s disease in early stage dementia? Brain 2006;129:729 –35. [210] Haley WE, Brown SL, Levine EG. Experimental evaluation of the
[190] Cohen-Mansfield J, Werner P, Watson V, et al. Agitation among effectiveness of group intervention for dementia caregivers. Geron-
elderly persons at adult day-care centres: the experiences of relatives tologist 1987;27:376 – 82.
and staff members. Int Psychogeriatr 1995;7:447–58. [211] Hébert R, Levesque L, Vezina J, et al. Efficacy of a psychoeducative
[191] Phillips VL, Diwan S. The incremental effect of dementia-related group program for caregivers of demented persons living at home: a
problem behaviors on the time to nursing home placement in poor, randomized controlled trial. J Gerontol B Psychol Sci Soc Sci
frail, demented older people. J Am Geriatr Soc 2003;51:188 –93. 2003;58:S58 – 67.
[192] Diwan S, Phillips VL. Agitation and dementia-related problem be- [212] Eloniemi-Sulkava U, Notkola IL, Hentinen M, et al. Effects of
haviors and case management in long-term care. Int Psychogeriatr supporting community-living demented patients and their caregiv-
2001;13:5–21. ers: a randomized trial. J Am Geriatr Soc 2001;49:1282–7.
[193] Canadian Study of Health and Aging Working Group. Canadian [213] Hall GR, Buckwalter KC. Progressively lowered stress threshold: a
Study of Health and Aging. study methods and prevalence of de- conceptual model for care of adults with Alzheimer’s disease. Arch
mentia. CMAJ 1994;150:899 –913. Psychiatr Nurs 1987;1:399 – 406.
[194] Czaja SJ, Eisdorfer C, Schulz R. Future directions in caregiving: [214] Gerdner LA, Buckwalter KC, Reed D. Impact of a psychoeduca-
implications for intervention research. In: Schulz R, ed. Handbook tional intervention on caregiver response to behavioral problems.
of dementia caregiving intervention research. New York: Springer Nurs Res 2002;51:363–74.
Publishing Company; 2000. p. 283–319. [215] Huang HL, Shyu YI, Chen MC, et al. A pilot study on a home-based
[195] Canadian Study of Health and Aging Working Group. Patterns of caregiver training program for improving caregiver self-efficacy and
caring for people with dementia in Canada. Can J Aging 1994;13: decreasing the behavioral problems of elders with dementia in Tai-
470 – 87. wan. Int J Geriatr Psychiatry 2003;18:337– 45.
D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384 383

[216] Gormley N, Lyons D, Howard R. Behavioural management of [240] Gouvier WD, Maxfield MW, Schweitzer JR, et al. Psychometric
aggression in dementia: a randomized controlled trial. Age Ageing prediction of driving performance among the disabled. Arch Phys
2001;30:141–5. Med Rehabil 1989;70:745–50.
[217] Bourgeois MS, Burgio LD, Schulz R, et al. Modifying repetitive [241] Kumar R, Powell B, Tani N, Naliboff B, Metter EJ. Perceptual
verbalizations of community-dwelling patients with AD. Gerontol- dysfunction in hemiplegia and automobile driving. Gerontology
ogist 1997;37:30 –9. 1991;31:807–10.
[218] Rowe M, Alfred D. The effectiveness of slow-stroke massage in [242] Uc E, Rizzo M, Anderson SW, Shi Q, Dawson JD. Driver landmark
diffusing agitated behaviors in individuals with Alzheimer’s disease. and traffic sign identification in early Alzheimer’s disease. J Neurol
J Gerontol Nurs 1999;25:22–34. Neurosurg Psychiatr 2005;76:764 – 8.
[219] Baker R, Bell S, Baker E, et al. A randomized controlled trial of the [243] Galski T, Bruno RL, Ehle HT. Driving after cerebral damage: a
effects of multi-sensory stimulation (MSS) for people with demen- model with implications for evaluation. Am J Occup Ther 1992;46:
tia. Br J Clin Psychol 2001;40(Pt 1):81–96. 324 –32.
[220] Keplinger FS. The elderly driver: who should continue to drive? [244] Simms B. Perception and driving: theory and practice. Br J Occup
Phys Med Rehabil 1998;12:147–55. Ther 1985;48:363– 6.
[221] Marottoli RA, Mendes de Leon CF, Glass TA, et al. Driving ces- [245] Sundet K, Goffeng L, Hofft E. To drive or not to drive: neuropsy-
sation and increased depressive symptoms: prospective evidence chological assessment for driver’s license among stroke patients.
from the New Haven EPESE. J Am Geriatr Soc 1997;45:202– 6. Scand J Psychol 1995;36:47–58.
[222] Marottoli RA, Glass TA, Williams CS, Cooney LMJ, Berkman LF. [246] Sivak M, Olson P, Kewman DG, Won H, Henson DL. Driving and
Consequences of driving cessation: decreased out-of-home activity perceptual/cognitive skills: behavioral consequences of brain dam-
levels. J Gerontol 2000;55:S334 – 40. age. Arch Phys Med Rehabil 1981;62:476 – 83.
[223] Johnson JE. Urban older adults and the forfeiture of a driver’s [247] Molnar FJ, Patel A, Marshall SC, Man-Son-Hing M, Wilson KG.
license. J Gerontol Nurs 1999;12:12– 8. Clinical utility of office-based cognitive predictors of fitness to drive
[224] Bonnel WB. Giving up the car: older women’s losses and experi- in persons with dementia: a systematic review. J Am Geriatr Soc
ences. J Psychosoc Nurs Mental Health Services 1999;37:10 –5. 2006;54:1809 –24.
[225] O’Neill D. Predicting and coping with the consequences of stopping [248] Folstein JF, Folstein SE, McHugh PR. Mini-mental State: a practical
driving. Alzheimer Dis Assoc Disord 1997;11(Suppl 1):70 –2.
method for grading the cognitive state of patients for the clinician.
[226] Johnson JE. Older rural adults and the decision to stop driving: the
J Psychiatr Res 1975;12:189 –98.
influence of family and friends. J Community Health Nurs 1998;15:
[249] Dubinsky RM, Stein AC, Lyons K. Practice parameter: risk of
105–16.
driving in Alzheimer’s disease. Neurology 2000;54:2205–11.
[227] Rosenbloom S. Transportation needs of the elderly population. Clin
[250] Australian Society for Geriatric Medicine. Dementia and driving.
Geriatr Med 1993;9:297–310.
Accessed February 10, 2006. Available at: http://www.asgm.org.au/
[228] Marshall S, Gilbert N. Saskatchewan physicians’ knowledge and
pdfdocs/position_statements/PositionStatementNo11.pdf. Accessed
attitudes regarding assessment of medical fitness to drive. Can Med
February 10, 2006.
Assoc J 1999;160:1701– 4.
[251] Hunt L, Morris JC, Edwards D, Wilson BS. Driving performance in
[229] Friedland R, Koss E, Kumar A, et al: Motor vehicle crashes in
persons with mild senile dementia of the Alzheimer type. J Am
dementia of the Alzheimer type. Ann Neurol 1988;24:782– 6.
Geriatr Soc 1993;41:747–52.
[230] Trobe JD, Waller PF, Cook-Flannagan CA, Teshima SM, Bieliaus-
[252] Fox GK, Bowden SC, Bashford GM, Smith DS. Alzheimer’s disease
kas LA. Crashes and violations among drivers with Alzheimer
and driving: prediction and assessment of driving performance.
disease. Arch Neurol 1996;53:411– 6.
[231] Carr DB, Duchek JMJ. Characteristics of motor vehicle crashes of J Am Geriatr Soc 1997;45:949 –53.
drivers with dementia of the Alzheimer type. J Am Geriatr Soc [253] Schulz R, O’Brien AT, Bookwala J, Fleissner K. Psychiatric and
2000;48:18 –22. physical morbidity effects of dementia caregiving: prevalence, cor-
[232] Fitten LJ, Perryman KM, Wilkinson CJ, Little RJ, Burns MM, relates and causes. Gerontologist 1995;35:771–91.
Pachana N, et al. Alzheimer and vascular dementias and driving: a [254] Schulz R, Martire LM. Family caregiving of persons with dementia.
prospective road and laboratory study. JAMA 1995;273:1360 –5. Am J Geriatr Psychiatry 2004;12:240 –9.
[233] Galski T, Ehle HT, Bruno RL. An assessment of measures to predict [255] Hébert R, Dubois MF, Wolfson C, Chambers L, Cohen C. Factors
the outcome of driving evaluations in patients with cerebral damage. associated with long-term institutionalization of older people with
Am J Occup Ther 1990;44:709 –13. dementia: data from the Canadian Study of Health and Aging. J
[234] Korteling JE, Kaptein MA. Neuropsychological driving fitness tests Gerontol Biol Sci Med Sci 2001;56:M693–9.
for brain-damaged subjects. Arch Phys Med Rehabil 1996;77: [256] Pusey H, Ricards D. A systematic review of the effectiveness of
138 – 46. psychosocial interventions for carers of people with dementia. Ag-
[235] Odenheimer GL, Beaudet M, Jette AM, Albert MS, Grande L, ing Ment Health 2001;5:107–19.
Minaker KL. Performance-based driving evaluation of the elderly [257] Schulz R, O’Brien A, Czaja A, Ory M, Norris R, Martire LM, et al.
driver: safety, reliability, and validity. J Gerontol 1994;49:M153–9. Dementia caregiver intervention research: In search of clinical sig-
[236] Fox GK, Bashford GM, Caust SL. Identifying safe versus unsafe nificance. Gerontologist 2002;42:589 – 602.
drivers following brain impairment: the Coorabel Programme. Dis- [258] Brodaty H, Green A, Koschera A. Meta-analysis of psychosocial
abil Rehabil 1992;14:140 –5. intervention for caregivers of people with dementia. J Am Geriatr
[237] van Zomeren AH, Brouwer WH, Rothengatter JA, Snoek JW. Fit- Soc 2003;51:657– 64.
ness to drive a car after recovery from a severe head injury. Arch [259] Lee H, Cameron M. Respite care for people with dementia and their
Phys Med Rehabil 1988;69:90 – 6. carers. Cochrane Database Syst Rev 2004.CD004396.
[238] Brooke MM, Questad KA, Patterson DR, Valois TA. Driving eval- [260] Lingler JH, Martire LM, Schulz R. Caregiver-specific outcomes in
uation after traumatic brain injury. Am J Phys Med Rehabil 1992; antidementia clinical drug trials: a systematic review and meta-
71:177– 82. analysis. J Am Geriatr Soc 2005;53:983–90.
[239] Mazer BL, Korner-Bitensky NA, Sofer S. Predicting ability to drive [261] Doody RS, Stevens JC, Beck C, Dubinsky RM, Kaye JA, Gwyther
after stroke. Arch Phys Med Rehabil 1998;79:743–50. L, et al. Practice parameter: management of dementia (an evidence-
384 D.B. Hogan et al. / Alzheimer’s & Dementia 3 (2007) 355–384

based review—report of the Quality Standards Subcommittee of the [281] Defilippi JL, Crismon ML. Drug interactions with cholinesterase
American Academy of Neurology. Neurology 2001;56:1154 – 66. inhibitors. Drugs Aging 2003;20:437– 44.
[262] American Association of Geriatric Psychiatry. Family and caregiver [282] Bieliauskas LA, Roper BR, Trobe J, Green P, Lacy M. Cognitive
counseling in dementia: medical necessity. Available at: http:// measures, driving safety and Alzheimer’s disease. Clin Neuropsy-
www.aagponline.org/prf/position_dementia.asp. Accessed February chol 1998;12:206 –12.
12, 2006. [283] Carr DB, Labarge E, Dunnigan K, Storandt M. Differentiating
[263] American Medical Association. Caregiver health. Available at: drivers with dementia of the Azheimer type from healthy older
http://www.ama-assn.org/ama/pub/category/5099.html. Accessed July persons with a traffic sign recognition test. J Gerontol 1998;53:
2, 2007. M135–9.
[264] Foster NL. Barriers to treatment: the unique challenges for physi- [284] Cooper PJ, Tallman K, Tuokko H, Beattie BL. Vehicle crash in-
cians providing dementia care. J Geriatr Psychiatry Neurol 2001;14: volvement and cognitive deficit in older drivers. J Safety Res 1993;
188 –98. 24:9 –17.
[265] Barrett JJ, Haley WE, Harrell LE, Powers RE. Knowledge about [285] Cox DJ, Quillian WC, Thorndike FP, Kovachev BP, Hanna G.
Alzheimer disease among primary care physicians, psychologists, Evaluating driving performance of outpatients with Alzheimer Dis-
nurses, and social workers. Alzheimer Dis Assoc Disord 1997;11: ease. J Am Board Fam Pract 1998;11:264 –71.
99 –106. [286] Cushman L. Cognitive capacity and concurrent driving performance
[266] Pucci E, Angeleri F, Borsetti G, Brizioli E, Cartechini E, Giuliana G, in older drivers. IATSS Research 1996;20:38 – 45.
et al. General practitioners facing dementia: are they fully prepared? [287] Drachman DA, Swearer JM. Driving and Alzheimer’s disease: the
Neurol Sci 2004;24:384 –9. risk of crashes. Neurology 1993;43:2448 –56.
[267] Turner S, Iliffe S, Downs M, Wilcock J, Bryans M, Levin E, et al. [288] Duchek JM, Hunt L, Ball K, Buckles V, Morris JC. Attention and
General practitioners’ knowledge, confidence and attitudes in the driving performance in Alzheimer’s disease. J Gerontol 1998;53:
diagnosis and management of dementia. Age Ageing 2004;33: 130 – 41.
461–7. [289] Duchek JM, Carr DB, Hunt L, et al. Longitudinal driving perfor-
[268] Olafsdottir M, Foldevi M, Marcusson J. Dementia in primary care: mance in early stage dementia of the Alzheimer type. J Am Geriatr
why the low detection rate? Scand J Prim Health Care 2001;19: Soc 2003;51:1342–7.
[290] Harvey R, Fraser D, Bonner D, Warnes A, Warrington E, Rossor M.
194 – 8.
Dementia and driving: results of a semi-realistic simulator study. Int
[269] Gifford DR, Holloway RG, Frankel MR, Albright CL, Meyerson R,
J Geriatr Psychiatr 1995;10:859 – 64.
Griggs RC, et al. Improving adherence to guidelines through edu-
[291] Hunt L. Dementia and road test performance. VTI Rapport 372A
cation and opinion leaders: a randomized, controlled trail. Ann
1989.
Intern Med 1999;131:237– 46.
[292] Hunt LA, Morris JC, Edwards D, Wilson BS. Driving performance
[270] Waldorff FB, Almind G, Makela M, Moller S, Waldemar G. Im-
in persons with mild senile dementia of the Alzheimer type. J Am
plementation of a clinical dementia guideline: a controlled study on
Geriatr Soc 1993;41:747–52.
the effect of a multifaceted strategy. Scand J Prim Health Care
[293] Hunt LA, Murphy CF, Carr D, Duchek JM, Buckles V, Morris JC.
2003;21:142–7.
Reliability of the Washington University Road Test: a performance-
[271] Cairns R, Evans J, Prince M. The impact of the NICE guidelines on
based assessment for drivers with dementia of the Alzheimer type.
the diagnosis and treatment of Alzheimer’s disease among general
Arch Neurol 1997;54:707–12.
medical inpatients. Int J Geriatr Psychiatry 2004;19:800 –2.
[294] Ott BR, Heindel WC, Whelihan WM, Caron MD, Piatt AL, DiCarlo
[272] Pereles I, Russell ML. Needs for CME in geriatrics. Part 2: physi- MA. Maze test performance and reported driving ability in early
cian priorities and perceptions of community representatives. Can dementia. J Geriatr Psychiatr Neurol 2003;16:151–5.
Fam Physician 1996;42:632– 40. [295] Rebok GW, Keyl PM, Blaustein MJ, Tune L. The effects of Alz-
[273] Gaugler JE, Kane RL, Kane RA, Newcomer R. Early community- heimer disease on driving-related abilities. Alzheimer Dis Assoc
based service utilization and its effects on institutionalization in Disord 1994;8:228 – 40.
dementia caregiving. Gerontologist 2005;45:177– 85. [296] Rizzo M, Reinach S, McGehee D, Dawson J. Simulated car crashes
[274] Wolff JL, Boult C. Moving beyond round pegs and square holes: and crash predictors in drivers with Alzheimer disease. Arch Neurol
restructuring Medicare to improve chronic care. Ann Intern Med 1997;54:545–51.
2005;143:439 – 45. [297] Rizzo M, McGehee D, Dawson J, Anderson SN. Simulated car
[275] Fillit HM. Managed care decisions in Alzheimer disease. Am J crashes at intersection in drivers with Alzheimer disease. Alzheimer
Managed Care 2000;6(Suppl):S1149 – 6. Dis Assoc Disord 2001;15:10 –20.
[276] Cherry DL, Vickrey BG, Schwankovsky L, Heck E, Plauchm M, [298] Tuokko H, Tallman K, Beattie BL, Cooper P, Weir J. An examina-
Yep R. Interventions to improve quality of care: the Kaiser tion of driving records in a dementia clinic. J Gerontol 1995;50:
Permanente-Alzheimer’s Association Dementia Care Project. S173– 81.
Am J Manag Care 2004;10:553– 60. [299] Wald JL. Neuropsychological impairment and driving performance
[277] Rockman P, Salach L, Gotlib D, Cord M, Turner T. Shared mental in dementia. PhD thesis, University of Alberta. 1998.
health care: model for supporting and mentoring family physicians. [300] Waller PF, Trobe JD, Olson PL. Crash characteristics associated
Am Fam Physician 2004;50:397– 402. with early Alzheimer disease. Proc Assoc Advance Automotive Med
[278] Iliffe S, Wilcock J, Haworth D. Obstacles to shared care for patients 1993;1:157–73.
with dementia: a qualitative study. Fam Pract 2006;23:353– 62. [301] Whelihan WM, Dicarlo MA, Paul RH. The relationship of neuro-
[279] Fillit H, Geldmacher DS, Welter RT, Maslow K, Fraser M. Opti- psychological functioning to driving competence in older persons
mizing coding and reimbursement to improve management of Alz- with early cognitive decline. Arch Clin Neuropsychol 2005;20:
heimer’s disease and related dementias. J Am Geriatr Soc 2002;50: 217–28.
1871– 8. [302] Zuin D, Ortiz H, Boromei D, Lopez OL. Motor vehicle crashes and
[280] Mintzer J, Burns A. Anticholinergic side-effects of drugs in elderly abnormal driving behaviors in patients with dementia in Mendoza,
people. J R Soc Med 2000;93:457– 62. Argentina. Eur J Neurol 2002;9:29 –34.
Alzheimer’s & Dementia 3 (2007) 385–397

Clinical practice guidelines for severe Alzheimer’s disease


Nathan Herrmanna,*, Serge Gauthierb, Paul G. Lysyc
a
Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
b
Alzheimer’s Disease Research Unit, McGill Center for Studies in Aging, and Department of Neurology, McGill University, Montreal, Quebec, Canada
c
Department of Family Medicine, McGill University, Montreal, Quebec, Canada

Abstract Background: Although severe Alzheimer’s disease (AD) represents a prevalent, serious, and
costly public health problem, few practice guidelines exist to help physicians manage this disorder.
Methods: A search of English language medical databases was performed from 1996 to the present
for articles pertaining to the management of AD. The focus of this review was on studies that
included patients with severe disease. Studies were assessed by considering the subjects, trial design,
analysis, and results. Recommendations were based on the best available evidence.
Results: Severe AD can be defined and diagnosed reliably by using measures of cognition,
function, behavior, and global staging. Specific assessments would also include medical status,
safety issues, and the health status of the caregiver. Disease-specific management would include
treatment with cholinesterase inhibitors and/or memantine. Treatment of neuropsychiatric symptoms
begins with nonpharmacologic behavioral and environmental approaches. Severe agitation, aggres-
sion, and psychosis that are potentially dangerous to the patient, caregiver, and others in the
environment can be treated with atypical antipsychotics, with consideration of their increased risk
of cerebrovascular adverse events and mortality. All pharmacologic approaches require careful
monitoring and regular periodic reassessments to determine whether ongoing treatment is necessary.
Conclusions: Evidence-based guidelines for the management of severe AD have the potential to
improve the quality of life for the patient and their caregiver. More randomized controlled trials
aimed specifically at this phase of illness are still urgently required.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Alzheimer’s disease; Severe dementia; Therapy; Practice guidelines

1. Introduction often considered a hallmark of the more severe stages of the


illness. This dependence leads to increased caregiver time,
Alzheimer’s disease (AD) is characterized by a progressive increased caregiver burden and stress, and increased risk of
impairment of cognition, function, and behavior. Although institutionalization [2]. The costs of caring for patients with
numerous reviews and clinical practice guidelines (CPGs) for AD rise dramatically from mild to moderate to severe illness
AD exist, most have focused on mild to moderate illness. Only [9], and the severe stages of the illness might account for up to
recently has focus shifted to more advanced stages of the 70% to 80% of the total costs [10].
illness [1–5]. The Canadian Study of Health and Aging The significant prevalence, disability, burden, and cost as-
(CSHA) estimated that 50% of individuals diagnosed with AD sociated with severe AD clearly make this an important soci-
were in the moderate to severe stages of the illness, and 90% etal health problem. Development of CPGs for this stage of
of residents in long-term care facilities were in moderate to illness was, therefore, recommended.
severe stages [6]. Disease severity increases with age [7] and
negatively affects survival [8]. With progression of the disease,
patients with AD become totally dependent on caregivers, 2. Methods
This article represents the background for CPGs for
*Corresponding author. Tel.: 416-480-6133; Fax: 416-480-6022. severe AD as part of the Third Canadian Consensus Con-
E-mail address: n.herrmann@utoronto.ca ference on Diagnosis and Treatment of Dementia. A liter-
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.007
386 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

ature review was performed with PubMed and Embase Table 1


databases that included articles in English language, human Recommendations for the management of severe AD (levels of evidence
appear in parentheses)
research, and publications from 1996 to the present. Specific
search terms included the following: dementia or Alzhei- 1. Severe AD can be defined as the stage in which the patient
becomes totally dependent on a caregiver for survival. This will
mer’s disease, severe, therapy or treatment, cholinesterase
typically correspond to MMSE ⬍10 and GDS 6 to7
inhibitors, antioxidants, hormones, anti-inflammatory drugs, (Grade B, Level 2).
neurotropics, metabolic enhancers, neurotropic agents, me- 2. Patients with severe AD should be assessed at least every 4 months
mantine, gingko, hypnotics, antidepressants, anxiolytics, se- or if treated with pharmacotherapy at least every 3 months
lective serotonin reuptake inhibitors, sleep, depression, anx- (Grade C, Level 3).
3. Assessment should include cognition (eg MMSE) function behavior
iety, agitation, disinhibition, affective disorders, counseling,
medical status nutrition safety and caregiver health
education, caregiver environment, behavior, and rehabilita- (Grade B, Level 3).
tion. We attempt to provide recommendations on the defi- 4. The goals for management are to improve the quality of life for
nition, assessment, and management of severe AD by using patient and caregivers maintain optimal function and provide
an evidence-based approach and focusing particularly on maximum comfort (Grade B, Level 3).
5. Medical management includes treatment of intercurrent medical
studies published since 1998 (the date of the last Canadian
conditions (eg infections parkinsonian symptoms seizures pressure
Consensus Conference on Dementia). Recommendations ulcers) ameliorating pain improving nutritional status and
(Table 1) were rated on the basis of rules of evidence optimizing sensory function (Grade B, Level 3).
developed by the Canadian Task Force on the periodic 6. Patients with severe AD can be treated with ChEIs memantine or
health examination [11]. the combination. Expected benefits would include modest
improvements or slower decline in cognition function and behavior
(Grade A, Level 1).
7. Treatment with ChEIs and/or memantine should persist until
3. Results clinical benefit can no longer be demonstrated. Treatment should
not be discontinued simply because of institutionalization
3.1. Definition (Grade C, Level 3).
8. The management of BPSD should begin with appropriate
There are no current widely accepted consensus criteria
assessments diagnosis and identification of target symptoms and
for severe AD. In the only published CPGs for severe AD consideration of safety of the patient their caregiver and others in
[5], moderately severe AD was defined as a Mini-Mental their environment (Grade B, Level 3).
State Examination score (MMSE) [12] of 10 to 15, severe 9. Nonpharmacologic treatments should be initiated first. Approaches
AD 3 to 9, and very severe AD ⬍3. These authors noted that that might be useful for severe AD include behavioral management
for depression and caregivers/staff education programs for a variety
although these cognitive function scores generally correlate
of behaviors. Music and multi-sensory intervention (Snoezelen) are
with increasing dependence and neuropsychiatric symp- useful during treatment sessions but longer-term benefits have not
toms, this definition requires validation and clearly does not been demonstrated (Grade B, Level 1).
encompass a rating of the functional capacity, behavior, and 10. Pharmacologic interventions should be initiated concurrently with
medical status of the patient. Several other reviews have nonpharmacologic approaches in the presence of severe depression
psychosis or aggression that puts the patient or others at risk of
also suggested that MMSE scores ⬍10 represent severe AD
harm (Grade B, Level 3).
[1,13–15]. 11. Pharmacologic interventions for BPSD should be initiated at the
Another approach is to define severe AD as a stage in lowest doses titrated slowly and monitored for effectiveness and
which the patient becomes dependent on a caregiver for safety (Grade B, Level 3).
their survival. This approach encompasses not only cogni- 12. Attempts to taper and withdraw medications for BPSD after a
period of 3 months of behavioral stability should occur in a
tive decline but also functional impairment and forms the
standardized fashion (Grade A, Level 1).
basis of several commonly used global staging instruments. 13. Risperidone and olanzapine can be used for severe agitation
For example, the Global Deterioration Scale (GDS) [16] aggression and psychosis. The potential benefit of all antipsychotics
defines moderately severe dementia as the stage in which must be weighed against the potential risks such as cerebrovascular
the patient can no longer survive without some assistance, adverse events and mortality (Grade A, Level 1).
14. There is insufficient evidence to recommend for or against the use
severe dementia as the stage when patients are entirely
of trazodone in the management of nonpsychotic agitated patients
dependent on a caregiver for survival, and very severe (Grade C, Level 3).
dementia as the stage in which basic psychomotor and 15. Benzodiazepines should be used only for short periods as prn
verbal skills are lost and “the brain appears to no longer be agents (Grade B, Level 1).
able to tell the body what to do.” Similarly, the Clinical 16. Selective serotonin reuptake inhibitors can be used for the
treatment of severe depression (Grade B, Level 3).
Dementia Rating Scale (CDR) [17,18] suggests that for
17. If BPSD fails to improve after appropriate nonpharmacologic and
moderate and severe stages, there is no pretense of inde- pharmacologic interventions refer to a specialty service
pendent function outside the home. In their stages, both (Grade B, Level 3).
scales include clinical descriptors related to cognition (eg,
memory, orientation, language), function, and physical
characteristics (eg, incontinence, bedridden), whereas the
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 387

GDS also includes descriptors of behavior (eg, delusions, AD. Although not designed specifically for moderate to
agitation). severe AD, the Disability Assessment for Dementia (DAD)
Because global staging is far more holistic, it would [29], a 40-item functional inventory, has also been used
theoretically be the preferred approach for defining severe successfully in a clinical trial of moderately severe AD.
dementia. The advantages of defining severe dementia as an Both of these scales are fairly lengthy; therefore they would
MMSE score ⬍10 is the fact that this instrument is struc- be unsuitable for use in primary care settings. In contrast,
tured and already well-known to Canadian physicians, and the ADL Index evaluates only six basic ADLs assessed for
its use is already incorporated in drug benefit formulary degree of assistance required (none, some, or complete)
requirements for most provinces. Furthermore, studies have [30]. This scale has been recommended by previous severe
suggested that MMSE scores are surprisingly strongly cor- AD guidelines [5]. Finally, it must be noted that assessment
related with global staging scores [19]. If clinicians should of ADL function in severe AD will need to consider the
decide to rely on the MMSE scores for the definition of context and environment in which the assessment occurs;
severe AD, they must be aware of the limitations of this many patients, especially those in long-term care, will not
scale’s use in this population, described in the following have the opportunity to demonstrate competence in many
section. areas, because their caregivers might assist or perform these
to facilitate care.
3.2. Assessment
3.2.3. Global assessment
Assessing patients with severe dementia requires a multi-
As noted previously, global assessment or staging can be
factorial approach that would include cognition, function,
used for defining severe AD. The potential advantages of
behavior, medical status, safety, and medicolegal issues, as
global assessment include avoiding the floor effects of other
well as caregiver status. Patients with severe AD live 3 to 4
instruments, assessing longitudinal change and the effects
[8] years during which numerous medical and psychosocial
of treatment, and helping predict clinical course for care
crises might arise. It is, therefore, recommended that pa-
planning [31].
tients in this stage be assessed at least every 4 months.
Among the most commonly used global assessment
3.2.1. Cognitive assessment scales in clinical research are the GDS and CDR. The CDR
It is widely recognized the MMSE suffers from a floor uses a semistructured interview that requires 30 to 40 min-
effect and is relatively insensitive to change in severe AD utes to administer and evaluates cognition (memory, orien-
when scores drop below 10 [15], and it can no longer be tation, judgment/problem solving), community affairs,
used when language is severely affected [1]. The Severe home and hobbies, and personal care [17,18]. Because of
Impairment Battery (SIB) is a reliable and well-validated the time required to administer and the need for a semi-
cognitive assessment instrument that was developed specif- structured interview, it is not suitable for use in primary care
ically for patients with severe dementia [20]. It examines a and clinical practice. The GDS is a seven-stage instrument
broader range of cognitive functions and is better able to in which stage 6 represents severe and stage 7 represents
grade variations of cognition in patients whose MMSE very severe dementia [16]. The scale provides multiple
score is ⬍15 [21]. Unfortunately, the SIB requires 20 to 30 clinical anchors including cognition, function, and behavior.
minutes to administer, making it impractical for primary It has excellent inter-rater and test/retest reliability [32] and
care settings. A shorter version of the SIB has recently been content validity [33] and correlates significantly with the
developed, although its use in clinical settings requires MMSE [19].
further evaluation [22].
3.2.4. Behavioral assessment
With the caveats noted above, the MMSE is therefore the
The neuropsychiatric symptoms of dementia, also known
recommended cognitive assessment instrument at the
as the behavioral and psychological symptoms of dementia
present time. It should be noted that clinical trials in mod-
(BPSD), become increasingly prevalent and disturbing in
erate to severe AD have incorporated the MMSE, with
severe AD [34]. Agitation and aggressive behaviors in-
benefits being noted in some of these studies [23–26].
crease in prevalence with increased disease severity and
3.2.2. Functional assessment occur in up to two thirds of patients with severe AD.
Because loss of independent function is a hallmark of Aberrant motor behaviors such as restlessness, pacing, and
severe AD, functional scales that are heavily weighted to- wandering are also common and particularly prevalent and
ward instrumental activities of daily living have limited disturbing in residents of long-term care who remain am-
utility. The Alzheimer’s Disease Cooperative Study–Activi- bulatory [1,34,35]. Depression, apathy, anxiety, delusions,
ties of Daily Living Inventory (ADCS-ADL) [27] is a well- and hallucinations remain common in severe AD as well
validated functional assessment scale that has been modified [1]. Monitoring and assessment of BPSD are essential in
for use in moderate to severe AD. The ADCS-ADLsev [28] severe AD because these behaviors not only cause suffering
uses a subset of 19 items from the original version and has for the patient but are also associated with stress, burden,
already been used in clinical trials of moderate to severe and increased rates of depression in the caregiver [36 –38].
388 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

They are associated with more rapid cognitive and func- plies therapeutic activism (ie, avoiding nihilism) and in-
tional decline and increased mortality [39 – 43] and are sig- volving caregivers not only in decision making but also in
nificant risk factors for institutionalization [44,45]. therapeutic maneuvers as well. Although maintenance of
A variety of rating scales for BPSD exist that can be used function, providing maximum comfort, and prolonging life
in both long-term care patients and community-dwelling have been recognized as major goals [57], the latter might
individuals [46,47]. The Neuropsychiatric Inventory (NPI) not necessarily be a goal for end-stage disease. Palliative
[48] measures the frequency and severity of 12 behaviors: and end-of-life care have been reviewed elsewhere [58 – 60]
delusions, hallucinations, agitation, depression/dysphoria, and will be discussed in other guidelines in this series.
anxiety, euphoria/elation, apathy/indifference, disinhibition, Medical management includes treating intercurrent medical
irritability/lability, aberrant motor behaviors, sleep and ap- conditions such as infections, seizures, parkinsonian symp-
petite/eating disturbances. Other useful easily administered toms, and pressure ulcers. The detection of pain can be
scales include Cornell Depression Rating Scale [49] and the challenging, although treatment is essential and evidence
Cohen-Mansfield Agitation Inventory [50]. The use of de- suggests that pain is currently undertreated in these patients
pression rating scales has been shown to increase the sen- [61]. Malnutrition can be treated with nutritional supple-
sitivity of depression diagnosis in nursing home residents ments and extra care during meals [62]. Optimizing hearing
[51], and systematic use of structured rating scales can also and vision can be important [63]. The clinician should
reduce agitated aggressive behaviors in hospitalized demen- ensure caregivers are provided with education and support
tia patients [52]. including linkages to the local Alzheimer Society Chapter.
Home-care and day programs should be used. Finally, help-
3.2.5. Medical assessment
ing caregivers around decisions regarding institutionaliza-
The medical assessments typically recommended by
tion is an important function at this stage of disease.
CPGs for mild to moderate dementia often focus on reveal-
ing potentially reversible causes of dementia. In contrast, 3.4. Disease-specific therapies
the medical assessment of severe dementia should focus on
identifying medical causes of excess morbidity specifically Despite the increased attention, there are remarkably few
associated with this stage of illness. Any acute change in randomized controlled trials (RCTs) that have focused on
cognition, function, or behavior should alert the clinician to cognitive enhancement or disease modification for severe
an intercurrent medical problem causing delirium and AD. This is a concern because data from treatment trials of
should lead to appropriate physical and laboratory exami- mild to moderate AD might not necessarily translate to
nations. The value, if any, of neuroimaging in severe de- severe illness, given emerging data that suggest differences
mentia has not been demonstrated. in neuropathology and neurochemistry at different stages.
Severe AD is often associated with progressive gait in- For example, stage-specific changes in neuritic plaques and
stability, risk of falls, and parkinsonian symptoms. Myoc- neurofibrillary tangles have been documented [64,65] as
lonus and seizures can also occur [1]. Loss of ambulation well as changes in neurotransmitters such as catecholamines
leading to being bedridden is associated with the develop- [66,67], ␥-aminobutyric acid [68], and glutamate [69]. Per-
ment of problems such as contractures and pressure ulcers haps the best example of stage-specific neurotransmitter
[53,54]. Continence needs to be monitored because incon- changes is the cholinergic system, in which cholinergic
tinence is associated with the decision to institutionalize markers, such as choline acetyltransferase, do not decline
[55,56]. Weight and nutritional status should also be mon- significantly until later stages of AD, and milder stages are
itored. Potential problems with constipation should be con- characterized by persevered choline acetyltransferase
sidered. Swallowing difficulties might appear in very severe [70,71]. These stage-specific changes suggest that treat-
dementia, increasing the risk of aspiration, malnutrition, and ments studied in milder stages of AD might not necessarily
dehydration. be effective in severe AD.
3.2.6. Other assessments 3.4.1. Cholinesterase inhibitors
It is essential to assess the safety of the patient and their There are three cholinesterase inhibitors (ChEIs) ap-
caregiver. This requires review of issues such as aggression, proved for use in Canada for the treatment of mild to
wandering, neglect, and abuse. Caregiver stress and health, moderate AD: donepezil, galantamine, and rivastigmine.
both physical and emotional, require ongoing monitoring These agents have demonstrated consistent, significant, but
because they ultimately determine whether and when insti- modest improvements in cognition, global assessment, and
tutionalization is necessary. function in patients with MMSEs that range from 10 to 26
3.3. Management [72]. There is also evidence of benefit on neuropsychiatric
symptoms from these trials [73] and from one trial designed
3.3.1. General management principles specifically with behavior as the primary outcome measure
The goal of management of severe AD is to improve the [74]. It is interesting to note that post hoc analyses of some
quality of life for the patient and caregiver. This goal im- of the mild to moderate AD ChEI trials suggested greater
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 389

benefit in patients with moderate compared with mild illness on ADLs compared with placebo [80]. There was no sig-
[75–77]. nificant benefit noted on behavior in this study. Although
There are four published double-blind randomized pla- the authors argued that their results suggest that even in
cebo-controlled trials of ChEIs that included patients with institutionalized AD patients with severe dementia, donepe-
severe AD. Donepezil was studied for 24 weeks in 290 zil improves well-being and can facilitate caregiving, others
patients with moderate to severe AD whose average base- have suggested that it is still unclear how clinically mean-
line MMSE score was 12, and average baseline SIB scores ingful these results are [81].
were 79 to 80 [23]. Inclusion criteria for this study were Several post hoc analyses of rivastigmine- and galantamine-
MMSE scores 5 to 17, although 50% of enrolled patients treated patients with MMSE scores ⱕ14 have been reported,
scored 5 to 12 on baseline MMSE, and 86% of these were suggesting improved cognition and functional benefits com-
rated as severe dementia on global staging. Treatment with pared with placebo [77,82,83]. Similarly in a randomized con-
donepezil was associated with statistically significant ben- trolled head-to-head study of rivastigmine and donepezil, 43%
efits on cognition (as measured by both SIB and MMSE), of the total 994 patients included had MMSE scores of 10 to 14
global outcome, function, and behavior (as measured by the at baseline [84]. The declines in SIB and MMSE scores were
NPI). Similarly, in a post hoc analysis of the patients in this similar with both drugs and significantly smaller than would be
trial with severe dementia (MMSE 5 to 12, n ⫽ 145), expected from patients in untreated naturalistic studies. None
donepezil treatment demonstrated statistically significant of these studies, however, would have included patients de-
benefits on global, cognition, functional, and behavioral fined as severe by MMSE scores ⬍10. In a 52-week open-label
measures [78]. With respect to neuropsychiatric symptoms, study of 173 nursing home residents with moderately severe
there was a 5.5-point treatment difference in favor of done- AD, the effects of rivastigmine were recorded [85]. Average
pezil (P ⫽ .006), with significant improvement noted in baseline MMSE was 9.2. After 52 weeks the average change
depression/dysphoria, anxiety, and apathy. from baseline was an improvement of 0.1 point, and a dose-
In another randomized double-blind placebo-controlled dependent effect was noted (patients on 12 mg per day im-
24-week trial of donepezil, 208 nursing home residents were proved significantly by 1.4 points; P ⫽ .021). Furthermore,
enrolled, with inclusion criteria that included baseline patients in the study experienced improvement in most behav-
MMSE scores of 5 to 26 and at least one neuropsychiatric iors measured by the NPI, and 40% of patients who had been
symptom on the NPI that occurred several times per week treated with psychotropics for BPSD discontinued or reduced
[24]. The average MMSE score was 14, although only 22% the dosage of their medication by week 52.
to 26% of the sample had severe dementia. The primary In summary, although use of a ChEI in severe AD is
outcome measure, neuropsychiatric symptoms scores on the supported by at least one well-designed RCT, not all results
NPI, was not affected by treatment, although secondary are positive, and more confirmatory studies in patients ex-
measures of ADL function and disease stage improved clusively with severe AD are required.
significantly with donepezil compared with placebo. Suba-
nalysis of the patients with severe AD was not reported. 3.4.2. Memantine
In a randomized placebo-controlled study with exclu- The best studied treatment for severe AD is memantine,
sively severe AD patients, rivastigmine was compared with a noncompetitive N-methyl-D-aspartate receptor antagonist.
quetiapine in 93 patients in a 26-week trial [79]. The inclu- Three RCTs have been published to date. In a 12-week
sion criteria for this relatively small study focused on the randomized double-blind placebo-controlled trial, meman-
presence of agitation, irritability, and/or aberrant motor be- tine was studied in 166 patients with MMSE ⬍10 and GDS
haviors in institutionalized patients. The resultant average 5 to 7 [86]. Average MMSE scores were 6, with 96% of
baseline SIB score was 62, making this one of the more patients being staged as severe or very severe, although only
cognitively impaired patient samples to date. Although about half the samples were diagnosed with AD. Global
there were no statistically significant differences at 6 or 26 outcomes, behavior, and function all improved significantly
weeks between rivastigmine and placebo, placebo-treated with memantine. In a larger 28-week double-blind con-
patients (n ⫽ 19) improved on average by 3.3 points on the trolled trial, 252 patients with moderate to severe AD were
SIB compared with 3.1-point worsening for patients on treated with either memantine or placebo. Inclusion criteria
rivastigmine (n ⫽ 15). Besides the very small sample size, included MMSE scores of 3 to 14 and GDS 5 or 6 [25].
placebo-treated patients had a 10-point average higher SIB Average baseline MMSE score was 8, with 56% staged as
score at baseline compared with the rivastigmine patients, moderate to severe (GDS, 6). Global outcome demonstrated
possibly biasing these results. a trend in favor of memantine (P ⫽ .06), with statistically
Finally, in the most recent double-blind RCT, AD pa- significant benefits noted on function and cognition. These
tients with MMSE scores of 1 to 10 living in nursing homes two studies have been analyzed with a number needed to
were treated with either donepezil or placebo. After 6 treat (NNT) analysis [87]. A responder analysis from the
months of treatment, donepezil-treated patients improved study by Winblad and Poritis [86] was statistically signifi-
statistically more on cognitive measures and declined less cant with NNTs of three and four. In the study by Reisberg
390 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

et al [25], NNT for global improvement or stabilization and 3.4.4. Summary


one secondary outcome (ADL or SIB) was 6 (P ⫽ .004; There is evidence from large well-designed randomized
95% confidence interval, 4 to 12). Interestingly, the num- placebo-controlled trials to recommend the use of ChEIs,
bers needed to harm for adverse events were similar to memantine, and the combination for the treatment of severe
placebo except for agitation, in which placebo-treated pa- AD. Treatment benefits appear to include modest improve-
tients actually had a significantly higher risk. These authors ments in cognition, function, and behavior. None of the
concluded that on the basis of the low NNTs, a medium studies were longer than 6 months, leaving questions about
effect size, and an extremely benign safety profile, meman- duration of benefits. It does appear clear, however, that
tine’s benefit in moderate to severe AD was of similar admission to a long-term care facility does not in itself
magnitude to that of the ChEIs in mild to moderate AD [87]. justify treatment discontinuation. The RCTs reviewed
In a third study, 404 patients with moderate to severe AD above included patients with MMSEs as low as 3 to 5, and
were randomized to either memantine or placebo for 24 although recommendations to discontinue treatment below
weeks [26]. Inclusion criteria included MMSE 5 to 14 and these scores might not be unreasonable at the present, it is
having received a stable dose of donepezil for at least 6 still possible that benefits might persist at this stage. We
months before randomization. Average baseline MMSE in prefer to recommend that treatment persist until clinical
these patients was 10. All outcome measures including benefit can no longer be demonstrated, and/or the patient
cognition (SIB), global function, and behavior (NPI) statis- has reached a very severe stage of AD (GDS, 7).
tically favored memantine. 3.5. Management of BPSD
In a post hoc analysis of the studies by Reisberg et al [25]
and Tariot et al [26], behavioral improvements favored The management of BPSD begins with appropriate as-
memantine in both studies and reached statistical signifi- sessment and diagnosis, the identification of target symp-
cance in the latter [88]. In both studies, agitation/aggression toms, and consideration of the safety of the patient, their
improved significantly with memantine therapy, perhaps caregiver, and others in their environment. After optimizing
suggesting a slightly different profile of behavioral benefits medical conditions [93,94] and concomitant medications
compared with the ChEIs. [95,96] as well as improving sensory impairment [63,97–
In summary, use of memantine in severe AD is supported 99], specific treatment proceeds to nonpharmacologic inter-
by at least one high quality RCT as monotherapy or in ventions such as caregiver education, environmental, and
combination with a ChEI. behavioral approaches. If behaviors persist and are suffi-
ciently disturbing to patient or caregiver or endanger safety,
3.4.3. Selegiline/␣-tocopherol pharmacologic interventions can be instituted with appro-
In a double-blind placebo-controlled trial, 341 moder- priate monitoring of effectiveness and safety.
ately severe AD patients were treated with ␣-tocopherol 3.5.1. Nonpharmacologic interventions
(vitamin E), selegiline, or the combination for up to 2 years Nonpharmacologic interventions include environmental
[89]. Inclusion criteria were based on a CDR of 2 (moder- (eg, wandering tracks, bright light, aromatherapy), behav-
ately severe), with baseline MMSEs averaging 11 to 13. The ioral (eg, differential reinforcement), sensory (eg, music,
primary outcome measure was time to occurrence of any of Snoezelen), physical activity (eg, walking), social contact
death, institutionalization, loss of two or three basic ADLs, (eg, simulated presence), psychotherapeutic (eg, reminis-
or conversion to a CDR stage of 3 (severe dementia). There cence therapy), and caregiver/staff training interventions. A
were no differences in the primary outcomes, although be- number of systematic reviews have critically appraised the
cause of a failed randomization, baseline MMSEs differed evidence for these interventions [100 –103]. Unfortunately,
significantly among the treatment groups. After correcting none of these reviews focused on severe AD, and although
statistically for baseline differences in MMSE, it appeared some of the studies reviewed included patients with severe
there was a statistically significant delay in outcome for AD, most included patients with a variety of dementia
selegiline, ␣-tocopherol, and the combination. There were diagnoses and illness severity. The most comprehensive
no significant differences, however, in cognitive measures. review was recently published by the Old Age Task Force
Because of the need for statistical adjustment and the lack of of the World Federation of Biological Psychiatry [104].
benefit on cognitive measures, treatment with selegiline or After identifying 162 studies that met inclusion criteria,
vitamin E was not recommended in the previous Canadian only nine studies were judged to be of high quality (level 1
Consensus Guidelines [90]. Subsequently, enthusiasm for evidence). These authors concluded that only behavioral
treatment with vitamin E has waned even further because of management, specific types of caregiver and staff education,
a large negative trial of vitamin E in mild cognitive impair- and possibly cognitive stimulation appear to have lasting
ment [91] as well as concerns that doses of vitamin E in benefits. Unfortunately, a closer evaluation of the level I
excess of 400 IU per day are associated with increased studies identified by these authors suggests their application
mortality [92]. to severe AD is questionable. For example, in the highest
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 391

quality study of behavioral treatment of depression in de- patients can be withdrawn from antipsychotics without ex-
mentia [105], average MMSE scores were 16.5, whereas the acerbating behavior [111–114]. Finally, before initiating
highest quality study of cognitive stimulation therapy en- therapy with other forms of pharmacotherapy for BPSD,
rolled patients with average MMSE scores of 14.4 and an clinicians should consider use of the disease-specific treat-
average CDR of 1.4 [106]. Music therapy (no high quality ments, because treatment with ChEIs might improve apathy,
RCTs) and Snoezelen (one high quality RCT) were also depression, anxiety, and psychosis, whereas memantine
rated as potentially useful, although benefits have only been might improve agitation and aggression as noted previously.
documented during treatment sessions and not in the longer
term. Not included in this review was use of light therapy 3.5.2.1. Antipsychotics
and aromatherapy, although these have been examined in Antipsychotics potentially improve delusions, hallucina-
Cochrane Reviews. These reviews concluded that there is tions, aggression, agitation, and sleep disturbance in AD
insufficient evidence to assess the value of light therapy (on patients [115]. Meta-analyses of the RCTs of typical anti-
the basis of five published studies) [107], and more studies psychotics such as haloperidol suggest response rates
were required to assess the effectiveness of aromatherapy ⬎60% with benefits 18% to 26% greater than placebo
(on the basis of two studies) [108]. With respect to aroma- [116,117]. In these studies, however, the efficacy rate was
therapy, a high quality double-blind placebo-controlled trial approximately equivalent to the side effect rate, and the
with Melissa (lemon balm) included institutionalized pa- risks of developing parkinsonism and tardive dyskinesia are
tients with severe dementia only (CDR, 3) [109]. Treatment significant even when treated with low doses of typical
with active aromatherapy significantly improved scores on antipsychotics [118,119]. There is also evidence that links
agitation, aggression, irritability, and aberrant motor behav- the use of typical antipsychotics with increased cognitive
iors, with benefits occurring during the first week of treat- decline in AD [120]. As a result, previous reviews and
ment. Although limitations of the study included the method guidelines have preferentially recommended atypical anti-
of randomization and blinding, perhaps the major limitation psychotics for treatment of agitation, aggression, and psy-
to aromatherapy is the increasing move to scent-free work- chosis [121–124], although most of these recommendations
ing environments, which might make this intervention im- were published before safety warnings regarding the risk of
practical in many long-term care institutions. cerebrovascular adverse events (CVAEs) and mortality. It
Nonpharmacologic interventions clearly require more re- is, therefore, important to briefly review their efficacy and
search particularly for patients with severe AD. In spite of safety before making recommendations about their use.
the lack of high quality rigorous RCTs, organizations such More than 2,000 patients, mostly with severe dementia,
as the American Geriatric Society and the American Asso- were included in the published RCTs of risperidone [125–
ciation for Geriatric Psychiatry have recently recommended 128], olanzapine [129,130], and quetiapine [79]. For exam-
that after the assessment and treatment of associated med- ple, in studies with risperidone, the average MMSE scores
ical conditions, the initial treatment of BPSD in the absence ranged from 5 to 9. The studies with risperidone and olan-
of psychosis or immediate danger to self or others should be zapine demonstrated efficacy greater than placebo, with
nonpharmacologic approaches [110]. reasonable tolerability of doses of risperidone approxi-
mately 1 mg per day and olanzapine 5 to 10 mg per day. In
3.5.2. Pharmacologic interventions the only published RCT of quetiapine [79], treatment with
Before initiation of pharmacotherapy, a patient’s medical 100 mg per day was no more effective than placebo and
status must be reviewed to ensure avoidance of allergies and significantly worsened cognition. This is in contrast with a
drug-drug interactions as well as specific underlying comor- large unpublished trial of quetiapine that suggested efficacy
bidities that might increase the possibility of adverse drug greater than placebo at 200 mg per day and no significant
reactions (eg, parkinsonism). It is important to identify changes in cognitive function [131].
target symptoms that are more likely to respond to pharma- After reviewing data from the published and unpublished
cologic interventions. BPSDs such as agitation, aggression, RCTs of atypical antipsychotics for BPSD, health regula-
psychosis, apathy, and depression are likely to respond to tory agencies including Health Canada issued warnings
pharmacotherapy, whereas wandering, perseverative shout- about increased CVAEs and mortality in drug-treated pa-
ing, and some sexually inappropriate behaviors are unlikely tients relative to placebo-treated patients. In 11 RCTs with
to respond. Pharmacotherapy might be considered first line olanzapine and risperidone, 48 of 2,187 (2.2%) drug-treated
in the presence of severe depression, psychosis, and aggres- patients experienced CVAEs compared with 10 of 1,190
sion [110]. All forms of pharmacotherapy must be moni- (0.8%) placebo-treated patients, resulting in a relative risk
tored for effectiveness and adverse events. If effective and of 2.7 (95% confidence interval, 1.4 to 5.3) [132]. Similarly,
well-tolerated, attempts to taper and discontinue these med- increased mortality was noted with risperidone (4/1,009, 4%
ications should occur in a standardized fashion. Four ran- versus placebo 22/712; 3.1%), quetiapine (20/365; 5.51%
domized double-blind placebo-controlled trials with institu- versus placebo 7/217; 3.2%), and olanzapine (42/1,184;
tionalized severe dementia patients demonstrated that most 3.5% versus placebo 7/478; 1.5%) [133].
392 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

With respect to CVAEs, the increased incidence might with buspirone [147]. Although trazodone was significantly
be accounted for by nonspecific vascular events rather more effective than buspirone and placebo, this study in-
than completed strokes [132], and large observational cluded patients with mild-moderate dementia (GDS aver-
database studies have failed to confirm an increased risk age, 4.4). The second study was a randomized trial com-
of stroke compared with typical antipsychotics and even paring trazodone with haloperidol in moderate to severe
untreated dementia patients [134 –137]. In terms of mor- dementia [148] in which both medications appeared equally
tality data, a meta-analysis of 15 BPSD RCTs compared efficacious. Only one of these trials was a double-blind
3,353 drug-treated patients with 1,757 placebo-treated placebo-controlled parallel group design comparing traz-
patients [138]. There were 118 (3.5%) deaths with atyp- odone, haloperidol, and behavior therapy, which included
ical antipsychotics compared with 40 (2.3%) deaths with patients with average MMSEs of 12 to 14 [149]. Trazodone
placebo and no evidence that risk varied by drug, sever- treatment was no better than placebo, and the most frequent
ity, or diagnosis. Furthermore, in two studies that in- reason for dropouts with trazodone was actually an increase
cluded haloperidol arms, a similar magnitude of risk was in agitation. It is important to note that this was a negative
noted. In contrast, four observational studies have found study, and none of the interventions were significantly bet-
no risk of increased mortality associated with atypical ter than placebo; however, a number of methodologic issues
antipsychotic use for BPSD [139 –142]. have been raised that question the interpretation and con-
In summary, efficacy of the atypical antipsychotics, ris- clusions of this study [150]. Although previous recommen-
peridone and olanzapine, is supported by well-designed dations included the use of trazodone for management of
RCTs, although these data are tempered by increased agitation and sleep disturbance [90], a recent meta-analysis
CVAEs and mortality. Although observational studies fail of the use of trazodone for BPSD concluded that there was
to confirm these risks, these potentially serious adverse insufficient evidence to recommend its use for BPSD at the
events must be considered if their use is necessary. There is present time [151].
no rationale for using typical antipsychotics because their In summary, although evidence from RCTs supports the
use is clearly associated with increased extra-pyramidal use of antidepressants, especially selective serotonin re-
symptoms, probably similar rates of CVAEs and mortality, uptake inhibitors, for the treatment of depression in mild to
as well as worsened cognition. moderate AD, similar studies have not been completed in
patients with severe AD. The use of citalopram for the
3.5.2.2. Antidepressants treatment of agitation appears promising but requires repli-
cation in a larger, longer-term study. At the present time
A number of well-designed RCTs suggest that treatment there is insufficient evidence to recommend for or against
with antidepressants is useful for depression in patients with the use of trazodone for the treatment of BPSD in severe
mild to moderate AD [143]. For example, in one of the most AD.
rigorous studies published to date, AD patients with major
depression were randomized to treatment with sertraline or 3.5.2.3. Anticonvulsants
placebo [144]. Although treatment with sertraline demon-
strated significant overall benefit, inclusion criterion was There have been four published RCTs with carbamaz-
MMSE ⬎10, and baseline MMSE scores were 16.3 to 17.5. epine and four RCTs with valproate for the treatment of
Specific criteria for diagnosis of depression in AD have BPSD in severe dementia. The trials of carbamazepine
been proposed [145], although it is not clear how valid or [152–155] demonstrated significant benefit compared with
reliable they are in patients with severe AD. Clinicians placebo, although concerns about carbamazepine’s tolera-
bility and potential for drug interactions have limited its use.
should avoid the use of tricyclic antidepressants as a result
None of the valproate studies [156 –159] demonstrated ben-
of anticholinergic effects and concerns about worsening
efit on primary outcome measures compared with placebo,
cognition.
and higher doses are associated with unacceptable side
Antidepressants have also been used for the treatment of
effects, such as excessive sedation [160].
agitation in AD. In a small randomized double-blind con-
In summary, there are RCT data to support the use of
trolled trial, 85 patients with average MMSEs of 6.4 to 9.9
carbamazepine for agitation and aggression, although side
were randomized to citalopram, perphenazine, or placebo
effects and drug interactions probably relegate its use to a
[146]. Only citalopram-treated patients experienced statis-
second line agent. There is no evidence to support the use of
tically more improvement compared with placebo, with
valproate for BPSD.
benefits noted on agitation, aggression, psychosis, lability/
tension, and retardation.
3.5.2.4. Other pharmacologic approaches
Finally, there have been three RCTs published that used
trazodone for the treatment of agitation and aggression There is only one recent RCT examining the use of
[147–149]. One of the studies was a small double-blind benzodiazepines for BPSD, an RCT comparing intramuscu-
placebo-controlled crossover trial comparing trazodone lar lorazepam with olanzapine and placebo during a 24-hour
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 393

period [161]. Both treatments were better than placebo, with [4] Farlow MR. Moderate to severe Alzheimer disease: definition and
no differences in adverse events including sedation. Use of clinical relevance. Neurology 2005;65:S1– 4.
[5] Vellas B, Gauthier S, Allain H, Andrieu S, Aquino JP, Berrut G, et
benzodiazepines should be limited to short-term, prn (as al. Consensus statement on dementia of Alzheimer type in the severe
circumstances may require) use for behavioral emergencies stage. J Nutr Health Aging 2005;9:330 – 8.
or sedation for procedures. Because of their well-known [6] Canadian Study of Health and Aging. Study methods and prevalence
adverse effects including excessive sedation, falls, and cog- of dementia. CMAJ 1994;150:899 –913.
nitive impairment [162], regular, daily use should be [7] Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzhei-
mer disease in the US population: prevalence estimates using the
avoided. There are a number of small RCTs with variable
2000 census. Arch Neurol 2003;60:1119 –22.
results investigating the use of beta-blockers, such as pro- [8] Larson EB, Shadlen MF, Wang L, McCormick WC, Bowen JD, Teri
pranolol and pindolol [163–166]. Other RCTs for BPSD L, et al. Survival after initial diagnosis of Alzheimer disease. Ann
have included buspirone [147,167], conjugated estrogens Intern Med 2004;140:501–9.
[168 –170], dronabinol [171], long-acting opioids [172], and [9] Hux MJ, O’Brien BJ, Iskedjian M, Goeree R, Gagnon M, Gauthier
melatonin [173,174]. There is insufficient evidence to sup- S. Relation between severity of Alzheimer’s disease and costs of
caring. CMAJ 1998;159:457– 65.
port the use of any of these interventions at the present time. [10] Wimo A, Ljunggren G, Winblad B. Costs of dementia and dementia
care: a review. Int J Geriatr Psychiatry 1997;12:841–56.
4. Conclusions [11] Woolf SH, Battista RN, Anderson GM, Logan AG, Wang E. As-
sessing the clinical effectiveness of preventive maneuvers: analytic
Severe AD represents a huge societal burden as a result principles and systematic methods in reviewing evidence and de-
of its prevalence, costs, and the suffering incurred by pa- veloping clinical practice recommendations—a report by the Cana-
dian Task Force on the Periodic Health Examination. J Clin Epide-
tients and their caregivers. This review has emphasized the miol 1990;43:891–905.
increased attention this phase of illness has received re- [12] Folstein MF, Folstein SE, McHugh PR. ”Mini-mental state”: a
cently, including the recognition that it requires specific practical method for grading the cognitive state of patients for the
CPGs. The recommended interventions have the potential to clinician. J Psychiatr Res 1975;12:189 –98.
improve the quality of life for both patient and caregiver and [13] Reisberg B, Sclan G, Franssen EH, Klugger A, Ferris S. Dementia
staging in chronic care populations. Alzheimer Disease Related
are supported by a reasonable amount of RCT data. More
Disorders 1944;8:S188 –205.
research is still required to operationalize the definition of [14] Ferris SH, Yan B. Differential diagnosis and clinical assessment of
severe AD and develop clinically useful measures of cog- patients with severe Alzheimer disease. Alzheimer Dis Assoc Disord
nition. More RCTs are required to elaborate the use of 2003;17(Suppl 3):S92–5.
ChEIs and memantine and better define the length of useful [15] Feldman HH, Woodward M. The staging and assessment of mod-
therapy as well as the greatest severity of illness in which erate to severe Alzheimer disease. Neurology 2005;65:S10 –7.
[16] Reisberg B, Ferris SH, de Leon MJ, Crook T. The Global Deterio-
benefit can be elucidated. More effective nonpharmacologic ration Scale for assessment of primary degenerative dementia. Am J
approaches must be developed and examined with rigorous Psychiatry 1982;139:1136 –9.
RCTs. Safer and effective pharmacotherapy for BPSD must [17] Berg L. Clinical Dementia Rating (CDR). Psychopharmacol Bull
also be developed. Ultimately, effective disease-modifying 1988;24:637–9.
therapy for mild AD should hopefully preclude the need to [18] Morris JC. Clinical dementia rating: a reliable and valid diagnostic
and staging measure for dementia of the Alzheimer type. Int Psy-
treat this phase of illness.
chogeriatr 1997;9(Suppl 1):173– 8.
[19] Solomon PR, Adams FA, Groccia ME, DeVeaux R, Growdon JH,
Author Disclosures Pendlebury WW. Correlational analysis of five commonly used
measures of mental status/functional abilities in patients with Alz-
Nathan Herrmann has received research support, hono- heimer disease. Alzheimer Dis Assoc Disord 1999;13:147–50.
raria, and/or consultant fees from Lundbeck, Janssen Ortho [20] Saxton J, McGonigle-Gibson G, Swihart A, Miller VJ, Boller F.
Assessment of the severely impaired patient: description and vali-
Inc, Pfizer, Novartis, and Eli Lilly. dation of a new neuropsychological test battery. Psychol Assess-
Serge Gauthier has been an investigator and/or consul- ment: J Consulting Clin Psychology 1990;2:2.
tant for Lundbeck, Pfizer, and Merz. [21] Panisset M, Roudier M, Saxton J, Boller F. Severe impairment
Paul G. Lysy has nothing to disclose. battery: a neuropsychological test for severely demented patients.
Arch Neurol 1994;51:41–5.
[22] Saxton J, Kastango KB, Hugonot-Diener L, Boller F, Verny M,
References Sarles CE, et al. Development of a short form of the Severe Impair-
ment Battery. Am J Geriatr Psychiatry 2005;13:999 –1005.
[1] Boller F, Verny M, Hugonot-Diener L, Saxton J. Clinical features [23] Feldman H, Gauthier S, Hecker J, Vellas B, Subbiah P, Whalen E.
and assessment of severe dementia: a review. Eur J Neurol 2002;9: A 24-week, randomized, double-blind study of donepezil in mod-
125–36. erate to severe Alzheimer’s disease. Neurology 2001;57:613–20.
[2] Tariot PN. Medical management of advanced dementia. J Am Geri- [24] Tariot PN, Cummings JL, Katz IR, Mintzer J, Perdomo CA,
atr Soc 2003;51:S305–13. Schwam EM, et al. A randomized, double-blind, placebo-controlled
[3] Voisin T, Reynish E, Portet F, Feldman H, Vellas B. What are the study of the efficacy and safety of donepezil in patients with Alz-
treatment options for patients with severe Alzheimer’s disease? CNS heimer’s disease in the nursing home setting. J Am Geriatr Soc
Drugs 2004;18:575– 83. 2001;49:1590 –9.
394 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

[25] Reisberg B, Doody R, Stoffler A, Schmitt F, Ferris S, Mobius H. [46] Weiner MF, Koss E, Wild KV, Folks DG, Tariot P, Luszczynska H,
Memantine in moderate-to-severe Alzheimer’s disease. N Engl et al. Measures of psychiatric symptoms in Alzheimer patients: a
J Med 2003;348:1333– 41. review. Alzheimer Dis Assoc Disord 1996;10:20 –30.
[26] Tariot PN, Farlow MR, Grossberg GT, Graham SM, McDonald S, [47] Cummings JL. Neuropsychiatric and behavioral alterations and their
Gergel I. Memantine treatment in patients with moderate to severe management in moderate to severe Alzheimer disease. Neurology
Alzheimer disease already receiving donepezil: a randomized con- 2005;65:S18 –24.
trolled trial. JAMA 2004;291:317–24. [48] Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi
[27] Galasko D, Bennett D, Sano M, Ernesto C, Thomas R, Grundman DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive
M, et al. An inventory to assess activities of daily living for clinical assessment of psychopathology in dementia. Neurology 1994;44:
trials in Alzheimer’s disease: the Alzheimer’s Disease Cooperative 2308 –14.
Study. Alzheimer Dis Assoc Disord 1997;11(Suppl 2): S33–9. [49] Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell
[28] Galansko DR, Schmitt FA, Jin S. Detailed assessment of cognition Scale for Depression in Dementia. Biol Psychiatry 1988;23:271– 84.
and activities of daily living in moderate to severe Alzheimer’s [50] Cohen-Mansfield J, Billig N. Agitated behaviors in the elderly. I: a
disease. Neurobiol Aging 2000;21:S168. conceptual review. J Am Geriatr Soc 1986;34:711–21.
[29] Gelinas I, Gauthier L, McIntyre M, Gauthier S. Development of a [51] Teresi J, Abrams R, Holmes D, Ramirez M, Eimicke J. Prevalence
functional measure for persons with Alzheimer’s disease: the dis- of depression and depression recognition in nursing homes. Soc
ability assessment for dementia. Am J Occup Ther 1999;53:471– 81. Psychiatry Psychiatr Epidemiol 2001;36:613–20.
[30] Katz S, Akpom CA. 12: Index of ADL. Med Care 1976;14:116 – 8. [52] Nilsson K, Palmstierna T, Wistedt B. Aggressive behavior in hos-
[31] Cohen-Mansfield J, Reisberg B, Bonnema J, Berg L, Dastoor DP, pitalized psychogeriatric patients. Acta Psychiatr Scand 1988;78:
Pfeffer RI, et al. Staging methods for the assessment of dementia: 172–5.
perspectives. J Clin Psychiatry 1996;57:190 – 8. [53] Souren LE, Franssen EH, Reisberg B. Contractures and loss of
[32] Reisberg B, Franssen EH, Bobinski M, Auer S, Monteiro I, Boksay function in patients with Alzheimer’s disease. J Am Geriatr Soc
I, et al. Overview of methodologic issues for pharmacologic trials in 1995;43:650 –5.
mild, moderate, and severe Alzheimer’s disease. Int Psychogeriatr [54] Auer SR, Sclan SG, Yaffee RA, Reisberg B. The neglected half of
1996;8:159 –93. Alzheimer disease: cognitive and functional concomitants of severe
[33] Overall JE, Scott J, Rhoades HM, Lesser J. Empirical scaling of the dementia. J Am Geriatr Soc 1994;42:1266 –72.
stages of cognitive decline in senile dementia. J Geriatr Psychiatry [55] O’Donnell BF, Drachman DA, Barnes HJ, Peterson KE, Swearer
Neurol 1990;3:212–20. JM, Lew RA. Incontinence and troublesome behaviors predict in-
[34] Lopez OL, Becker JT, Sweet RA, Klunk W, Kaufer DI, Saxton J, et stitutionalization in dementia. J Geriatr Psychiatry Neurol 1992;5:
al. Psychiatric symptoms vary with the severity of dementia in 45–52.
probable Alzheimer’s disease. J Neuropsychiatry Clin Neurosci [56] Thomas P, Ingrand P, Lalloue F, Hazif-Thomas C, Billon R, Vieban
2003;15:346 –53. F, et al. Reasons of informal caregivers for institutionalizing demen-
[35] Wood S, Cummings JL, Hsu MA, Barclay T, Wheatley MV, tia patients previously living at home: the Pixel study. Int J Geriatr
Yarema KT, et al. The use of the neuropsychiatric inventory in Psychiatry 2004;19:127–35.
nursing home residents: characterization and measurement. Am J [57] Gillick M, Berkman S, Cullen L. A patient-centered approach to
Geriatr Psychiatry 2000;8:75– 83. advance medical planning in the nursing home. J Am Geriatr Soc
[36] Clyburn LD, Stones MJ, Hadjistavropoulos T, Tuokko H. Predicting 1999;47:227–30.
caregiver burden and depression in Alzheimer’s disease. J Gerontol [58] Sampson EL, Ritchie CW, Lai R, Raven PW, Blanchard MR. A
B Psychol Sci Soc Sci 2000;55:S2–13. systematic review of the scientific evidence for the efficacy of a
[37] Coen RF, Swanwick GR, O’Boyle CA, Coakley D. Behaviour palliative care approach in advanced dementia. Int Psychogeriatr
disturbance and other predictors of carer burden in Alzheimer’s 2005;17:31– 40.
disease. Int J Geriatr Psychiatry 1997;12:331– 6. [59] Lyness JM. End-of-life care: issues relevant to the geriatric psychi-
[38] Donaldson C, Tarrier N, Burns A. Determinants of carer stress in atrist. Am J Geriatr Psychiatry 2004;12:457–72.
Alzheimer’s disease. Int J Geriatr Psychiatry 1998;13:248 –56. [60] Sachs GA, Shega JW, Cox-Hayley D. Barriers to excellent end-of-
[39] Miller TP, Tinklenberg JR, Brooks JO, 3rd, Fenn HH, Yesavage JA. life care for patients with dementia. J Gen Intern Med 2004;19:
Selected psychiatric symptoms associated with rate of cognitive 1057– 63.
decline in patients with Alzheimer’s disease. J Geriatr Psychiatry [61] Scherder E, Oosterman J, Swaab D, Herr K, Ooms M, Ribbe M, et
Neurol 1993;6:235– 8. al. Recent developments in pain in dementia. BMJ 2005;330:461– 4.
[40] Moritz DJ, Fox PJ, Luscombe FA, Kraemer HC. Neurological and [62] Magri F, Borza A, del Vecchio S, Chytiris S, Cuzzoni G, Busconi L,
psychiatric predictors of mortality in patients with Alzheimer dis- et al. Nutritional assessment of demented patients: a descriptive
ease in California. Arch Neurol 1997;54:878 – 85. study. Aging Clin Exp Res 2003;15:148 –53.
[41] Walsh JS, Welch HG, Larson EB. Survival of outpatients with [63] Allen NH, Burns A, Newton V, Hickson F, Ramsden R, Rogers J, et
Alzheimer-type dementia. Ann Intern Med 1990;113:429 –34. al. The effects of improving hearing in dementia. Age Ageing
[42] Lopez OL, Wisniewski SR, Becker JT, Boller F, DeKosky ST. 2003;32:189 –93.
Psychiatric medication and abnormal behavior as predictors of pro- [64] Tiraboschi P, Hansen LA, Thal LJ, Corey-Bloom J. The importance
gression in probable Alzheimer disease. Arch Neurol 1999; of neuritic plaques and tangles to the development and evolution of
56:1266 –72. AD. Neurology 2004;62:1984 –9.
[43] Mortimer JA, Ebbitt B, Jun SP, Finch MD. Predictors of cognitive [65] Di Patre PL, Read SL, Cummings JL, Tomiyasu U, Vartavarian LM,
and functional progression in patients with probable Alzheimer’s Secor DL, et al. Progression of clinical deterioration and patholog-
disease. Neurology 1992;42:1689 –96. ical changes in patients with Alzheimer disease evaluated at biopsy
[44] Ryden MB. Aggressive behavior in persons with dementia who live and autopsy. Arch Neurol 1999;56:1254 – 61.
in the community. Alzheimer Dis Assoc Disord 1988;2:342–55. [66] Lanctot KL, Herrmann N, Mazzotta P. Role of serotonin in the
[45] Chenoweth B, Spencer B. Dementia: the experience of family care- behavioral and psychological symptoms of dementia. J Neuropsy-
givers. Gerontologist 1986;26:267–72. chiatry Clin Neurosci 2001;13:5–21.
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 395

[67] Herrmann N, Lanctot KL, Khan LR. The role of norepinephrine in severe Alzheimer’s disease over a 2-year period. Curr Med Res Opin
the behavioral and psychological symptoms of dementia. J Neuro- 2005;21:1317–27.
psychiatry Clin Neurosci 2004;16:261–76. [85] Edwards K, Koumaras B, Chen M, Gunay I, Mirski D. Long-term
[68] Lanctot KL, Herrmann N, Mazzotta P, Khan LR, Ingber N. effects of rivastigmine treatment on the need for psychotropic med-
GABAergic function in Alzheimer’s disease: evidence for dysfunc- ications in nursing home patients with Alzheimer’s disease. Clin
tion and potential as a therapeutic target for the treatment of behav- Drug Invest 2005;25:507–15.
ioural and psychological symptoms of dementia. Can J Psychiatry [86] Winblad B, Poritis N. Memantine in severe dementia: results of the
2004;49:439 –53. 9M-Best Study (benefit and efficacy in severely demented patients
[69] Cacabelos R, Takeda M, Winblad B. The glutamatergic system and during treatment with memantine). Int J Geriatr Psychiatry 1999;14:
neurodegeneration in dementia: preventive strategies in Alzheimer’s 135– 46.
disease. Int J Geriatr Psychiatry 1999;14:3– 47. [87] Livingston G, Katona C. The place of memantine in the treatment of
[70] Davis KL, Mohs RC, Marin D, Purohit DP, Perl DP, Lantz M, et al. Alzheimer’s disease: a number needed to treat analysis. Int J Geriatr
Cholinergic markers in elderly patients with early signs of Alzhei- Psychiatry 2004;19:919 –25.
mer disease. JAMA 1999;281:1401– 6. [88] Gauthier S, Wirth Y, Mobius HJ. Effects of memantine on behav-
[71] DeKosky ST, Ikonomovic MD, Styren SD, Beckett L, Wisniewski ioural symptoms in Alzheimer’s disease patients: an analysis of the
S, Bennett DA, et al. Upregulation of choline acetyltransferase Neuropsychiatric Inventory (NPI) data of two randomised, con-
activity in hippocampus and frontal cortex of elderly subjects with trolled studies. Int J Geriatr Psychiatry 2005;20:459 – 64.
mild cognitive impairment. Ann Neurol 2002;51:145–55. [89] Sano M, Ernesto C, Thomas RG, Klauber MR, Schafer K, Grund-
[72] Lanctot KL, Herrmann N, Yau KK, Khan LR, Liu BA, LouLou man M, et al. A controlled trial of selegiline, alpha-tocopherol, or
MM, et al. Efficacy and safety of cholinesterase inhibitors in Alz- both as treatment for Alzheimer’s disease: the Alzheimer’s Disease
heimer’s disease: a meta-analysis. CMAJ 2003;169:557– 64. Cooperative Study. N Engl J Med 1997;336:1216 –22.
[73] Trinh NH, Hoblyn J, Mohanty S, Yaffe K. Efficacy of cholinesterase [90] Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW,
inhibitors in the treatment of neuropsychiatric symptoms and func- Feldman H, et al. The recognition, assessment and management of
tional impairment in Alzheimer disease: a meta-analysis. JAMA dementing disorders: conclusions from the Canadian Consensus
2003;289:210 – 6. Conference on Dementia. CMAJ 1999;160:S1–15.
[74] Holmes C, Wilkinson D, Dean C, Vethanayagam S, Olivieri S,
[91] Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R,
Langley A, et al. The efficacy of donepezil in the treatment of
Ferris S, et al. Vitamin E and donepezil for the treatment of mild
neuropsychiatric symptoms in Alzheimer disease. Neurology 2004;
cognitive impairment. N Engl J Med 2005;352:2379 – 88.
63:214 –9.
[92] Miller ER, 3rd, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel
[75] Schneider LS, Anand R, Farlow MR. Systematic review of the
LJ, Guallar E. Meta-analysis: high-dosage vitamin E supplementa-
efficacy of rivastigmine for patients with Alzheimer’s disease. In-
tion may increase all-cause mortality. Ann Intern Med 2005;142:
ternational Journal of Geriatric Psychopharmacology 1998;1:S26 –
37– 46.
34.
[93] Kiely DK, Morris JN, Algase DL. Resident characteristics associ-
[76] Wilkinson DG, Hock C, Farlow M, van Baelen B, Schwalen S.
ated with wandering in nursing homes. Int J Geriatr Psychiatry
Galantamine provides broad benefits in patients with ’advanced
2000;15:1013–20.
moderate’ Alzheimer’s disease (MMSE ⬍ or ⫽ 12) for up to six
[94] Cohen-Mansfield J, Billig N, Lipson S, Rosenthan AS, Pawlson LG.
months. Int J Clin Pract 2002;56:509 –14.
Medical correlates of agitation in nursing home residents. Geron-
[77] Blesa R, Davidson M, Kurz A, Reichman W, van Baelen B,
tology 1990;36:150 – 8.
Schwalen S. Galantamine provides sustained benefits in patients
with ‘advanced moderate’ Alzheimer’s disease for at least 12 [95] Learoyd BM. Psychotropic drugs and the elderly patient. Med J Aust
months. Dement Geriatr Cogn Disord 2003;15:79 – 87. 1972;1:1131–3.
[78] Feldman H, Gauthier S, Hecker J, Vellas B, Xu Y, Leni JR, et al. [96] Lanctot KL, Bowles SK, Herrmann N, Best TS, Naranjo CA. Drugs
Efficacy and safety of donepezil in patients with more severe Alz- mimicking dementia: dementia symptoms associated with psycho-
heimer’s disease: a subgroup analysis from a randomized, placebo- tropic drugs in institutionalised cognitively impaired patients. CNS
controlled trial. Int J Geriatr Psychiatry 2005;20:559 – 69. Drugs 2000;14:381–90.
[79] Ballard C, Margallo-Lana M, Juszczak E, Douglas S, Swann A, [97] Corbin SL, Eastwood MR. Sensory deficits and mental disorders of
Thomas A, et al. Quetiapine and rivastigmine and cognitive decline old age: causal or coincidental associations? Psychol Med 1986;16:
in Alzheimer’s disease: randomised double blind placebo controlled 251– 6.
trial. BMJ 2005;330:874. [98] Horowitz A. The relationship between vision impairment and the
[80] Winblad B, Kilander L, Eriksson S, Minthon L, Batsman S, Wet- assessment of disruptive behaviors among nursing home residents.
terholm AL, et al. Donepezil in patients with severe Alzheimer’s Gerontologist 1997;37:620 – 8.
disease: double-blind, parallel-group, placebo-controlled study. [99] Chapman FM, Dickinson J, McKeith I, Ballard C. Association
Lancet 2006;367:1057– 65. among visual hallucinations, visual acuity, and specific eye pathol-
[81] Hogan DB. Donepezil for severe Alzheimer’s disease. Lancet 2006; ogies in Alzheimer’s disease: treatment implications. Am J Psychi-
367:1031–2. atry 1999;156:1983–5.
[82] Doraiswamy PM, Krishnan KR, Anand R, Sohn H, Danyluk J, [100] Cohen-Mansfield J. Nonpharmacologic interventions for inappropri-
Hartman RD, et al. Long-term effects of rivastigmine in moderately ate behaviors in dementia: a review, summary, and critique. Am J
severe Alzheimer’s disease: does early initiation of therapy offer Geriatr Psychiatry 2001;9:361– 81.
sustained benefits? Prog Neuropsychopharmacol Biol Psychiatry [101] Snowden M, Sato K, Roy-Byrne P. Assessment and treatment of
2002;26:705–12. nursing home residents with depression or behavioral symptoms
[83] Burns A, Spiegel R, Quarg P. Efficacy of rivastigmine in subjects associated with dementia: a review of the literature. J Am Geriatr
with moderately severe Alzheimer’s disease. Int J Geriatr Psychiatry Soc 2003;51:1305–17.
2004;19:243–9. [102] Opie J, Rosewarne R, O’Connor DW. The efficacy of psychosocial
[84] Bullock R, Touchon J, Bergman H, Gambina G, He Y, Rapatz G, et approaches to behaviour disorders in dementia: a systematic litera-
al. Rivastigmine and donepezil treatment in moderate to moderately- ture review. Aust N Z J Psychiatry 1999;33:789 –99.
396 N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397

[103] Verkaik R, van Weert JC, Francke AL. The effects of psychosocial [121] Alexopoulos GS, Silver JM, Kahn DA, Frances A, Carpenter D. The
methods on depressed, aggressive and apathetic behaviors of people expert consensus guideline series: treatment of agitation in older
with dementia: a systematic review. Int J Geriatr Psychiatry 2005; persons with dementia. Postgrad Med 1998;26:1– 88.
20:301–14. [122] Herrmann N. Recommendations for the management of behavioral
[104] Livingston G, Johnston K, Katona C, Paton J, Lyketsos CG. Sys- and psychological symptoms of dementia. Can J Neurol Sci 2001;
tematic review of psychological approaches to the management of 28(Suppl 1): S96 –107.
neuropsychiatric symptoms of dementia. Am J Psychiatry 2005;162: [123] Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neu-
1996 –2021. ropsychiatric symptoms of dementia: a review of the evidence.
[105] Teri L. Behavioral treatment of dementia: an overview. Semin Clin JAMA 2005;293:596 – 608.
Neuropsychiatry 1997;2:100 –1. [124] Alexopoulos GS, Streim J, Carpenter D, Docherty JP. Using anti-
[106] Spector A, Thorgrimsen L, Woods B, Royan L, Davies S, Butter- psychotic agents in older patients. J Clin Psychiatry 2004;65(Suppl
worth M, et al. Efficacy of an evidence-based cognitive stimulation 2): 5–104.
therapy programme for people with dementia: randomised con- [125] Katz IR, Jeste DV, Mintzer JE, Clyde C, Napolitano J, Brecher M.
trolled trial. Br J Psychiatry 2003;183:248 –54.
Comparison of risperidone and placebo for psychosis and behavioral
[107] Forbes D, Morgan DG, Bangma J, Peacock S, Pelletier N, Adamson
disturbances associated with dementia: a randomized, double-blind
J. Light therapy for managing sleep, behaviour, and mood distur-
trial—Risperidone Study Group. J Clin Psychiatry 1999;60:107–15.
bances in dementia. Cochrane Database Syst Rev 2005;2:
[126] De Deyn PP, Rabheru K, Rasmussen A, Bocksberger JP, Dautzen-
CD003946.
berg PL, Eriksson S, et al. A randomized trial of risperidone,
[108] Thorgrimsen L, Spector A, Wiles A, Orrell M. Aroma therapy for
placebo, and haloperidol for behavioral symptoms of dementia.
dementia. Cochrane Database Syst Rev 2003;CD003150.
[109] Ballard CG, O’Brien JT, Reichelt K, Perry EK. Aromatherapy as a Neurology 1999;53:946 –55.
safe and effective treatment for the management of agitation in [127] Chan WC, Lam LC, Choy CN, Leung VP, Li SW, Chiu HF. A
severe dementia: the results of a double-blind, placebo-controlled double-blind randomised comparison of risperidone and haloperidol in
trial with Melissa. J Clin Psychiatry 2002;63:553– 8. the treatment of behavioural and psychological symptoms in Chinese
[110] Consensus statement on improving the quality of mental health care dementia patients. Int J Geriatr Psychiatry 2001;16:1156 – 62.
in U.S. nursing homes: management of depression and behavioral [128] Brodaty H, Ames D, Snowdon J, Woodward M, Kirwan J, Clarnette
symptoms associated with dementia. J Am Geriatr Soc 2003;51: R, et al. A randomized placebo-controlled trial of risperidone for the
1287–98. treatment of aggression, agitation, and psychosis of dementia. J Clin
[111] Cohen-Mansfield J, Lipson S, Werner P, Billig N, Taylor L, Woos- Psychiatry 2003;64:134 – 43.
ley R. Withdrawal of haloperidol, thioridazine, and lorazepam in the [129] Street JS, Clark WS, Gannon KS, Cummings JL, Bymaster FP,
nursing home: a controlled, double-blind study. Arch Intern Med Tamura RN, et al. Olanzapine treatment of psychotic and behavioral
1999;159:1733– 40. symptoms in patients with Alzheimer disease in nursing care facil-
[112] van Reekum R, Clarke D, Conn D, Herrmann N, Eryavec G, Cohen ities: a double-blind, randomized, placebo-controlled trial—the
T, et al. A randomized, placebo-controlled trial of the discontinua- HGEU Study Group. Arch Gen Psychiatry 2000;57:968 –76.
tion of long-term antipsychotics in dementia. Int Psychogeriatr [130] De Deyn PP, Carrasco MM, Deberdt W, Jeandel C, Hay DP, Feld-
2002;14:197–210. man PD, et al. Olanzapine versus placebo in the treatment of psy-
[113] Ballard CG, Thomas A, Fossey J, Lee L, Jacoby R, Lana MM, et al. chosis with or without associated behavioral disturbances in patients
A 3-month, randomized, placebo-controlled, neuroleptic discontin- with Alzheimer’s disease. Int J Geriatr Psychiatry 2004;19:115–26.
uation study in 100 people with dementia: the neuropsychiatric [131] Zhong KX, Tariot PN, Mintzer J, Minkwitz MC, Devine NA.
inventory median cutoff is a predictor of clinical outcome. J Clin Quetiapine to treat agitation in dementia: a randomized, double-
Psychiatry 2004;65:114 –9. blind, placebo-controlled study. Curr Alzheimer Res 2007;4:81–93.
[114] Ruths S, Straand J, Nygaard HA, Bjorvatn B, Pallesen S. Effect of [132] Herrmann N, Lanctot KL. Do atypical antipsychotics cause stroke?
antipsychotic withdrawal on behavior and sleep/wake activity in CNS Drugs 2005;19:91–103.
nursing home residents with dementia: a randomized, placebo-con- [133] Health Canada. Health Canada endorsed important safety informa-
trolled, double-blinded study: the Bergen District Nursing Home tion on atypical antipsychotic drugs and dementia. Available at:
Study. J Am Geriatr Soc 2004;52:1737– 43.
http://www.hc-sc.gc.ca/dhp-mps/medeff/advisories-avis/prof/atyp-
[115] Rabinowitz J, Katz IR, De Deyn PP, Brodaty H, Greenspan A,
antipsycho_hpc_e.html. Accessed May 12, 2005.
Davidson M. Behavioral and psychological symptoms in patients
[134] Liperoti R, Gambassi G, Lapane KL, Chiang C, Pedone C, Mor V,
with dementia as a target for pharmacotherapy with risperidone.
et al. Cerebrovascular events among elderly nursing home patients
J Clin Psychiatry 2004;65:1329 –34.
treated with conventional or atypical antipsychotics. J Clin Psychi-
[116] Schneider LS, Pollock VELyness SA. A metaanalysis of controlled
atry 2005;66:1090 – 6.
trials of neuroleptic treatment in dementia. J Am Geriatr Soc 1990;
38:553– 63. [135] Layton D, Harris S, Wilton LV, Shakir SA. Comparison of incidence
[117] Lanctot KL, Best TS, Mittmann N, Liu BA, Oh PI, Einarson TR, et rates of cerebrovascular accidents and transient ischaemic attacks in
al. Efficacy and safety of neuroleptics in behavioral disorders asso- observational cohort studies of patients prescribed risperidone, quetia-
ciated with dementia. J Clin Psychiatry 1998;59:550 – 63. pine or olanzapine in general practice in England including patients
[118] Sweet RA. Neuroleptic-induced parkinsonism in elderly patients with dementia. J Psychopharmacol 2005;19:473– 82.
diagnosed with psychotic major depression and dementia of the [136] Herrmann N, Mamdani M, Lanctot KL. Atypical antipsychotics and risk of
Alzheimer type. Am J Geriatr Psychiatry 1996;4:311–9. cerebrovascular accidents. Am J Psychiatry 2004;161:1113–5.
[119] Caligiuri MP, Rockwell E, Jeste DV. Extrapyramidal side effects in [137] Gill SS, Rochon PA, Herrmann N, Lee PE, Sykora K, Gunraj N, et
patients with Alzheimer’s disease treated with low-dose neuroleptic al. Atypical antipsychotic drugs and risk of ischaemic stroke: pop-
medication. Am J Geriatr Psychiatry 1998;6:75– 82. ulation based retrospective cohort study. BMJ 2005;330:445.
[120] McShane R, Keene J, Gedling K, Fairburn C, Jacoby R, Hope T. Do [138] Schneider LS, Dagerman K, SInsel P. Risk of death with atypical
neuroleptic drugs hasten cognitive decline in dementia? prospective antipsychotic drug treatment for dementia: meta-analysis of random-
study with necropsy follow up. BMJ 1997;314:266 –70. ized placebo-controlled trials. JAMA 2005;294:1934 – 43.
N. Herrmann et al. / Alzheimer’s & Dementia 3 (2007) 385–397 397

[139] Raivio MM, Laurila JV, Strandberg TE, Tilvis RS, Pitkala KH. [158] Tariot PN, Schneider LS, Mintzer JE, Cutler AJ. Safety and toler-
Psychotropic medication and stroke outcome. Am J Psychiatry ability of divalproex sodium in treatment of signs and symptoms of
2005;162:1027– 8. mania in elderly patients with dementia: results of a double-blind
[140] Nasrallah HA, White T, Nasrallah AT. Lower mortality in geriatric placebo-controlled trial. Curr Ther Res Clin Exp 2001;62:51– 67.
patients receiving risperidone and olanzapine versus haloperidol: [159] Tariot PN, Raman R, Jakimovich L, Schneider L, Porteinsson A,
preliminary analysis of retrospective data. Am J Geriatr Psychiatry Thomas R, et al. Divalproex sodium in nursing home residents with
2004;12:437–9. possible or probable Alzheimer disease complicated by agitation: a
[141] Suh GH, Shah A. Effect of antipsychotics on mortality in elderly randomized, controlled trial. Am J Geriatr Psychiatry 2005;
patients with dementia: a 1-year prospective study in a nursing 13:942–9.
home. Int Psychogeriatr 2005;17:429 – 41. [160] Lonergan ET, Luxenberg J. Valproate preparations for agitation in
[142] Wang PS, Schneeweiss S, Avorn J, Fischer MA, Mogun H, Solomon dementia. Cochrane Database Syst Rev 2005;3.
DH, et al. Risk of death in elderly users of conventional vs. atypical [161] Meehan KM, Wang H, David SR, Nisivoccia JR, Jones B, Beasley
antipsychotic medications. N Engl J Med 2005;353:2335– 41. SM, Jr, et al. Comparison of rapidly acting intramuscular olanzap-
[143] Lyketsos CG, Olin J. Depression in Alzheimer’s disease: overview ine, lorazepam, and placebo: a double-blind, randomized study in
and treatment. Biol Psychiatry 2002;52:243–52. acutely agitated patients with dementia. Neuropsychopharmacology
[144] Lyketsos CG, DelCampo L, Steinberg M, Miles Q, Steele CD, 2002;26:494 –504.
Munro C, et al. Treating depression in Alzheimer disease: efficacy [162] Stern RG, Duffelmeyer ME, Zemishlani Z, Davidson M. The use of
and safety of sertraline therapy, and the benefits of depression benzodiazepines in the management of behavioral symptoms in
reduction: the DIADS. Arch Gen Psychiatry 2003;60:737– 46. demented patients. Psychiatr Clin North Am 1991;14:375– 84.
[145] Olin JT, Schneider LS, Katz IR, Meyers BS, Alexopoulos GS, [163] Greendyke RM, Kanter DR, Schuster DB, Verstreate S, Wootton J.
Breitner JC, et al. Provisional diagnostic criteria for depression of Propranolol treatment of assaultive patients with organic brain dis-
Alzheimer disease. Am J Geriatr Psychiatry 2002;10:125– 8. ease: a double-blind crossover, placebo-controlled study. J Nerv
[146] Pollock BG, Mulsant BH, Rosen J, Sweet RA, Mazumdar S, Bha- Ment Dis 1986;174:290 – 4.
rucha A, et al. Comparison of citalopram, perphenazine, and placebo [164] Greendyke RM, Kanter DR. Therapeutic effects of pindolol on
for the acute treatment of psychosis and behavioral disturbances in behavioral disturbances associated with organic brain disease: a
hospitalized, demented patients. Am J Psychiatry 2002;159:460 –5. double-blind study. J Clin Psychiatry 1986;47:423– 6.
[147] Lawlor BA, Radcliffe J, Molchan SE, Martinez RA, Hill JL, Sun- [165] Herrmann N, Lanctot KL, Eryavec G, Khan LR. Noradrenergic
derland T. A pilot placebo-controlled study of trazodone and buspi- activity is associated with response to pindolol in aggressive Alz-
rone in Alzheimer’s disease. Int J Geriatr Psychiatry 1994;9:55–9. heimer’s disease patients. J Psychopharmacol 2004;18:215–20.
[148] Sultzer DL, Gray KF, Gunay I, Berisford MA, Mahler ME. A [166] Peskind ER, Tsuang DW, Bonner LT, Pascualy M, Riekse RG,
double-blind comparison of trazodone and haloperidol for treatment Snowden MB, et al. Propranolol for disruptive behaviors in nursing
of agitation in patients with dementia. Am J Geriatr Psychiatry home residents with probable or possible Alzheimer disease: a
1997;5:60 –9. placebo-controlled study. Alzheimer Dis Assoc Disord 2005;19:
[149] Teri L, Logsdon RG, Peskind E, Raskind M, Weiner MF, Tracten- 23– 8.
berg RE, et al. Treatment of agitation in AD: a randomized, placebo- [167] Cantillon M, Brunswick R, Molina D, Bahro M. Buspirone vs.
controlled clinical trial. Neurology 2000;55:1271– 8. haloperidol: a double-blind trial for agitation in nursing home pop-
[150] Herrmann N, Black SE. Behavioral disturbances in dementia: will ulation with Alzheimer’s disease. Am J Geriatr Psychiatry 1996;4:
the real treatment please stand up? Neurology 2000;55:1247– 8. 263–7.
[151] Martinon-Torres G, Fioravanti M, Grimley Evans J. Trazodone for [168] Kyomen HH, Hennen J, Gottlieb GL, Wei JY. Estrogen therapy and
agitation in dementia. Cochrane Database Syst Rev 2004; noncognitive psychiatric signs and symptoms in elderly patients
CD004990. with dementia. Am J Psychiatry 2002;159:1225–7.
[152] Tariot PN, Erb R, Leibovici A, Podgorski CA, Cox C, Asnis J, et al. [169] Kyomen HH, Satlin A, Hennen J, Wei JY. Estrogen therapy and
Carbamazepine treatment of agitation in nursing home patients with aggressive behavior in elderly patients with moderate-to-severe de-
dementia: a preliminary study. J Am Geriatr Soc 1994;42:1160 – 6. mentia: results from a short-term, randomized, double-blind trial.
[153] Cooney C, Mortimer A, Smith A, Newton K, Wrigley M. Carbam- Am J Geriatr Psychiatry 1999;7:339 – 48.
azepine use in aggressive behaviour associated with senile dementia. [170] Hall KA, Keks NA, O’Connor DW. Transdermal estrogen patches
Int J Geriatr Psychiatry 1996;11:901–5. for aggressive behavior in male patients with dementia: a random-
[154] Tariot PN, Erb R, Podgorski CA, Cox C, Patel S, Jakimovich L, et ized, controlled trial. Int Psychogeriatr 2005;17:165–78.
al. Efficacy and tolerability of carbamazepine for agitation and [171] Volicer L, Stelly M, Morris J, McLaughlin J, Volicer BJ. Effects of
aggression in dementia. Am J Psychiatry 1998;155:54 – 61. dronabinol on anorexia and disturbed behavior in patients with
[155] Olin JT, Fox LS, Pawluczyk S, Taggart NA, Schneider L. A pilot Alzheimer’s disease. Int J Geriatr Psychiatry 1997;12:913–9.
randomized trial of carbamazepine for behavioral symptoms in treat- [172] Manfredi PL, Breuer B, Wallenstein S, Stegmann M, Bottomley G,
ment-resistant outpatients with Alzheimer disease. Am J Geriatr Libow L. Opioid treatment for agitation in patients with advanced
Psychiatry 2001;9:400 –5. dementia. Int J Geriatr Psychiatry 2003;18:700 –5.
[156] Porsteinsson AP, Tariot PN, Erb R, Cox C, Smith E, Jakimovich L, [173] Serfaty M, Kennell-Webb S, Warner J, Blizard R, Raven P. Double
et al. Placebo-controlled study of divalproex sodium for agitation in blind randomised placebo controlled trial of low dose melatonin for
dementia. Am J Geriatr Psychiatry 2001;9:58 – 66. sleep disorders in dementia. Int J Geriatr Psychiatry 2002;17:
[157] Sival RC, Haffmans PM, Jansen PA, Duursma SA, Eikelenboom P. 1120 –7.
Sodium valproate in the treatment of aggressive behavior in patients [174] Singer C, Tractenberg RE, Kaye J, Schafer K, Gamst A, Grundman
with dementia: a randomized placebo controlled clinical trial. Int J M, et al. A multicenter, placebo-controlled trial of melatonin for
Geriatr Psychiatry 2002;17:579 – 85. sleep disturbance in Alzheimer’s disease. Sleep 2003;26:893–901.
Alzheimer’s & Dementia 3 (2007) 398 – 403

Management of dementia with a cerebrovascular component


Christian Boctia,*, Sandra Blackb, Chris Frankc
a
Division of Neurology, Department of Medicine, Université de Montréal, Montréal, Quebec, Canada
b
Division of Neurology, Department of Medicine, University of Toronto, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
c
Program for Care of the Elderly, Department of Medicine, Queen’s University Providence Continuing Care Center, Kingston, Ontario, Canada

Abstract Background: Cerebrovascular disease (CVD) has been reconceptualized as an important factor in
cognitive decline and dementia. The concept of vascular cognitive impairment (VCI) has been
introduced to capture the whole spectrum of cognitive decline associated with CVD, and to
emphasize the possibility of early intervention on vascular risk factors.
Methods: Intervention studies in prevention and symptomatic therapy of dementia associated with
CVD are reviewed.
Results: We present recommendations based on the review of current literature. There is some
evidence that treating hypertension can prevent cognitive decline. Galantamine has shown some
benefit in the symptomatic treatment of possible Alzheimer’s disease with CVD and donepezil has
shown some benefits in vascular dementia. Other pharmacologic interventions lack sufficient
empirical support.
Conclusions: A more pro-active stance towards cognitive decline of vascular etiology is warranted,
with some evidence supporting both prevention and symptomatic therapy.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Vascular dementia; Vascular cognitive impairment; Alzheimer’s disease; Guidelines; Prevention; Therapy

1. Introduction cholinesterase inhibitors have been carried out in dementia


associated with cerebrovascular disease [15–17].
Since the last Canadian Consensus Conference on De-
mentia [1], cerebrovascular disease has been reconceptual-
ized as a major factor in cognitive decline and dementia in 2. Definitions
the elderly [2– 4]. Some of the most important concepts are
(1) virtually all traditional risk factors for cerebrovascular These recent concepts are just beginning to generate new
disease also are risk factors for Alzheimer’s disease [5,6]; approaches not only to vascular dementia (VaD) but also to
(2) subcortical ischemic lesions such as lacunar infarcts cognitive decline at any degree of severity, including the
greatly influence the clinical syndrome of dementia [7–9]; “brain-at-risk” and predementia stages in which the most
and (3) the most common pathologic substrate of dementia efficient preventive strategies could be implemented [18].
in population-based autopsy series is combined cerebrovas- The concept of vascular cognitive impairment (VCI) has
cular disease and Alzheimer’s pathology [10,11]. In addi- been introduced to emphasize the high prevalence of cog-
tion to these epidemiologic and pathologic interactions, nitive impairment with a vascular component and foster a
some evidence supports a cholinergic deficit in vascular more proactive stance toward cognitive decline. VCI en-
dementia [12], presumably through vascular damage to the compasses all degrees of severity of cognitive deficits re-
cholinergic pathways [13,14]. Clinical trials with two of the sulting from vascular disease: vascular cognitive impair-
ment, no dementia (V-CIND); VaD, which is itself
heterogeneous; and mixed Alzheimer’s disease and cerebro-
vascular disease (AD-CVD) [4,19,20]. An alternative um-
*Corresponding author. Tel.: 514-252-3528; Fax: 514-252-3529. brella term to VCI has been proposed to further clarify the
E-mail address: cbocti@gmail.com same concept, vascular cognitive disorders [21]. Here the
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.009
C. Bocti et al. / Alzheimer’s & Dementia 3 (2007) 398 – 403 399

term VCI is equivalent to V-CIND or vascular mild cogni- 3. Methods


tive impairment, and the other two main subtypes remain
The current review considered all randomized controlled
VaD and AD-CVD. In both of these schemes, the inclusion
trials of interventions to improve VaD. A preliminary list of
of the mildest form of cognitive impairment will hopefully
potential articles was obtained through an exhaustive En-
enable both patients and health professionals to optimize
glish language literature review on PubMed and Embase
preventive strategies before the stage of VaD is attained. with the terms “Vascular dementia OR Multi-infarct demen-
Many of the propositions just presented have been for- tia OR Vascular cognitive impairment OR Vascular cogni-
mulated in response to the consensus-derived National In- tive disorders OR Subcortical Ischemic Vascular Dementia”
stitute of Neurological Disorders and Stroke–Association In- and “management OR treatment OR prevention OR cholin-
ternationale pour la Recherche et l’Enseignement (NINDS– ergic agents”. The list was reduced to 29 articles on the
AIREN) VaD criteria [22]. These criteria are the most basis of methodologic quality and non-overlap with other
commonly used and include three concepts: (1) evidence of reports. We included here only those that dealt with drugs
dementia, (2) evidence of vascular disease of the brain, and easily available in Canada.
(3) evidence of a temporal link between (1) and (2). They A standardized analysis of the quality of the evidence
have been used successfully in clinical trials to identify was undertaken, integrating a rating of the quality of the
patients who evolve differently than AD patients [16,17]. report with the Jadad scale [34] and such quantitative as-
However, many critiques are well-founded and justify the pects as statistical significance and magnitude of effects
current effort to better define the spectrum of VaD. One (naturalstandard.com) [35].
fundamental limitation is that the definition is modeled on The following categories will be utilized to grade the
the cognitive profile of AD, in which memory is promi- levels of evidence.
nently affected. The most common cognitive deficit of VaD,
(I) Evidence obtained from at least one properly ran-
in contrast, is frontal/executive dysfunction [23,24]. Most
domized controlled trial.
clinical tools developed for AD do not capture this type of (II-1) Evidence obtained from well-designed controlled tri-
cognitive deficit [25], with the consequence that the contri- als without randomization.
bution of vascular disease is often clinically unrecognized. (II-2) Evidence obtained from well-designed cohort or case
A possible solution to that problem is the recent publication control analytic studies preferably from more than
of a neuropsychology protocol that is specifically aimed at one center or research group.
screening patients with VCI, including a 5-minute battery (II-3) Evidence obtained from comparisons between times
with a real potential for widespread clinical use [26]. An- or places with or without the intervention. Dramatic
other limitation of the VaD criteria comes from the imaging results in uncontrolled experiments are included in
criteria; they are quite complex to apply [27] and do not this category.
appear to be specific in unselected stroke populations [28]. (III) Opinions of respected authorities based on clinical
There is a need for prospective, detailed information about experience, descriptive studies, or reports of expert
the clinical, cognitive, imaging, and functional aspects of committees [36].
VaD to develop new criteria. There are no universally ac-
cepted criteria for V-CIND. The criteria derived from the Recommendations were graded as follows:
Canadian Study of Health and Aging have been shown to (A) There is good evidence to support this maneuver.
have important clinical implications; rates of death or insti- (B) There is fair evidence to support this maneuver.
tutionalization were similar to those of AD subjects after 5 (C) There is insufficient evidence to recommend for or
years [29]. Other criteria have been proposed by Italian against this maneuver, but recommendations might
investigators for subcortical vascular MCI, and the clinical be made on other grounds.
outcomes have similarly been far from benign [30]. These (D) There is a fair evidence to recommend against this
criteria are modified from research criteria for subcortical procedure.
vascular dementia proposed in 2000, defining a more ho- (E) There is good evidence to recommend against this
mogeneous subgroup of patients within the VaD category procedure.
[31]. A major strength of these criteria is the adequate
emphasis on executive dysfunction as the major cognitive
domain affected. As for the AD-CVD category or mixed 4. Nonpharmacologic interventions
dementia, there is an important need for more elaborate and Few studies have been published on this topic. A
valid criteria. The original AD criteria provide the “possible Cochrane database review included six randomized con-
AD with cerebrovascular disease” category [32], but the trolled trials of cognitive training in dementia [37]. Only
relative contribution of each pathologic process is a chal- one of these articles included patients with vascular demen-
lenge to establish in most cases [33]. tias [38]. No benefit was reported in any of the main out-
400 C. Bocti et al. / Alzheimer’s & Dementia 3 (2007) 398 – 403

comes for any of these studies (cognitive, global, functional, rates were low, resulting in very wide confidence intervals,
and behavioral outcomes). and a meta-analysis of pooled data from the four aforemen-
tioned trials failed to reach significance [48].
4.1. Recommendation 1
6.1. Recommendation 3
4.1.1. Use of nonpharmacologic interventions
There is currently insufficient evidence to recommend 6.1.1. Investigations for vascular risk factors
the use of cognitive training for vascular dementia (Grade It is recommended that vascular risk factors are identified
C, Level 2). in all patients with vascular cognitive impairment (Grade C,
Level 3).
5. Pharmacologic therapy in vascular dementia 6.2. Recommendation 4

5.1. Aspirin 6.2.1. Treating hypertension


There is some evidence that treating hypertension might
Although the use of aspirin for prevention of recurrent prevent further cognitive decline associated with cerebro-
stroke is supported by considerable evidence [39], its use in vascular disease. There is no compelling evidence that one
vascular dementia is not. According to a Canadian survey, class of agent is superior to another; calcium channel block-
the clinical practice of prescribing aspirin as an antiplatelet ers or angiotensin-converting enzyme inhibitors might be
in the context of VaD is very common (86% of respondents) considered (Grade B, Level 1).
[40]. There are no clinical trials supporting potential bene- Treatment for hypertension should be implemented for
fits of this intervention [41]. This information needs to be other reasons, including the prevention of recurrent stroke
weighted against the potential risk of hemorrhage, which (Grade A, Level 1).
might be increased in some patients under consideration here;
cerebral microbleeds as detected by gradient-echo magnetic
resonance imaging are very prevalent in subjects with subcor- 7. Symptomatic treatment
tical ischemic damage. Microbleeds could be a marker of Several molecules have been the object of a controlled
increased hemorrhagic risk, although a recent review failed to trial in symptomatic treatment of VaD. Nimodipine, me-
find compelling evidence to modify clinical practice [42]. mantine, donepezil, and galantamine all had randomized
5.2. Recommendation 2 controlled trials supporting some benefits in VaD.
7.1. Nimodipine
5.2.1. Antiplatelet therapy with aspirin
There is currently no evidence to support the use of Several trials have been conducted with nimodipine in
aspirin to specifically treat dementia associated with cere- both degenerative and vascular dementias. A recent
brovascular disease (Grade C, Level 3). Aspirin or other Cochrane review included a total of 14 trials of nimodipine,
antiplatelet therapies should be used for prevention of recur- of which seven reported sufficient data to be included in the
rent ischemic stroke in appropriate patients [43] (Grade A, meta-analysis [49]. The total number of subjects included
Level 1). was 2,492, the typical trial duration was 12 weeks, and the
dosage of nimodipine was 90 mg/day. Results favored treat-
6. Blood pressure reduction ment over placebo for global scales and cognitive outcomes
in treatment completers with “cerebrovascular dementia”.
In recent years there has been increasing interest in blood The drug was well-tolerated, with remarkably low dropout
pressure lowering as a means to prevent dementia or protect rates in most studies. The benefits were of small magnitude,
against further cognitive decline. The evidence supporting and no benefit could be demonstrated for behavioral or
such a practice is included here. Because hypertension is a functional outcomes in any of the trials. The strength of the
major risk factor for stroke and stroke can be associated conclusions is undermined by the lack of data for fully half
with dementia, it is biologically plausible that reducing of the identified reports.
hypertension will reduce dementia and cognitive decline.
7.2. Recommendation 5
Although there are promising results from two of four large
randomized controlled trials of antihypertensive drugs 7.2.1. Nimodipine in vascular dementia
(Syst-Eur and Progress) [44,45], two others failed to show a There is insufficient evidence for or against the use of
benefit for cognitive outcomes in the treatment arm (SHEP, nimodipine for VaD (Grade C, Level 1).
SCOPE) [46,47]. Methodologic issues limit the conclusions
that can be drawn specifically for the prevention of demen- 8. Cholinesterase inhibitors
tia. Although numbers of subjects were large in the two
positive trials, cognitive testing was relatively crude, and One large-scale (n ⫽ 592), high quality (Jadad scale ⫽ 5)
primary end points were noncognitive. In addition, event trial with galantamine in a mixed population of patients with
C. Bocti et al. / Alzheimer’s & Dementia 3 (2007) 398 – 403 401

probable VaD (n ⫽ 252) and possible AD with CVD (n ⫽ There is fair evidence of benefits of small magnitude for
295) showed overall positive results on cognitive, func- donepezil in cognitive and global outcomes, with less robust
tional, and behavioral measures. The effect size was small benefits on functional measures. Donepezil can be considered
(0.3 for cognitive outcome), commensurate with that ob- a treatment option for vascular dementia (Grade B, Level 1).
served in trials of AD. Dropout rates as a result of gastro-
intestinal adverse events were significantly higher with the
titration dosage used in that study. The overall effect was 9. Memantine
driven by the AD-CVD group, because the results did not
reach statistical significance for the probable VaD group. Memantine, an N-methyl-D-aspartate receptor antago-
This trial was not powered to draw conclusions on the nist, has been the object of two trials in vascular dementia
probable VaD subgroup [14]. with a total sample of 900 patients [52,53], and a further
Two identical large-scale double-blind randomized con- trial included a subgroup of VaD patients, but the results are
trolled trials (Jadad scale ⫽ 5) have been conducted with not reported separately [54]. The quality of the two reports
donepezil in vascular dementia, with a total sample size of reviewed here varies from good to excellent (Jadad scale, 4
1,219 subjects [16,17]. Outcomes have been statistically and 5). Primary outcomes have been favorable to treatment
significant for cognition in both trials, with small effect size only for cognitive measures in both trials, with small effect
(Alzheimer’s Disease Assessment Scale, cognitive portion size in both (0.22). The global scales failed to capture any
and Mini-Mental State Examination). For global scales and significant benefit (Clinical Global Impression of Change,
functional outcomes, there were inconsistencies between CIBIC-plus). The drug was well-tolerated.
both dosages used, 5 mg or 10 mg daily. There was benefit
on the Clinician’s Interview-Based Impression of Change 9.1. Recommendation 8
(CIBIC)-plus for both dosages. On another measure, the 9.1.1. Use of memantine
Clinical Dementia Rating-Sum of Boxes, the lower 5-mg There is some evidence of small magnitude of cognitive
dosage did not produce significant results, but the10-mg benefit that is not captured in global measures for patients
dose did. The same situation prevailed for the functional with VaD. There is insufficient information to recommend
outcome, the Alzheimer’s Disease Functional Assessment memantine for the treatment of vascular dementia (Grade C,
and Change Scale. The subscore for Instrumental Activities Level 1).
of Daily Living, however, was significant for both doses.
Tolerability was apparently better for the lower dosage, with a
differential dropout rate for the 10-mg dose. It was suggested
that the lower dosage might be preferable in this population. Author Disclosures
There is no high quality trial for rivastigmine in vas- Christian Bocti has received support from Janssen Ortho
cular dementia. A post hoc analysis that compared AD Inc. (Speaker, Consultant), Novartis (Speaker) and Pfizer
patients with and without vascular risk factors showed a Canada (Speaker).
somewhat increased magnitude of benefit in the first Sandra Black has received support from Eisai (Clinical
group [50]. An open uncontrolled trial of rivastigmine investigation, CME lecturer, Ad hoc consultant), Pfizer
showed benefits of small magnitude in cognitive func- (Clinical investigation, CME lecturer, Ad hoc consultant),
tions and behavior [51]. Janssen-Ortho (Clinical investigation, CME lecturer, Ad
hoc consultant), Novartis (Clinical investigation, CME lec-
8.1. Recommendation 6 turer, Ad hoc consultant), Lundbeck (Ad hoc consultants,
8.1.1. Use of cholinesterase inhibitors in dementia caused CME lecturer), Sanofi-Aventis (Trial investigator), and
by AD-CVD Myriad (Trial investigator, Consultant—Ad hoc).
There is fair evidence of benefits of small magnitude for Chris Frank has nothing to disclose.
galantamine in cognitive, functional, behavioral, and global
measures in AD with CVD. Galantamine can be considered
a treatment option for mixed AD with CVD (Grade B, Acknowledgments
Level 1).
Christian Bocti has been supported by a scholarship from
8.2. Recommendation 7 the Alzheimer Society of Canada, and an operating grant
from Canadian Institutes for Health Research (CIHR).
8.2.1. Use of cholinesterase inhibitors in Sandra Black’s work has been supported by operating
probable/possible vascular dementia with the NINDS- grants from the CIHR, Alzheimer Association US and Alz-
AIREN diagnostic criteria heimer Society of Canada. She receives salary support from
There is insufficient evidence for or against the use of the Department of Medicine, University of Toronto, and
galantamine (Grade C, Level 1). Sunnybrook Research Institute.
402 C. Bocti et al. / Alzheimer’s & Dementia 3 (2007) 398 – 403

References [21] Roman GC, Sachdev P, Royall DR, Bullock RA, Orgogozo JM,
Lopez-Pousa S, et al. Vascular cognitive disorder: a new diagnostic
[1] Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW, category updating vascular cognitive impairment and vascular de-
mentia. J Neurol Sci 200415;226:81–7.
Feldman H, et al. The recognition, assessment and management of
[22] Roman GC, Tatemichi TK, Erkinjuntti T, Cummings JL, Masdeu JC,
dementing disorders: conclusions from the Canadian Consensus Con-
Garcia JH, et al. Vascular dementia: diagnostic criteria for research
ference on Dementia. 1999;160(12 Suppl):S1–15.
studies—report of the NINDS-AIREN International Workshop. Neu-
[2] Bowler JV, Steenhuis R, Hachinski V. Conceptual background to
rology 1993;43:250 – 60.
vascular cognitive impairment. Alzheimer Dis Assoc Disord 1999;
[23] Kertesz A, Clydesdale S. Neuropsychological deficits in vascular
13(Suppl 3): S30 –7.
dementia vs Alzheimer’s disease: frontal lobe deficits prominent in
[3] Roman GC. Vascular dementia may be the most common form of
vascular dementia. Arch Neurol 1994;51:1226 –31.
dementia in the elderly. J Neurol Sci 2002;203-204:7–10.
[24] Sachdev PS, Brodaty H, Valenzuela MJ, Lorentz L, Looi JC, Wen W,
[4] O’Brien JT, Erkinjuntti T, Reisberg B, Roman G, Sawada T, Pantoni
et al. The neuropsychological profile of vascular cognitive impair-
L, et al. Vascular cognitive impairment. Lancet Neurol. 2003;2:89 –
ment in stroke and TIA patients. Neurology 2004;62:912–9.
98.
[25] O’Sullivan M, Morris RG, Markus HS. Brief cognitive assessment for
[5] Decarli C. Vascular factors in dementia: an overview. J Neurol Sci
patients with cerebral small vessel disease. J Neurol Neurosurg Psy-
2004;226:19 –23.
chiatry 2005;76:1140 –5.
[6] Honig LS, Kukull W, Mayeux R. Atherosclerosis and AD: analysis of
[26] Hachinski V, Iadecola C, Petersen RC, Breteler MM, Nyenhuis DL,
data from the US National Alzheimer’s Coordinating Center. Neu-
Black SE, et al. National Institute of Neurological Disorders and
rology 2005;64:494 –500.
Stroke-Canadian Stroke Network vascular cognitive impairment har-
[7] Snowdon DA, Greiner LH, Mortimer JA, Riley KP, Greiner PA,
monization standards. Stroke 2006;37:2220 – 41.
Markesbery WR. Brain infarction and the clinical expression of
[27] van Straaten EC, Scheltens P, Knol DL, van Buchem MA, van Dijk
Alzheimer disease: the Nun Study. JAMA 1997;277:813–7.
EJ, Hofman PA, et al. Operational definitions for the NINDS-AIREN
[8] Esiri MM, Nagy Z, Smith MZ, Barnetson L, Smith AD. Cerebrovas-
criteria for vascular dementia: an interobserver study. Stroke 2003;
cular disease and threshold for dementia in the early stages of Alz-
34:1907–12.
heimer’s disease. Lancet 1999;354:919 –20. [28] Ballard CG, Burton EJ, Barber R, Stephens S, Kenny RA, Kalaria RN,
[9] Petrovitch H, Ross GW, Steinhorn SC, Abbott RD, Markesbery W, et al. NINDS AIREN neuroimaging criteria do not distinguish stroke
Davis D, et al. AD lesions and infarcts in demented and non- patients with and without dementia. Neurology 2004;63:983– 8.
demented Japanese-American men. Ann Neurol 2005;57:98 –103. [29] Rockwood K, Wentzel C, Hachinski V, Hogan DB, MacKnight C,
[10] Lim A, Tsuang D, Kukull W, Nochlin D, Leverenz J, McCormick W, McDowell I. Prevalence and outcomes of vascular cognitive impair-
et al. Clinico-neuropathological correlation of Alzheimer’s disease in ment: Vascular Cognitive Impairment Investigators of the Canadian
a community-based case series. J Am Geriatr Soc 1999;47:564 –9. Study of Health and Aging. Neurology 2000;54:447–51.
[11] Neuropathology Group of the Medical Research Council Cognitive [30] Frisoni GB, Galluzzi S, Bresciani L, Zanetti O, Geroldi C. Mild
Function and Ageing Study (MRC CFAS). Pathological correlates of cognitive impairment with subcortical vascular features: clinical char-
late-onset dementia in a multicentre, community-based population in acteristics and outcome. J Neurol 2002;249:1423–32.
England and Wales. Lancet 2001;357:169 –75. [31] Erkinjuntti T, Inzitari D, Pantoni L, Wallin A, Scheltens P, Rockwood
[12] Tohgi H, Abe T, Kimura M, Saheki M, Takahashi S. Cerebrospinal K, et al. Research criteria for subcortical vascular dementia in clinical
fluid acetylcholine and choline in vascular dementia of Binswanger trials. J Neural Transm Suppl 2000;59:23–30.
and multiple small infarct types as compared with Alzheimer-type [32] McKhann G, Drachman D, Folstein M, Katzman R, Price D, Stadlan
dementia. J Neural Transm 1996;103:1211–20. EM. Clinical diagnosis of Alzheimer’s disease: report of the
[13] Tomimoto H, Ohtani R, Shibata M, Nakamura N, Ihara M. Loss of NINCDS-ADRDA Work Group under the auspices of Department of
cholinergic pathways in vascular dementia of the Binswanger type. Health and Human Services Task Force on Alzheimer’s Disease.
Dement Geriatr Cogn Disord 2005;19:282– 8. Neurology 1984;34:939 – 44.
[14] Bocti C, Swartz RH, Gao FQ, Sahlas DJ, Behl P, Black SE. A new [33] Lopez OL, Kuller LH, Becker JT, Jagust WJ, DeKosky ST, Fitz-
visual rating scale to assess strategic white matter hyperintensities patrick A, et al. Classification of vascular dementia in the Cardiovas-
within cholinergic pathways in dementia. Stroke 2005;36:2126 –31. cular Health Study Cognition Study. Neurology 2005;64:1539 – 47.
[15] Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju [34] Jadad AR, Moore RA, Carroll D, Jenkinson C, Reynolds DJ, Ga-
CV. Efficacy of galantamine in probable vascular dementia and Alz- vaghan DJ, et al. Assessing the quality of reports of randomized
heimer’s disease combined with cerebrovascular disease: a random- clinical trials: is blinding necessary? Control Clin Trials 1996;17:
ised trial. Lancet 2002;359:1283–90. 1–12.
[16] Black S, Roman GC, Geldmacher DS, Salloway S, Hecker J, Burns [35] Available at: http://www.naturalstandard.com/explanation_column-
A, et al. Efficacy and tolerability of donepezil in vascular dementia: s.html. Accessed February 5, 2006.
positive results of a 24-week, multicenter, international, randomized, [36] Woolf SH, Battista RN, Anderson GM, Logan AG, Wang E. Assess-
placebo-controlled clinical trial. Stroke 2003;34:2323–30. ing the clinical effectiveness of preventive maneuvers: analytic prin-
[17] Wilkinson D, Doody R, Helme R, Taubman K, Mintzer J, Kertesz A, ciples and systematic methods in reviewing evidence and developing
et al. Donepezil in vascular dementia: a randomized, placebo- clinical practice recommendations—a report by the Canadian Task
controlled study. Neurology 2003;61:479 – 86. Force on the Periodic Health Examination. J Clin Epidemiol 1990;
[18] Hachinski V. Preventable senility: a call for action against the vas- 43:891–905.
cular dementias. Lancet 1992;340:645– 8. [37] Clare L, Woods RT, Moniz Cook ED, Orrell M, Spector A. Cognitive
[19] Hachinski V. Vascular dementia: a radical redefinition. Dementia. rehabilitation and cognitive training for early-stage Alzheimer’s dis-
1994;5:130 –2. ease and vascular dementia. Cochrane Database Syst Rev 2003(4):
[20] Rockwood K, Davis H, MacKnight C, Vandorpe R, Gauthier S, CD003260.
Guzman A, et al. The Consortium to Investigate Vascular Impairment [38] Quayhagen MP, Quayhagen M, Corbeil RR, Hendrix RC, Jackson JE,
of Cognition: methods and first findings. Can J Neurol Sci 2003;30: Snyder L, et al. Coping with dementia: evaluation of four nonphar-
237– 43. macologic interventions. Int Psychogeriatr 2000;12:249 – 65.
C. Bocti et al. / Alzheimer’s & Dementia 3 (2007) 398 – 403 403

[39] Antiplatelet Trialists’ Collaboration. Collaborative overview of trials learned from the systolic hypertension in the elderly program (SHEP)
of antiplatelet therapy. I: prevention of death, myocardial infarction, trial. Am J Epidemiol 2001;153:72– 8.
and stroke by prolonged antiplatelet therapy in various categories of [47] Lithell H, Hansson L, Skoog I, Elmfeldt D, Hofman A, Olofsson B,
patients. Br Med J 1994;308:81–106. et al. The study on cognition and prognosis in the elderly (SCOPE):
[40] Molnar FJ, Man Song Hing M, St John P, Brymer C, Rockwood K, principal results of a randomized double-blind intervention trial.
Hachinski V. Subcortical vascular dementia: survey of treatment J Hypertens 2003;21:875– 86.
patterns and research considerations. Can J Neurol Sci 1998;25: [48] Feigin V, Ratnasabapathy Y, Anderson C. Does blood pressure low-
320 – 4. ering treatment prevent dementia or cognitive decline in patients with
[41] Williams PS, Spector A, Orrell M, Rands G. Aspirin for vascular cardiovascular and cerebrovascular disease? J Neurol Sci 2005;229-
dementia. Cochrane Database Syst Rev 2000(2):CD001296. 230:151–5.
[42] Koennecke HC. Cerebral microbleeds on MRI: prevalence, associa- [49] Lopez-Arrieta JM, Birks J. Nimodipine for primary degenerative,
tions, and potential clinical implications. Neurology 2006;66:165–71. mixed and vascular dementia. Cochrane Database Syst Rev 2002(3):
[43] Goldstein LB, Adams R, Alberts MJ, Appel LJ, Brass LM, Bushnell CD000147.
CD, et al. Primary prevention of ischemic stroke: a guideline from the [50] Kumar V, Anand R, Messina J, Hartman R, Veach J. An efficacy and
American Heart Association/American Stroke Association Stroke safety analysis of Exelon in Alzheimer’s disease patients with con-
Council: cosponsored by the Atherosclerotic Peripheral Vascular Dis- current vascular risk factors. Eur J Neurol 2000;7:159 – 69.
ease Interdisciplinary Working Group; Cardiovascular Nursing Coun- [51] Moretti R, Torre P, Antonello RM, Cazzato G, Bava A. Rivastigmine
cil; Clinical Cardiology Council; Nutrition, Physical Activity, and in subcortical vascular dementia: a randomized, controlled, open
Metabolism Council; and the Quality of Care and Outcomes Research 12-month study in 208 patients. Am J Alzheimers Dis Other Demen
Interdisciplinary Working Group: the American Academy of Neurol- 2003;18:265–72.
ogy affirms the value of this guideline. Stoke 2006;37:1583– 633. [52] Orgogozo JM, Rigaud AS, Stoffler A, Mobius HJ, Forette F. Efficacy
[44] Forette F, Seux ML, Staessen JA, Thijs L, Birkenhager WH, Babar- and safety of memantine in patients with mild to moderate vascular
skiene MR, et al. Prevention of dementia in randomised double-blind dementia: a randomized, placebo-controlled trial (MMM 300). Stroke
placebo-controlled systolic hypertension in Europe (Syst-Eur) trial. 2002;33:1834 –9.
Lancet 1998;352:1347–51. [53] Wilcock G, Mobius HJ, Stoffler A. A double-blind, placebo-
[45] Tzourio C, Anderson C, Chapman N, Woodward M, Neal B, controlled multicentre study of memantine in mild to moderate vas-
MacMahon S, et al. Effects of blood pressure lowering with cular dementia (MMM500). Int Clin Psychopharmacol 2002;17:297–
perindopril and indapamide therapy on dementia and cognitive 305.
decline in patients with cerebrovascular disease. Arch Intern Med [54] Winblad B, Poritis N. Memantine in severe dementia: results of the
2003;163:1069 –75. 9M-Best Study (Benefit and efficacy in severely demented patients
[46] Di Bari M, Pahor M, Franse LV, Shorr RI, Wan JY, Ferrucci L, et al. during treatment with memantine). Int J Geriatr Psychiatry 1999;14:
Dementia and disability outcomes in large hypertension trials: lessons 135– 46.
Alzheimer’s & Dementia 3 (2007) 404 – 410

Disclosure of the diagnosis of dementia


John D. Fisk *, B. Lynn Beattieb, Martha Donnellyc, Anna Byszewskid, Frank J. Molnare
a,
a
QE II Health Sciences Centre; Departments of Psychiatry, Medicine, and Psychology, Dalhousie University, Halifax, Nova Scotia, Canada
b
Division of Geriatric Medicine, Department of Medicine, UBC, Vancouver, British Columbia, Canada
c
Division of Community Geriatrics, Department of Family Practice, and Division of Geriatric Psychiatry, Department of Psychiatry, UBC; and
Geriatric Psychiatry Outreach Team, Vancouver Hospital, Vancouver, British Columbia, Canada
d
Department of Medicine, University of Ottawa; Geriatric Day Hospital, Ottawa Hospital; and Ottawa Health Research Institute, Ottawa,
Ontario, Canada
e
Division of Geriatric Medicine, Department of Internal Medicine, Ottawa Hospital; Department of Medicine, University of Ottawa; Ottawa Health
Research Institute; and Elisabeth Bruyere Research Institute, Ottawa, Ontario, Canada

Abstract Most ethical guidelines strongly promote disclosure of a diagnosis of dementia to the affected
individual, based on the principle of autonomy. Nevertheless, codes of medical ethics allow for
various interpretations of this issue and surveys of clinical practice illustrate that such disclosure is
by no means the rule. We argue that diagnostic disclosure for persons with dementia must be
considered a process that begins when cognitive impairment is first suspected and that evolves over
time as information is obtained. Whenever possible and appropriate, this process should involve not
only the affected individual but also their family and/or other current or potential future care
providers. Once a diagnosis is established it should be disclosed in a manner consistent with the
expressed wishes of the patient, using an individualized patient-centered approach that maintains
the individual’s personal integrity. Diagnostic disclosure of dementia is a process that may require
additional time as well as follow-up or referral to other specialists. We recommend that a progres-
sive disclosure process be employed to address issues including: remaining diagnostic uncertainty,
treatment options, future plans, financial planning, assigning power of attorney, wills and “living
wills”, driving privileges and the need for eventual driving cessation, available support services, and
potential research participation. The potential for adverse psychological consequences to diagnostic
disclosure must be assessed and these should be addressed through education and support of the
patient and their family/caregivers throughout the diagnostic disclosure process. At present, few data
are available regarding patients’ perspectives on the diagnostic disclosure process and its conse-
quences. This limitation and the apparent discrepancies in physician and caregiver opinions about
the disclosure process, make it incumbent upon health care professionals to evaluate the diagnostic
disclosure process within their practice.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Alzheimer’s disease; Dementia; Diagnosis; Ethics

Although various ethical guidelines for medical care Guidelines [2]. This approach used focus group discussions
exist, all with common underlying principles, few have that included caregivers, individuals with mild Alzheimer’s
specific recommendations regarding dementia. In 1997 the disease, and professionals (eg, physicians, nurses, lawyers,
Alzheimer Society of Canada (ASC) released its Ethical ethicists, and administrators) but also extended this consul-
Guidelines [1] document, which represented the results of a tation process by disseminating the information nationally
2-year consultative process that was based on a “discourse for review and comment [3]. Both the Fairhill Guidelines
ethics” approach previously used in developing the Fairhill and the ASC Ethical Guidelines addressed issues important
in clinical practice and everyday life for persons with Alz-
*Corresponding author. Tel.: 902-473-2581; Fax: 902-473-7166. heimer’s disease, such as diagnostic disclosure, driving
E-mail address: John.Fisk@cdha.nshealth.ca privileges, autonomy in decision making, maintaining qual-
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.008
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410 405

ity of life, appropriate use of restraints, and end-of-life care. general practitioners in the United Kingdom identified com-
The ASC Ethical Guidelines, however, also addressed ge- munication about diagnosis as one of the “main difficulties”
netic testing and participation in research. In 1999 the Ca- experienced by general practitioners when dealing with pa-
nadian Consensus Conference on Dementia (CCCD) ac- tients with dementia [10]. Those who reported greater dif-
knowledged the diversity and scope of ethical issues ficulty with the diagnosis and management of dementia
addressed by the ASC guidelines and chose to focus on were also less likely to endorse open communications strat-
discussion of diagnostic disclosures and driving privileges egies for diagnostic disclosure [10]. This same association
[4]. Revision of the ASC Ethical Guidelines through an- between difficulty in establishing a diagnosis and disclosing
other consultative process was undertaken in 2001–2002 in the diagnosis was also reported in an American sample of
response to increased awareness, earlier diagnosis, the con- primary care physicians [11].
ceptualization of mild cognitive impairment (MCI) [5], and Although guidelines favoring diagnostic disclosure are
the availability of symptomatic treatments. For the current based on the principle of patient autonomy, the arguments
revision of the CCCD, the topics discussed in the original that have typically been put forth against diagnostic disclo-
document were reconsidered with a view to both updating sure are based largely on the principle of non-maleficence
them and adding new relevant issues. First, it was decided (ie, the obligation to avoid harm). Justification for the em-
that because of its complexity, the issue of driving needed to phasis on this latter principle has typically relied on (1) the
be considered as a separate topic with discussions not lim- lack of absolute diagnostic certainty from clinical informa-
ited to that of ethical issues. Second, it was determined that tion, (2) the absence of effective treatments of progressive
the issue of diagnostic disclosure warranted a review in light dementia, (3) the questionable ability of persons with more
of new developments and, in particular, because of the advanced disease to understand the implications of the di-
importance of diagnostic disclosure as the starting point agnosis, and (4) the potential for adverse psychological
from which treatment and management decisions follow. responses to diagnostic disclosure as their justification [12].
For this review the PubMed and Embase databases were Diagnostic certainty, although never 100%, has im-
searched for articles with the keywords “Dementia OR proved considerably through the development of diagnostic
Alzheimer’s disease AND ethics AND diagnosis.” For dis- criteria that now span a range of less prevalent forms of
cussion, preference was given to publications between 1996 dementia in addition to Alzheimer’s disease [13]. These
and 2006. criteria are unlikely to be used routinely outside specialty
As described by Maguire [6], “Clinicians who diagnose referral clinics, and lack of familiarity in their use among
patients with dementia are faced with a number of ethical primary care physicians might contribute to uncertainty in
dilemmas: They must be truthful, yet do no harm; they must dementia diagnosis. Nevertheless, lack of access to a diag-
respect patients’ autonomy, yet consider the concerns of nostic opinion from a specialty clinic and lack of certainty
those who live with and care for those patients.” Most about a suspected diagnosis do not negate the responsibility
ethical guidelines dealing with diagnostic disclosure to of primary care physicians to provide “an open, honest
those with dementia and their family/caregivers, including presentation of information as it is perceived and known”
those recommendations put forth in the previous CCCD, [12]. Moreover, as recommended by the previous CCCD,
strongly promote the disclosure of a diagnosis of dementia the topic of diagnostic uncertainty itself should be discussed
to the affected individual, on the basis of the principle of within the context of diagnostic disclosure (Recommenda-
autonomy [4]. This viewpoint is not universally held, how- tion 21) [4]. In recent years, MCI has become increasingly
ever, and codes of medical ethics in general allow for well-defined to the point where it can be considered as a
various interpretations of the issue of diagnostic disclosure diagnostic entity itself rather than just a risk factor for
[7]. Two relatively recent systematic reviews have been dementia [14], making it increasingly important that the
undertaken by groups in the United States [8] and in the same considerations for diagnostic disclosure apply to this
United Kingdom [9]. Both point out the need for continued condition. However, aside from diagnostic uncertainly, dis-
discussion, research, practice guideline refinement, and ed- closure of a diagnosis of MCI is also complicated by prog-
ucation of professionals and the public about this issue. The nostic uncertainty [15]. Explaining uncertainty to patients
need for a Canadian perspective on these issues is rein- and family members is never a simple matter, as was evi-
forced by the finding that only 4% of the articles discussing dent in a qualitative study of the psychosocial impact of
diagnostic disclosure were published in Canada [8]. disclosing a diagnosis of dementia to patients and their
We have little information about whether available prac- family/caregivers by Smith and Beattie [16]. They found
tice guidelines are known by practitioners and how practi- that family/caregivers’ understanding of the clinician’s un-
tioners’ experiences with dementia influence their accep- certainty about the diagnosis was subject to their precon-
tance and use of such guidelines. A review of studies of ceived ideas about the source of their loved ones’ cognitive
general practitioners’ practices in the United States regard- problems. Such findings illustrate the importance of estab-
ing diagnostic disclosure concluded that approximately half lishing a clear understanding of the perspectives of both the
routinely withhold disclosure to the patient [8]. A survey of patient and their family/caregivers to accurately convey
406 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410

both the diagnostic opinion and the uncertainty that might not absolve them of the responsibility to respect patients’
surround this opinion. autonomy. As described by Marzanski [7], “The truth may
The diagnosis of dementia presents a framework within be neither fully understood nor remembered by the patient
which target symptoms and symptomatic treatment options and difficult for the doctor, but neither of these problems
can be discussed. Although the absence of effective disease- should remove the obligation to be honest and truthful”.
modifying treatments had previously been raised as a po- Although the availability of clinical diagnostic criteria
tential argument against diagnostic disclosure, such argu- for improved early and differential diagnosis, as well as
ments were rejected in the ASC Ethical Guidelines [1] even symptomatic treatments for Alzheimer’s disease, has ad-
before the widespread availability of cholinesterase inhibi- dressed some of the arguments against diagnostic disclo-
tors for the treatment of Alzheimer’s disease. Smith and sure, the potential for adverse psychological consequences
Beattie [16] noted that “disclosure facilitated patient care remains an important consideration and is frequently cited
for all families,” and it is important that care not be equated by general practitioners, among others, as the rationale for
with pharmacologic treatment alone. Even in the absence of failing to disclose a diagnosis [19]. However, it is important
approved symptomatic pharmacologic treatments for a to note that concerns about adverse psychological reactions
given diagnosis, treatment of specific symptoms of demen- are most often raised by family/caregivers rather than the
tia might involve either pharmacologic or behavioral inter- patients themselves [20]. Surveys of nonimpaired elderly,
ventions. Prevention strategies for future problems can be faced with the hypothetical situation of having dementia,
discussed meaningfully with patients and their family/care- have consistently found that the majority indicate that they
givers once a diagnosis has been disclosed. Even though would want to be told of their diagnosis [21,22]. This was
approved pharmacologic treatments for Alzheimer’s disease found to be true even for family/caregivers who themselves
remain symptomatic and treatments for other forms of de- did not want their impaired family member to be told of the
mentias are limited, diagnostic disclosure can open a dia- diagnosis of dementia [20]. Despite the views favoring
logue among the patient, their family/caregivers, and health diagnostic disclosure to patients that have been expressed in
care professionals about a broad range of important issues. most guidelines, including the previous CCCD [4], surveys
These include, but are not limited to, financial planning, of patients and their family/caregivers have typically found
assigning power of attorney, wills and “living wills”, driv- that family/caregivers are told of the diagnosis more often
ing privileges, research participation, and the availability than the affected individual [23] and that family/caregivers
and access to formal and informal community services. frequently indicate that they do not want the affected indi-
Without an effective process of diagnostic disclosure, this vidual to be told [24]. If one respects the principle of the
dialogue is not possible. autonomy of the individual, however, decisions regarding
The previous CCCD cited “severe dementia where un- diagnostic disclosure should be based on an accurate ap-
derstanding of the diagnosis is unlikely” as a possible ex- praisal of the patient’s prior wishes rather than the caregiv-
ception to diagnostic disclosure to the patient (Recommen- er’s current wishes.
dation 21) [4]. Although this has historically been described Among the possible negative consequences of diagnostic
throughout discussions of diagnostic disclosure, one hopes disclosure, the potential for suicide is often cited as a reason
that a diagnosis will usually be made long before a severe to consider withholding the diagnosis [4]. Suicide among
stage is reached. Regardless, for some persons with demen- persons with dementia appears to be rare, however, with
tia, profound lack of insight is an early feature and is not a reports limited to only a few cases of individuals with
reflection of severe disease [17]. Although it can be argued relatively early stage Alzheimer’s disease who were de-
that the principle of autonomy no longer applies to persons scribed as having preserved insight into their disabilities
who are incapable of understanding what is being disclosed [25,26]. Varied approaches to this issue have so far failed to
to them, obtaining a clear understanding of the level of establish an association between increased suicide risk and
insight of persons with dementia is challenging [18]. More- the disclosure of a diagnosis of dementia. A study of family
over, an inability to fully appreciate the implications of a members attending a community dementia support group
diagnosis need not preclude the possibility of deriving any reported that suicidal thoughts were expressed by only two
benefit from diagnostic disclosure. The opinion expressed of 28 patients who were told of their diagnosis, and that
more than a decade ago that the individual patient has a neither had acted on these [23]. Another study found less
“moral and legal right . . . to receive a specific diagnosis than a 1% prevalence of suicidal thoughts in a sample of
unless he or she waives it” [2] asserts the principle of 221 demented patients as well as an association between
autonomy in ethical decision making and challenges clini- clinical depression and a stated “wish to die” that did not
cians to base their decisions on more than personal conve- appear different from that seen in general population sur-
nience and their presumptions about the patients’ potential veys of the elderly [27]. A survey of nondemented patients
to benefit from knowledge of their diagnosis. The difficulty attending an outpatient clinic found only three of 182 re-
faced by clinicians in disclosing a diagnosis, and in dealing spondents to a questionnaire about diagnostic disclosure
with the psychosocial consequences of this disclosure, does indicated that they would consider suicide if told of a
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410 407

diagnosis of Alzheimer’s disease [28]. Finally, the findings issue arising from the ASC guidelines [1] include ensuring
of a retrospective study of autopsy cases suggest that it that sufficient time is available for the disclosure process
might be those individuals with Alzheimer’s disease who and considering the possibility of follow-up sessions to
are undiagnosed who are at greater risk for committing address unresolved issues. Lack of follow-up appointments
suicide than the general population [29]. To date, there and limited discussion of information about community-
remains limited data regarding the adverse psychological based services and treatment options were indeed noted as
reactions to receiving a diagnosis of dementia. The need for sources of dissatisfaction in the diagnostic disclosure pro-
better understanding of the psychological consequences of a cess for family/caregivers in a qualitative study conducted
disclosure of a diagnosis of dementia is clear as is the need at U.S. Alzheimer’s Disease Centers [35]. Providing ade-
to constantly update such knowledge as new developments quate time for the detailed discussion of assessment results
in diagnosis and treatment occur. To fully understand the helps ensure that patients and family members appreciate
psychological consequences of diagnostic disclosure, we that the diagnostic process has been thorough, and even if
must consider not just the short-term consequences; in the diagnostic opinions are discrepant from their own, they
which shock, anger, denial, and depression might be more seem more likely to accept them as valid [16]. Providing
common, but also the long-term consequences in which the written general educational materials has also been recom-
benefits of disclosure on psychological well-being might mended [30], although details that are specific for the indi-
become more evident [8]. vidual might be helpful as well. For example, one study
The issue is not one of whether to disclose the diag- found that regardless of the severity or type of dementia,
nosis of dementia or not but rather how and when to do patients generally had a positive response to reading their
so [30]. The consensus opinion of the previous CCCD [4] unedited clinic notes [36].
and subsequent commentaries [31] favors an individual- Early diagnosis might be the key to resolving much of
ized approach to diagnostic disclosure that is sensitive to the debate surrounding the ethics of diagnostic disclosure.
each patient’s unique situation and that also involves Early diagnosis clearly limits arguments against disclosure
family/caregivers in the process. Although it might well that are based on a presumed failure to understand the
be the case that those who more fully appreciate the diagnosis, its implications, or its uncertainty. Because care-
implications of the diagnosis are the family/caregivers of givers seem more likely to express preferences for lack of
the patient, a patient-centered approach to diagnostic diagnostic disclosure when the patient is more severely
disclosure might help family/caregivers appreciate that affected, it seems likely that family/caregivers will be more
their loved one is respected as an individual, regardless of supportive of diagnostic disclosure to the patient if the
their medical condition. The process of diagnostic dis- diagnosis is made earlier [24,35]. However, important dis-
closure might itself serve as an opportunity to appropri- crepancies between physicians’ and family/caregivers’
ately model communication strategies for the patient and opinions about the disclosure process have also been de-
their family/caregivers. However, despite suggestions as scribed. Connell et al [35] found that although physicians
to how meetings with patients and their family/caregivers often believed that the disclosure process had gone well;
might take place, there remains no research demonstrat- many family/caregivers reported that the disclosure was too
ing the relative advantages of individual versus joint direct and insensitive. They concluded that family/caregiv-
disclosure to patients and their family/caregivers. ers “would have liked to have the diagnosis disclosed in a
The increasingly multicultural nature of North America compassionate manner and to include the patient in the
and the absence of research that identifies how ethnicity or office visit in a manner that preserves their dignity and a
belief in cultural idioms might affect the process of disclo- sense of hope” [35].
sure of the diagnosis of dementia also pose challenges for Much as establishing the diagnosis of dementia requires
clinicians. Studies suggest that ethnic minority seniors a process, so too does diagnostic disclosure. Disclosure in a
might be less familiar than Anglo-European Americans with manner that is compassionate for the patient and their fam-
the biomedical model of disease, instead normalizing de- ily/caregiver will vary with individual circumstances, but
mentia as part of the aging process [32,33]. However, in one the need to provide information about the diagnosis along
study, fully 54% of caregivers, including 41% of Anglo- with information about what needs to be done next cannot
European Americans, believed in mixed models of causa- be communicated effectively in a single office appointment.
tion that combined folk explanations and biomedical Follow-up sessions must be mandatory, although this can
elements [34]. take a variety of forms, potentially including meetings with
In the previous CCCD [4], Recommendation 21 stated the clinician responsible for the diagnostic investigations in
that the diagnosis of dementia should be disclosed to the concert with the family physician. This longitudinal process
patient and to their family, and that in addition to diagnostic of disclosure seems crucial for effective communication and
uncertainty, the discussions should include the topics of education of the patient and their family/caregivers.
prognosis, advance planning, treatment options, support Establishment of guidelines for diagnostic disclosure of
groups, and future plans. Other recommendations on this dementia is made difficult by the lack of methodologically
408 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410

adequate systematic study of this issue. Most quantitative individualized approach to disclosure is recom-
studies to date have focused on attitudes and practice rather mended to take into account variations in the needs
than the outcomes of disclosure [9]. Few studies have ex- of the patient and family/caregivers that reflect the
amined the perspectives of patients themselves; instead they nature and severity of the patient’s cognitive prob-
focus on those of physicians and family/caregivers [8,9]. A lems, their background knowledge and expectations,
recent qualitative study of the perspectives of patients with and the cultural factors that can influence the under-
dementia and their caregivers’ attending a Canadian day hos- standing and acceptance of the diagnosis. The pres-
pital program supported full disclosure involving a gradual ence of comorbid medical and mental health issues
process [37,38]. This study highlighted the need to provide that might influence the diagnostic disclosure pro-
hope in the face of a difficult diagnosis and both patients and cess should also be considered. These factors might
caregivers expressed the wish for detailed information and require that additional time, follow-up appointments,
referral to community supports. Though limited, the body of or referral to other specialists be used. Those pro-
literature relevant to this topic allows for some recommenda- viding the diagnosis must demonstrate empathy and
tions for clinical practice as a guide for health care profession- respect and instill a sense of hope. Although some
als’ behavior during the diagnostic disclosure process. patients prefer a very direct approach, others require
a more gentle explanation of their condition. The
progressive disclosure process should also be used to
1. Summary address remaining diagnostic uncertainty, treatment
options, future plans, financial planning, assigning
(1) Diagnostic disclosure must be recognized as a pro- power of attorney, wills and “living wills”, driving
gressive process that begins as soon as the possibility privileges, available support services, and potential
of cognitive impairment is suspected and that in- research participation if appropriate. If physicians
volves education and discussion throughout the in- lack knowledge of these issues, they must be pre-
vestigative phase of the diagnosis. During the initial pared to discuss this openly with the patient and to
investigations of complaints or suspicions of cogni- make appropriate referrals. Disclosure should take a
tive loss, the patient’s understanding and attitudes patient-centered approach that maintains the per-
about cognitive loss and dementia, as well as those sonal integrity of the individual and instills a sense
of their family members/caregivers, should be estab- of hope. The immediate psychological impact of the
lished. Diagnostic investigations, including the pas- diagnosis should be assessed.
sage of time when necessary, provide opportunities (3) The potential for adverse psychological conse-
to discuss diagnostic uncertainty and to address spe- quences must be assessed and should be addressed
cific fears, concerns, and preconceptions/misconcep- by education and support of the patient and family/
tions that patients and their family/caregivers might caregivers throughout the diagnostic disclosure pro-
have regarding dementia. Health care professionals cess. Reactions to a diagnosis of dementia can in-
should explore with patients and family/caregivers clude anger, denial, shock, and grief, and health
what their beliefs are about the causes of dementia professionals need to be prepared to deal with these
and how they wish to be informed about the biomed- difficult but often normal stages of emotional reac-
ical model. They should be educated as to how the tions to a life-altering diagnosis. Potential negative
process of making a diagnosis can help long-term reactions may be avoided or moderated through ed-
planning by both the patient and family/caregiver. ucational interventions early in the diagnostic pro-
Those involved in the diagnostic disclosure should cess. The benefits of knowing the diagnosis can be
establish who should be involved (eg, the patient highlighted early in the investigative process, and the
only, the patient and their spouse, the patient and roles of specific health professionals in the diagnos-
other family members). Although the autonomy of tic process as well as their potential roles in later
the patient must be the primary consideration, it is management issues can be clarified.
essential that those involved in the investigative pro- (4) Follow-up plans for the patient and their family/
cess encourage the involvement of relevant family caregivers must be made and discussed at the time of
members and/or other current or potential future care diagnostic disclosure. This follow-up should be tai-
providers in the investigative process and the even- lored to the individual needs of the patient and their
tual diagnostic disclosure. family/caregiver and should include education and
(2) Once a diagnosis is established, this must be dis- linkage to available community supports. Follow-up
closed to the patient and their family/caregivers in a appointments to discuss the diagnosis must be made
manner that is consistent with the expressed wishes available to the patient and their family/caregivers,
of the patient, with a patient-centered approach that including the possibility of separate appointments if
maintains the personal integrity of the individual. An appropriate or requested. If patients have chosen to
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410 409

not yet involve family/caregivers in the diagnostic (Speaker, Clinical Trials), Janssen Ortho Inc. (Medical Ad-
process, they should be strongly encouraged to do so visory Board, Speaker, Clinical Trials).
for follow-up appointments. Follow-up appoint-
ments might involve referral by the primary care
physician to other clinicians for specific services or References
might involve referral back to the primary care phy-
[1] Fisk JD, Sadovnik AD, Cohen CA, Gauthier S, Dossetor JB, Eberhart
sician for ongoing management in cases in which the
A, et al. Ethical guidelines of the Alzheimer Society of Canada. Can
diagnosis is disclosed in a specialized clinic setting. J Neurol Sci 1998;25:242– 8.
More specific information, such as written summa- [2] Post SG, Whitehouse PJ. Fairhill guidelines on ethics of the care of
ries, that is valued by patients and family/caregivers people with Alzheimer’s disease: a clinical summary. J Am Geriatr
can be provided through follow-up sessions. Data are Soc 1995;43:1423–9.
[3] Cohen CA, Whitehouse PJ, Post SG, Gauthier S, Eberhart A, LeDuc
lacking that reflect patients’ perspectives on the di-
L. Ethical issues in Alzheimer’s disease: the experience of a national
agnostic disclosure process, the consequences of di- Alzheimer Society Task Force. Alzheimer Dis Assoc Disord 1999;
agnostic disclosure, and in particular the long-term 13:66 –70.
consequences. Because the limited information [4] Patterson CJS, Gauthier S, Bergman H, Cohen CA, Feightner JW,
available points to discrepancies in physician and Feldman H, et al. The recognition, assessment and management of
dementing disorders: conclusions from the Canadian Consensus Con-
caregiver opinions about the disclosure process, it is
ference on Dementia. CMAJ 1999;160(Suppl):S1–15.
incumbent on health care professionals to evaluate [5] Petersen RC, Stevens JC, Ganguli M, Tangalos EG, Cummings JL,
this process within the context of their practice. Re- DeKosky ST. Practice parameter: early detection of dementia—mild
gardless of who discloses the diagnosis, everyone cognitive impairment (an evidence-based review). Neurology 2001;
involved in the care of the patient has a responsibil- 56:1133– 42.
ity to ensure that a plan for follow-up appointments [6] Maguire CP. Telling the diagnosis of dementia: consider each patient
individually. Int Psychogeriatrics 2002;14:123– 6.
is established and carried out. [7] Marzanski M. Would you like to know what is wrong with you? on
telling the truth to patients with dementia. Journal of Medical Ethics
2000;26:108 –13.
2. Recommendations Approved at the Third CCCDTD [8] Carpenter B, Dave J. Disclosing a dementia diagnosis: a review of
Conference opinion and practice, and a proposed research agenda. Gerontologist
2004;44:149 –58.
[9] Bamford C, Lamont S, Eccles M, Robinson L, May C, Bond J.
(1) The process of diagnostic disclosure for persons with Disclosing a diagnosis of dementia: a systematic review. Int J Geri-
cognitive impairment or dementia must begin as atric Psychiatry 2004;19:151– 69.
soon as the possibility of cognitive impairment is [10] Turner S, Iliffe S, Downs M, Wilcock J, Bryans M, Levin E, et al.
suspected (Grade A, Level 3). General practitioner’s knowledge, confidence and attitudes in then
(2) Both the diagnosis of dementia and the disclosure of diagnosis and management of dementia. Age Ageing 2004;33:461–7.
[11] Cody M, Beck C, Shue VM, Pope S. Reported practices of primary
the diagnosis must be considered processes that pro- care physicians in the diagnosis and management of dementia. Age-
vide opportunities for education and discussion ing Ment Health 2002;6:72– 6.
(Grade A, Level 3). [12] Drickamer MA, Lachs MS. Should patients with Alzheimer’s disease
(3) The potential for adverse psychological conse- be told their diagnosis? N Engl J Med 1992;326:947–51.
quences must be assessed and addressed through [13] Lopez OL, Litvan A, Catt KE, Stowe R, Klunk W, Kaufer DI, et al.
Accuracy of four clinical diagnostic criteria for the diagnosis of
education of the patient and family/caregivers neurodegenerative dementias. Neurology 1999;53:1292–9.
(Grade B, Level 3). [14] Petersen RC. Mild cognitive impairment as a diagnostic entity. J In-
(4) Once a diagnosis is established, this must be dis- tern Med 2004;256:183–94.
closed to the patient and their family/caregivers in a [15] Fisk JD, Rockwood K. Outcomes of incident mild cognitive impair-
manner that is consistent with the expressed wishes ment in relation to case definition. J Neurol Neurosurg Psychiatry
2005;76:1175–7.
of the patient (Grade B, Level 3). [16] Smith AP, Beattie BL. Disclosing a diagnosis of Alzheimer’s disease:
(5) Follow-up plans must be made and discussed at the patient and family experiences. Can J Neurol Sci 2001;28(Suppl
time of diagnostic disclosure (Grade A, Level 3). 1):S67–71.
[17] Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, et
al. Frontotemporal lobar degeneration: a consensus on clinical diag-
Author Disclosures nostic criteria. Neurology 1998;51:1546 –54.
[18] Mullen R, Howard R, David A, Levy R. Insight in Alzheimer’s
John Fisk has received support from AstraZenca (Consul- disease. Int J Geriatr Psychiat 1996;11:645–51.
tant), Bayer (Consultant), Biogen-Idec (Consultant), Bristol- [19] DeLepeleire J, Butinx F, Aertgeerts B. Disclosing the diagnosis of
dementia: the performance of Flemish general practitioners. Int Psy-
Myers Squibb (Consultant), Novartis (Consultant), Sanofi-
chogeriatrics 2004;16:421– 8.
Aventis (Consultant) and TEVA Neuroscience (Speaker). [20] Maguire CP, Kirby M, Coen RE, Coakley D, Lawlor BA, O’Neill D.
Lynn Beattie has received support from Novartis (Med- Family members’ attitudes toward telling the patient with Alzhei-
ical Advisory Board Member, Speaker), Pfizer Canada mer’s disease their diagnosis. Br Med J 1996;313:529 –30.
410 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 404 – 410

[21] Erde EL, Nadal EC, Scholl TO. On truth telling and the diagnosis of [30] Pinner G, Bouman WP. To tell or not to tell: on disclosing the
Alzheimer’s disease. J Fam Pract 1988;26:401– 6. diagnosis of dementia. Int Psychogeriatrics 2002;14:127–37.
[22] Holroyd S, Snustad DG, Chalifoux ZL. Attitudes of older adults on [31] Pinner G, Bouman WP. What should we tell people about dementia?
being told the diagnosis of Alzheimer’s disease. J Am Geriatr Soc Advances in Psychiatric Treatment 2003;9:335– 41.
1996;44:400 –3. [32] Henderson JN, Gutierrez-Mayka M. Ethnocultural themes in caregiv-
[23] Holroyd S, Turnbull Q, Wolf AM. What are patients and their fam- ing to Alzheimer’s disease patients in Hispanic families. Clin Ger-
ilies told about the diagnosis of dementia? results of a family survey. ontol 1992;11:59 –74.
Int J Geriatr Psychiatry 2002;17:218 –21. [33] Braun KL, Browne CV. Perceptions of dementia, caregiving and help
[24] Fahy M, Wald C, Walker Z, Livingston G. Secrets and lies: the seeking among Asian and Pacific Islander Americans. Health Soc
dilemma of disclosing the diagnosis to an adult with dementia. Age Work 1998;l23:262–73.
[34] Hinton L, Franz CE, Teo G, Levkoff SE. Conceptions of dementia in
Ageing 2003;32:439 – 41.
a multiethnic sample of family caregivers. J Am Geriatr Soc 2005;
[25] Ferris SH, Hofeldt GT, Carbone G, Masciandaro P, Troetel WM,
53:1405–10.
Imbimbo BP. Suicide in two patients with a diagnosis of probable
[35] Connell CM, Boise L, Stuckey JC, Holmes SB, Hudson ML. Atti-
Alzheimer disease. Alzheimer Dis Assoc Disord 1999;13:88 –90.
tudes toward the diagnosis and disclosure of dementia among family
[26] Vega U, Kishikawa, Ricanatti E, Friedland RP. Suicide and Alzhei-
caregivers and primary care physicians. Gerontologist 2004;44:
mer disease. Am J Geriatric Psychiatry 2002;10:484 –5. 500 –7.
[27] Draper B, Maccuspie-Moore C, Brodaty H. Suicidal ideation and the [36] Jha A, Tabet N, Orrell M. To tell or not to tell: comparison of older
‘wish to die’ in dementia patients: the role of depression. Age Ageing patients’ reaction to their diagnosis of dementia and depression. Int J
1998;27:503–7. Geriatric Psychiatry 2001;16:87– 885.
[28] Turnbull Q, Wolf AMD, Holroyd S. Attitudes of elderly subjects [37] Aminzadeh F, Byszewski AM, Molnar FJ, Eisner M. Emotional
toward “truth telling” for the diagnosis of Alzheimer’s disease. J impact of dementia diagnosis: exploring persons with dementia and
Geriatr Psychiatry Neurol 2003;16:90 –3. caregivers’ perspectives. Aging Ment Health 2007;11:281–90.
[29] Rubio A, Vestner AL, Stewart JM, Forbes NT, Conwell Y, Cox C. [38] Byszewski A, Molnar F, Aminzadeh F, Eisner M, Gardezi F, Bassett
Suicide and Alzheimer’s pathology in the elderly: a case-control R. Dementia diagnosis disclosure: a study of patient and caregiver
study. Biol Psychiatry 2001;49:137– 45. perspectives. Alzheimer Dis Assoc Disord 2007;21:107–14.
Alzheimer’s & Dementia 3 (2007) 411– 417

Ethical considerations for decision making for treatment


and research participation
John D. Fiska,*, B. Lynn Beattieb, Martha Donnellyc
a
QE II Health Sciences Centre; Department of Psychiatry, Department of Medicine, and Department of Psychology, Dalhousie University, Halifax, Nova
Scotia, Canada
b
Department of Medicine, Division of Geriatric Medicine, University of British Columbia, and the BC Network for Aging Research, Vancouver, British
Columbia, Canada
c
Division of Community Geriatrics, Department of Family Practice, and Division of Geriatric Psychiatry, Department of Psychiatry, University of
British Columbia, and Geriatric Psychiatry Outreach Team, Vancouver Hospital, Vancouver, British Columbia, Canada

Abstract Here we review issues of patient decision-making and consent to treatment and research by
persons with cognitive impairment and dementia. Clinicians and researchers must recognize their
primary duty to care for the individual and must clearly distinguish their role as a clinician and/or
researcher. Distinctions between standard care and research must be clearly understood by everyone,
as must the clinician’s role in each. Both actual and perceived conflicts of interest must be avoided.
At present there is insufficient evidence to recommend specific methods for determining compe-
tency for decision-making, but a diagnosis of cognitive impairment or dementia does not preclude
such competence. Competency is not a unitary or static construct and must be considered as the
ability to make an informed decision about participation in the particular context of the specific
treatment or study. Clinicians and researchers should consider consent as a process involving both
the patient with cognitive impairment and his or her family/caregiver, particularly given the
potential that competency for decision-making will change over time. As the availability of advance
directives remains limited, clinicians and researchers must make efforts to ensure that decisions
made by proxies are based on the prior attitudes and values of the patient.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Alzheimer’s disease; Dementia; Decision-making; Competency; Ethics

For the current revision of the Canadian Consensus Con- view, the PubMed and Embase databases were searched for
ference on Dementia (CCCD), the emergence of improved articles with the keywords “Dementia OR Alzheimer’s dis-
methods for early diagnosis of dementia as well as the ease AND ethics AND competency.” For discussion, pref-
availability of approved symptomatic treatments for Alzhei- erence was given to publications between 1996 and 2006.
mer’s disease meant that the issues of patient decision We also examined Canadian and international guidelines on
making for treatment and research required discussion. The the ethics of research, with particular attention to issues
ethical considerations surrounding these issues had not been relevant to the participation of individuals with cognitive
included in previous CCCD guidelines [1]. Not only did impairment and dementia in research.
these issues seem of greater current priority, but also the We review and discuss issues on consent to treatment in
ethical issues of decision making seemed to represent a clinical practice and consent to research participation by
logical extension of the discussions and recommendations persons with cognitive impairment and dementia. We also
surrounding the issue of diagnostic disclosure. Both have review studies that have examined patient decision making
the issue of patient autonomy at their center. For this re- for treatment and the participation of persons with dementia
in research. Although attempts have been made to draw
*Corresponding author. Tel.: 902-473-2581; Fax: 902-473-7166. distinctions between the ethics of clinical care and the ethics
E-mail address: John.Fisk@cdha.nshealth.ca of the conduct of research [2], this view is strongly opposed
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.08.001
412 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417

by many [3] and is inconsistent with most national and benefits, and the selection of subjects. In Canada, research
international ethical guidelines for research [4,5]. Thus on human subjects conducted at institutions that receive
many of the same principles apply, regardless of whether support for federal funding bodies must be conducted in
treatment or research participation is the issue of discussion. accordance with guidelines established jointly by the Cana-
For this reason, we have chosen to discuss these issues dian Institutes for Health Research, the Social Sciences and
together in a single document and have combined the dis- Humanities Research Council, and the National Science and
cussions and recommendations regarding treatment decision Engineering Research Council [10] that are largely based on
making and research participation decision making. these same ethical principles. The same principles are
Although there might not be clear distinctions between present in most international regulatory research guidelines
the ethical issues that clinicians and scientists must con- such as the World Medical Association’s Declaration of
sider, there are nonetheless clear distinctions between the Helsinki [11], the Ethical Guidelines for Biomedical Re-
roles of clinician and scientist. Specifically, appointments, search of the Council for International Organisations of
investigations, and data collection that represent manag- Medical Sciences (CIOMS) [5], and the Council of Eu-
ment of an individual’s health care must be clearly distin- rope’s Convention on Human Rights and Biomedicine [12].
guished from similar activities that represent the conduct of Internationally accepted guidelines for the conduct of clin-
a clinical trial for the research participant. Clinicians need to ical trials are published in the International Conference on
be keenly aware of the ethical issues, such as conflicts of Harmonisation of Technical Requirements for Registration
interest, that they are likely to face when they conduct of Pharmaceuticals for Human Use Harmonised Tripartite
clinical trials. They must also be aware of the values and Guideline (ICH) [13]. Unlike the ethical guidelines for
beliefs on which their own decision making is based. How- research, these guidelines, often referred to as the “good
ever, although research involving patients with cognitive clinical practice” (ICH-GCP) guidelines, were produced as
impairment and dementia presents its own particular chal- a joint regulatory and industry project “to facilitate the
lenges, the ethical considerations themselves are not unique adoption of new or improved technical research and devel-
to dementia. opment approaches which update or replace current prac-
In 1997 the Alzheimer Society of Canada’s ethical tices, where these permit a more economical use of human,
guidelines document [6,7] addressed both autonomy in de- animal and material resources, without compromising
cision making and participation in research. Even though safety” [13]. Given their origins and mandate, it is perhaps
the lack of available symptomatic treatment in Canada not surprising that despite recognizing the need to address
at that time limits their current applicability to some extent, issues specific to the conduct of clinical trials in elderly
these guidelines illustrate an important point. Specifically, populations and reference to research on Alzheimer’s dis-
they illustrate the differences that can emerge between gen- ease [14], ICH-GCP guidelines provide no insight into the
eral international guidelines on the ethical conduct of re- specific ethical issues that arise in those contexts. As with
search and guidelines that are developed by individuals who all guidelines, the challenge becomes how to find the right
are affected by specific medical conditions and their repre- balance when attempting to apply the general principles to
sentatives. Individuals affected by specific medical condi- the specific ethical dilemmas that arise.
tions and their advocates, while continuing to point out Perhaps the most obvious ethical dilemma associated
the importance of protecting the rights of vulnerable indi- with clinical trials for cognitive impairment and dementia is
viduals, generally place greater emphasis on ensuring that balancing respect for the autonomy of the individual with
the potential to benefit from participation in research is not the protection of vulnerable persons. Most ethical guide-
denied to specific groups of individuals. This inclusive lines regarding research focus on the individual in terms of
stance toward research participation by persons with de- the analysis of risk/benefit ratios and the consent process.
mentia has been articulated by High et al [8]: “To deny Guidelines developed for persons with Alzheimer’s disease,
persons access to research participation out of fear of ex- however, also acknowledge the need to consider the conse-
ploitation of specific groups of persons is to avoid rather quences of research participation on families and the im-
than accept and practice ethical responsibility.” portance of involving the caregiver/family in the consent
The principles that best describe ethical decision making process [8,15]. Even for individuals with Alzheimer’s dis-
regarding research participation are those articulated in the ease who are competent to provide informed consent, con-
Belmont Report, produced by the US National Commission sent might be required of their caregiver/family. Typically it
for the Protection of Human Subjects of Biomedical and is they who will be expected to bring the person to sched-
Behavioural Research [9]. These include the principles of uled appointments; observe the subject for adverse events,
respect for persons, beneficence (ie, the obligation to do no including those that should be reported immediately to the
harm and to maximize the potential for benefit while min- research team, as well as those of less urgent nature; ensure
imizing the potential for harm), and justice. These princi- that the subject takes medications as required; observe and
ples, in turn, are applied to clinical research through con- report possible treatment benefits; relay to the investigators
sideration of informed consent, assessment of risks and the subject’s continued assent to participation; and take part
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417 413

in evaluating treatment efficacy, such as reporting changes statement. However, this should not be construed to
in emotional burden or time spent on tasks for care. mean that the Association necessarily opposes involve-
Most guidelines promote the view that “the principle of ment of decidedly impaired individuals in all nonthera-
respect for human dignity entails high ethical obligations to peutic research involving minor increments over mini-
mal risk. The degree of risk is a judgment call frequently
vulnerable populations” [10]. However, most also agree
and appropriately made by IRBs [institutional review
with the concept that the “protection should be proportion- boards]” [20].
ate to the risk involved, with the least protection required
when research involves minimal risk” [16]. This consider- Although it has been argued that research ethics review
ation of the limits of the acceptable risk/benefit ratio is boards spend a disproportionate amount of time addressing
central to the evaluation of all clinical trials [17]. However, the issue of informed consent at the expense of addressing
it becomes particularly difficult when evaluating clinical the analysis of risk [17], free and fully informed consent is
trials for patients with cognitive impairment and dementia both central to the ethical conduct of research and particu-
because of the potential for uncertainty about the subject’s larly problematic for research in dementia. Most ethical
capacity for autonomous and informed decision making. A guidelines take a relatively protectionist stance with regard
decade ago, High et al [8] noted “no clear consensus exists to avoiding the potential for exploitation of individuals who
either in the literature or in regulatory guidelines as to what are not competent to provide informed consent, but vari-
constitutes an acceptable degree of risk when cognitively ability does exist among various guidelines [21]. A diagno-
impaired persons are involved in research.” This remains sis of dementia or other forms of cognitive impairment does
the case today. not preclude competence to provide informed consent for
The concept of minimal risk has been used in most participation in clinical trials [22] or in nontherapeutic re-
international ethical guidelines for research, although it has search, which contains minimal risk. Improved diagnostic
been described in various ways [17]. Usually, this is meant methods also mean that Alzheimer’s disease and other de-
to ensure that “the probability and magnitude of harm or mentias are detected at earlier stages in which many cogni-
discomfort anticipated in the research are not greater in and tive and functional abilities are relatively preserved. The
of themselves than those ordinarily encountered in daily life conceptualization of mild cognitive impairment (MCI) [23]
or during the performance of routine physical or psycho- as a diagnostic entity that warrants clinical therapeutic and
logical examinations or tests” [18]. Such is rarely the case in prevention trials reinforces this trend.
clinical trials of new treatments for dementia, however, and Competence to consent to either treatment or research
although it might be argued that research that does not participation is not a simple matter of the stage of dementia
involve “risk of harm beyond a minor increment over min- or severity of cognitive impairment for an individual. In
imal” [19] is acceptable when dealing with cognitively part, this is because competence is not a unitary construct.
impaired individuals, such terms are open to the criticism of One can be competent in some aspects of one’s life (eg,
being poorly defined and allowing wide interpretation. Also competent to consent to research) without being competent
contributing to difficulty in establishing consensus on the in others (eg, competent to drive a motor vehicle) [24]. As
issue of risk is the use of varied ethical guidelines for ethics the risk/benefit ratio and other aspects of a given treatment
review. In Canada, research on human subjects conducted at or study vary, so do the requirements for competence. This
institutions that receive support for federal funding bodies concept of a “sliding-scale” of competency [25] is most
must be conducted in accordance with the Tricouncil Policy typically considered in the context of research participation,
Statement [10], but research conducted outside such insti- as acknowledged in the Tri-Council Policy Statement:
tutions is governed by a patchwork of laws and voluntary “Competence to participate in research, then, is not an
industry guidelines such as the ICH-GCP guidelines [13]. all-or-nothing decision. It requires that they be compe-
Both international regulatory bodies and organizations with tent to make an informed decision about participation
the mandate of patient advocacy typically defer decisions of in particular research. Competence is neither a global
ethical acceptability of clinical trials to individual ethical condition nor a static one; it may be temporary or
review boards. The difficulties in coming to terms with permanent” [10].
the issue of risk, even for an organization such as the Although much of the discussion regarding the ethical
Alzheimer’s Association in the USA, are evident in their issues of decision making has been focused on the topic of
statement: research participation, most of the research on the topic
of competency has focused on consent to treatment. The use
“There is considerable disagreement in the ethics liter-
ature and in regulatory and statutory language about
of standardized methods to determine competency to pro-
definitions of risk. In addition to the two polar defini- vide informed consent has been promoted by some, but
tions, ‘minimal risk’ and ‘greater than minimal risk,’ there remains no clear consensus on the best standardized
there are also various gradations, such as ‘minor increase assessment methods to address this [24,26]. Moreover,
over minimal risk.’ The Alzheimer’s Association has given the evidence of inconsistencies in expert clinical judg-
chosen not to draw these finer distinctions in its position ments on the issue of competence for mildly affected pa-
414 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417

tients [27], the question can be raised as to what can be the context of diagnostic disclosure. First and foremost,
considered a gold standard for evaluating such methods. For informed proxy decision making by the family/caregiver
the primary care physician with limited time and access to requires that they be informed about dementia. Unfortu-
formal competency assessment methodologies, it would be nately, the limited numbers of studies to date that have
advantageous if standard mental status screening instru- examined caregiver knowledge about Alzheimer’s disease
ments could be used to determine consent capacity as well have found relatively poor understanding of issues such as
as for diagnostic screening purposes. There is limited evi- causes, symptoms, and drug treatments [37]. This highlights
dence to suggest that screening instruments can provide the need for education of patients and their family/caregivers in
some guidance as to the probability of competency to con- the diagnostic disclosure process before the possibility of treat-
sent to treatment [28,29] and to research [30], but the ment and/or research participation is raised. Clinicians must
complexity of the issues precludes any simple means of also recognize that, as with other demands of providing care
establishing competency in a specific treatment or research to persons with dementia, the process of providing proxy
setting with certainty [31]. Clearly, more impaired individ- consent can itself be burdensome, particularly when the
uals are less likely to be competent to make informed risks of clinical trial participation are not fully appreciated
decisions, but beyond this, the issues become much more [38]. Factors affecting proxy decision making were exam-
complex. ined by Karlawish et al [39] in a study of caregivers’ views
Research on competency for the provision of informed about the acceptance of risk associated with potential treat-
consent in a research context has been less common than for ments for Alzheimer’s disease. They noted that caregivers’
consent to treatment. Although Kim et al [32] have reported tolerance for risk of death associated with a hypothetical
that even mild Alzheimer’s disease has significant effects on treatment was influenced by demographic factors and that
competency to consent to research, this issue remains an working adult children who were caring for early-stage
important topic for further research. Even if standardized patients were more willing to accept that risk than spouses
assessments of competency to participate in clinical or re- of such patients. For the clinician, these views point out the
search decision making were to become available, signifi- need to recognize that although proxies are expected to
cant practical, social, cultural, and legislative variations are represent the patient, they are not without their own biases
still likely to limit their application. The absence of clear when it comes to decision making. It also points out the
and consistent legislation regarding competency for deci- importance of ensuring that decisions made by proxies re-
sion making and the important role of proxy decision mak- garding treatment and research participation reflect the prior
ing, particularly in the context of research participation, attitudes and values of the patient rather than those of the
have been recognized for some time [25,33]. Across Can- proxy.
ada, legislation regarding substitute decision making for the A study by Connell et al [40] of caregiver attitudes
participation of incompetent individuals in research is often toward participation in longitudinal, nontherapeutic re-
unclear and varies among provinces and territories [34]. search is also important in that it points out the possible
For both practical and ethical reasons in most situations, presence of expectations of improved access to clinical care
clinicians, researchers, and research ethics boards have typ- through research participation. Although the direct implica-
ically relied on a family member of the person with demen- tions of this American study to a Canadian context can be
tia to provide “third party authorization” for decision mak- questioned, it also pointed out the existence of ethnic/racial
ing, even when there has been no legal determination that differences in attitudes toward research, presenting barriers
the patient lacks competence to provide informed consent. to equitable research participation that are likely to gener-
In some cases, proxy consent is aided by the availability of alize beyond the USA. As was discussed in the context of
advance directives for research participation that specifi- diagnostic disclosure, we have very limited information
cally identify proxy decision makers. But despite the pro- regarding the potential influences of ethnic and cultural
motion of the use of advance directives by some [19], issues with regard to treatment and research participation
concerns about this have been raised because of their lim- decision making among patients with dementia and their
ited availability [35]. Even when consent from a legally family/caregivers. This is an important consideration with
authorized third party is given, the ongoing assent of the regard to the ethical principle of justice and the equal
individual subject is almost invariably necessary. distribution of the risks and benefits. The possibility of
Despite the common use of proxies in the decision- systematic differences in expected benefits of research par-
making process, reservations about proxy decision making ticipation reinforces the need to draw a clear distinction
for participation by cognitively impaired subjects in re- between research participation and clinical care for both the
search have been expressed for some time [36]. More patient and their family/caregiver. Although improved mon-
recently, the factors that influence caregivers’ decisions itoring and access to health care professionals might be the
regarding research participation have themselves become case in some clinical trials, the assumption that receiving
the source of study. Many of the considerations that have treatment through participation in a clinical trial results in
arisen are an extension of those raised previously in improved outcome over similar usual care has not been
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417 415

supported [41]. It is also the case that participation in studies ing for persons with dementia has its own set of challenges that
might reduce rather than improve access to care, for exam- require careful consideration. The existence of social and
ple, when subjects are enrolled into the placebo arm of a cultural barriers to the implementation of existing ethical
clinical trial of a new treatment rather than being started on guidelines for clinical practice must be acknowledged, and
an established therapy. the extent to which similar barriers exist in the implemen-
Research directly related to proxy decisions about enroll- tation of guidelines for the ethical conduct of research is
ment in clinical trials has been relatively rare. Those studies itself an important topic that warrants research [43].
that have been conducted suggest that although proxies
actively involve the research subjects in the decision- 1. Summary
making process [38,42], they do not always think that
their decisions are consistent with those of the research
(1) At present there is insufficient evidence to recom-
subject [15]. For the clinician and researchers, it is impor-
mend a specific standardized method for determining
tant to recognize that the interests of the person with de-
the competency of persons with dementia for deci-
mentia and those of a substitute decision maker might differ.
sion making for treatment or research. A diagnosis of
Thus, clinicians have an obligation to determine, to the best
dementia, or other forms of cognitive impairment,
of their ability, that the decisions regarding treatment and
does not preclude competence to provide informed
research participation are guided by the individual patient’s
consent for treatment or for participation in research,
wishes and, if this is not clearly known, that it has been
and competency must be considered as the ability to
made with the individual patient’s best interests in mind
make an informed decision about participation in the
[8,10,15].
particular context of the specific treatment or study.
As new potential treatments for dementia become avail-
(2) Even in the absence of a legal determination of
able for study and use, decision making on the part of
competency, clinicians and researchers should con-
patients and their family/caregivers will become increas-
sider obtaining consent as a process involving both
ingly complex and will require even greater educational
the patient with dementia and their family/caregiver.
efforts and systematic evaluation. Because changes in com-
(3) The distinctions between the clinician’s role in the
petency are to be expected over time in most of the demen-
management of the individual’s health care and his/
tias, there is also clear need for ongoing monitoring and
her potential role in the conduct of research must be
reaffirmation of consent/assent. Indeed, this could easily be
clearly understood by everyone, as must the proce-
considered an important requirement even when compe-
dures that represent standard care and research. Con-
tency of the subject can be assured. Thus, for all research, it
flicts of interest, both actual and perceived, must be
is reasonable to expect that the procedures to evaluate the
avoided. If conflicts arise in a research context, the
ability of the potential subject to understand the nature of the
opinion of a physician other than the research phy-
research, the consequences of participation (ie, potential risks
sician might be necessary to ensure that decisions
and benefits), and alternative choices are described [7,15].
regarding treatment and continued research partici-
In summary, it is important that the clinicians and re-
pation are made in the best interests of the patient.
searchers are aware of their primary duty to care for the
(4) The potential that competency for decision making
individual and to clearly distinguish their roles as a clinician
will change over time must be recognized. This
and/or researcher. They must also be aware of the values
might lead to a change from one of obtaining the
and beliefs that influence their own ethical decision making.
patient’s ongoing consent to one of obtaining ongo-
The ethical principles of autonomy, beneficence, and justice
ing assent. Assent is usually required.
represent a balance. In research, it is the principles of
(5) To the best of their ability, clinicians and researchers
autonomy (respect for persons) and beneficence (do no
must ensure that the decisions made by proxies re-
harm/do good) that might be most obviously in conflict, but
garding treatment and research are based on the prior
this is not unique to research, and this conflict will likely
attitudes and values of the patient. This might be
become even more apparent as new treatments for dementia
assisted by the availability of advance directives.
become available and/or as the risks of current treatments
become better understood. Finding balance requires the ethical
analysis of risk and the process of informed consent. However, 2. Recommendations approved at the Third
considerations of informed consent in dementia invariably Canadian Consensus Conference on Diagnosis and
come up against the issue of competency. It is important to Treatment of Dementia
recognize that competency is not a unitary or static construct,
and the variability of existing legislation and current ethical (1) Provision of the best standard of care for the patient
guidelines on this issue might be of limited help to the indi- must always remain the priority (Grade A, Level 3).
vidual clinician or researcher dealing with the individual pa- (2) Drawing a clear distinction between research partic-
tient and a specific set of circumstances. Proxy decision mak- ipation and clinical care is essential for both the
416 J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417

patient and their family/caregiver. The distinctions Author Disclosures


between the clinician’s role in the management of
John Fisk has received support from AstraZenca (Con-
the individual’s health care and his/her potential role
sultant), Bayer (Consultant), Biogen-Idec (Consultant),
in the conduct of research must be clearly under-
Bristol-Myers Squibb (Consultant), Novartis (Consultant),
stood by everyone, as must the procedures that rep-
Sanofi-Aventis (Consultant) and TEVA Neuroscience
resent standard care and research. In research set-
(Speaker).
tings, the availability of a physician other than the
Lynn Beattie has received support from Novartis (Med-
research physician to provide general care is recom-
ical Advisory Board Member, Speaker), Pfizer Canada
mended to ensure that decisions regarding treatment
(Speaker, Clinical Trials), Janssen Ortho Inc. (Medical Ad-
are made in the best interests of the patient (Grade A, visory Board, Speaker, Clinical Trials).
Level 3).
(3) A diagnosis of dementia, or other forms of cognitive
impairment, does not preclude competence to pro-
vide informed consent, whether it is for treatment References
decisions, for participation in clinical trials, or for
[1] Patterson CJS, Gauthier S, Bergman H, Cohen CA, Feightner JW,
participation in nontherapeutic research. Compe- Feldman H, et al. The recognition, assessment and management of
tency must be considered as the ability to make an dementing disorders: conclusions from the Canadian Consensus Con-
informed decision about participation in the partic- ference on Dementia. CMAJ 1999;160(Suppl):S1–15.
ular context of the specific treatment or research [2] Miller FG, Brody H. What makes placebo-controlled trials unethical?
Am J Bioeth 2002;2:3–9.
study (Grade A, Level 3).
[3] Lemmens T, Miller PB. Avoiding a Jekyll-and-Hyde approach to the
(4) For studies, it is reasonable to expect that the pro- ethics of clinical research and practice. Am J Bioeth 2002;2:14 –7.
cedures that will be used to evaluate the ability of the [4] American Medical Association. Code of medical ethics. Available at:
potential subject to understand the nature of the www.ama-assn.org/ama/pub/category/8292.html. Accessed: Septem-
research, the consequences of participation (ie, po- ber 6, 2007.
[5] Council for International Organizations of Medical Sciences. Interna-
tential risks and benefits), and alternative choices are tional ethical guidelines for biomedical research involving human sub-
described. However, at present there is insufficient jects, 2002. Available at: www.cioms.ch/frame_guidelines_nov_2002.
evidence available to recommend the use of a spe- htm. Accessed: September 6, 2007.
cific standardized method for determining compe- [6] Alzheimer Society of Canada. Ethical guidelines, 2003. Available at:
www.alzheimer.ca/english/care/ethics-intro.htm.
tency for decision making either for treatment or
[7] Fisk JD, Sadovnik AD, Cohen CA, Gauthier S, Dossetor JB, Eberhart
research (Grade B, Level 3). A, et al. Ethical guidelines of the Alzheimer Society of Canada. Can
(5) Even in the absence of a legal determination of the J Neurol Sci 1998;25:242– 8.
competency of the patient with cognitive impairment [8] High DM, Whitehouse PJ, Post SJ, Berg L. Guidelines for addressing
or dementia, it is important that the clinician and ethical and legal issues in Alzheimer disease research: a position
paper. Alzheimer Dis Assoc Disord 1994;8:66 –74.
researcher consider the consent process as one that [9] National Commission for the Protection of Human Subjects of Bio-
should involve both the patient and their family/ medical and Behavioural Research, the Belmont Report, ethical prin-
caregiver for treatment and research decision mak- ciples and guidelines for the protection of human subjects of research,
ing. In research settings, research ethics boards 1979. Available at: http://www.hhs.gov/ohrp/humansubjects/guidance/
belmont.htm. Accessed: September 6, 2007.
might explicitly require that consent/assent be ob-
[10] Tri-Council policy statement: ethical conduct for research involving
tained from both parties (Grade B, Level 3). humans, 1998 (with 2000, 2002 updates).Available at: http://pre.
(6) The potential that competency for treatment and re- ethics.gc.ca/english/policystatement/policystatement.cfm.
search decision making will change over time must [11] World Medical Association Declaration of Helsinki, ethical principles
be recognized. This might lead to a change from one for medical research involving human subjects. Available at: www.
wma.net/e/policy/b3.htm. Accessed: September 6, 2007.
of obtaining the patient’s ongoing consent to one of [12] Council of Europe, Convention for the Protection of Human Rights
obtaining ongoing assent. Assent is almost invari- and Dignity of the Human Being with Regard to the Application of
ably required, and the decision to discontinue treat- Biology and Medicine: Convention on Human Rights and Biomedi-
ment, whether it is therapy or research, must always cine, Oviedo, 4.IV. 1997. Available at: http://conventions.coe.int/
treaty/en/Treaties/Html/164.htm. Accessed: September 6, 2007.
be an option (Grade A, Level 3).
[13] International Conferences on Harmonization of Technical Require-
(7) To the best of their ability, clinicians and researchers ments for Registration of Pharmaceuticals for Human Use (ICH).
must ensure that the decisions made by proxies re- Revised ICH Terms of Reference. Available at: http://www.ich.org/
garding treatment and research are based on the prior cache/compo/276-254-1.html. Accessed: September 6, 2007.
attitudes and values of the patient. Proxies have a [14] International Conference on Harmonization of Technical Require-
ments for Registration of Pharmaceuticals for Human Use, 1993. ICH
responsibility to represent the patient, and all parties Harmonised Tripartite Guideline, Studies in Support of Special Pop-
must recognize the challenges of doing so (Grade A, ulations: Geriatrics E7. Available at: http://www.ich.org/cache/
Level 3). compo/276-254-1.html. Accessed: September 6, 2007.
J.D. Fisk et al. / Alzheimer’s & Dementia 3 (2007) 411– 417 417

[15] Karlawish JHT. Research involving cognitively impaired adults. [30] Kim SYH, Caine ED. Utility and limits of the Mini Mental State
N Engl J Med 2003;348:1389 –92. Examination in evaluating consent capacity in Alzheimer’s disease.
[16] Office of Human Subjects Research, information sheet 7, National In- Psychiatric Services 2002;3:1322– 4.
stitutes of Health. Available at: http://ohsr.od.nih.gov/info/sheet3.html. [31] McQuillan MP, Tariot P. Who cand say yes (or no) to a physician: and
[17] Weijer C. The ethical analysis of risk. J Law Med Ethics 2000;28: how does the physician know they can? Neurology 2005;64:1494 –5.
344 – 61. [32] Kim SY, Caine ED, Currier GW, Leibovici A, Ryan JM. Assessing
[18] Department of Health and Human Services. Code of federal regula- the competence of persons with Alzheimer’s disease in providing
tions title 45 part 46: regulations for the protection of human subjects, informed consent for participation in research. Am J Psychiatry 2001;
section 46.102.i, 2001. Available at: http://www.hhs.gov/ohrp/ 158:712–7.
humansubjects/guidance/45cfr46.htm. Accessed: September 6, 2007.
[33] The Ethics and Humanities Subcommittees of the American Acad-
[19] Keyserlingk EW, Glass K, Kogan S, Gauthier S. Proposed guidelines
emy of Neurology. Ethical issues in clinical research in neurology.
for the participation of persons with dementia as research subjects.
advancing knowledge and protecting human research subjects. Neu-
Perspect Biol Med 1995;38:319 – 62.
rology 1998;50:592–5.
[20] Alzheimer’s Association. Ethical Issues in Dementia Research (with
special emphasis on “informed consent”). Available at: http://www. [34] Alzheimer Society of Canada. Tough issues: ethical guidelines of
alz.org/national/documents/statements_ethicalissues.pdf. Accessed: the Alzheimer Society of Canada. Toronto: Alzheimer Society of
September 6, 2007. Canada; 1997.
[21] Wendler D, Prasad K. Core safe guards for clinical research with [35] Sachs GA, Stocking CB, Stern R, et al. Ethical aspects of dementia
adults who are unable to consent. Ann J Intern Med 2001;135: research: informed consent and proxy consent. Clin Res 1994;42:
514 –23. 403–12.
[22] Karlawish JHT, Casarett DJ, James BD. Alzheimer’s disease patients’ [36] Candilis PJ, Wesley RW, Wichman A. A survey of researchers using
and caregivers’ capacity, competency, and reasons to enroll in an a consent policy for cognitively impaired human research subjects.
early-phase Alzheimer’s disease clinical trial. J Am Geriatr Soc IRB 1993;15:1– 4.
2002;50:2019 –24. [37] Werner P. Correlates of family caregivers knowledge about Alzhei-
[23] Petersen RC, Stevens JC, Ganguli M, Tangalos EG, Cummings JL, mer’s disease. Int J Geriatr Psychiatry 2001;16:32– 8.
DeKosky ST, et al. Practice parameter: early detection of dementia: [38] Sugarman J, Cain C, Wallace R, Welsh-Bohmer KA. How proxies
mild cognitive impairment (an evidence-based review). Neurology make decisions about research for patients with Alzheimer’s disease.
2001;56:1133– 42. J Am Geriatr Soc 2001;49:1110 –9.
[24] Marson DC. Loss of competency in Alzheimer’s disease: conceptual [39] Karlawish JHT, Klocinski JL, Merz J, Clark CM, Asch DA. Care-
and psychometric approaches. Int J Law Ethics 2001;24:267– 83. givers’ preferences for the treatment of patients with Alzheimer’s
[25] High DM. Research with Alzheimer’s disease subjects: informed disease. Neurology 2000;55:1008 –14.
consent and proxy decision-making. J Am Geriatr Soc 1992;40:
[40] Connell CM, Shaw BA, Holmes SB, Foster N. Caregiver’s attitudes
950 –7.
toward their family members’ participation in Alzheimer disease
[26] Moye J, Karel M, Azar AR, Gurrera RJ. Capacity to consent to
research: implications for recruitment and retention. Alzheimer Dis
treatment: empirical comparison of the instruments in older adults
Assoc Disord 2001;15:137– 45.
with and without dementia. Gerontologist 2004;44:166 –75.
[27] Marson DC, McInturff B, Hawkins L, Bartolucci A, Harrell LE. [41] Vist GE, Hagen KB, Devereaux PJ, Bryant D, Kristoffersen DT,
Consistency of physician judgements of capacity to consent in Alz- Oxman AD. Systematic review to determine whether participation in
heimer’s disease. J Am Geriatr Soc 1997;45:453–7. a trial influences outcome BMJ 2005;330:1175– 81.
[28] Etchells E, Darzins P, Silberferfeld M, Singer PA, McKenny J, Naglie [42] Karlawish JHT, Casarett D, Klocinski J, Sankar P. How do AD
G, et al. Assessment of patient capacity to consent to treatment. J Gen patients and their caregivers decide whether to enroll in a clinical
Intern Med 1999;14:27–34. trial? Neurology 2001;56:789 –92.
[29] Karlawish JHT, Casarett DJ, James BD, Xie SX, Kim SYH. The [43] Graham C, Holmes C, Lindesay J. Clinical involvement in anti-
ability of persons with Alzheimer’s disease (AD) to make a decision dementia drug trials: why bother? Int J Geriatr Psychiatry 1999;14:
about taking an AD treatment. Neurology 2005;64:1514 –9. 257– 60.
Alzheimer’s & Dementia 3 (2007) 418 – 427

Genetics and dementia: Risk factors, diagnosis, and management


Ging-Yuek Robin Hsiunga,b,c,*, A. Dessa Sadovnickb,d
a
Division of Neurology, University of British Columbia, Vancouver, BC, Canada
b
Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
c
St. Paul’s Hospital, Providence Health Care Center, Vancouver, British Columbia, Canada
d
Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada

Abstract New developments in molecular genetics have improved our understanding on a number of
neurodegenerative dementias considerably, especially Alzheimer’s disease and frontotemporal de-
mentia. However, this explosion of information can be overwhelming to clinicians, making it
difficult to integrate into regular clinical practice. In this article, we briefly reviewed our current
understanding regarding causative genetic mutations and genetic risk factors on the major forms of
dementia, which provided the background information for discussion in the third Canadian Con-
sensus Conference on Diagnosis and Treatment of Dementia. The principles of genetic counselling
were applied. Guidelines and recommendations on the application of genetics in the assessment,
diagnosis, and management of patients and families with dementia were summarized.
© 2007 The Alzheimer’s Association. All rights reserved.

Keywords: Genetic counselling; Alzheimer’s disease; Frontotemporal dementia; APOE; PSEN1; PSEN2; MAPT; PGRN

1. Background 2. Methods
Advances in genetics, especially with completion of the We initially retrieved 1721 articles by a PubMed search
human genome project, have improved our understanding on genetic risk factors for AD and other dementias from
of the pathogenesis of many diseases, including Alzhei- January 1996 up to and including December 2005. Relevant
mer’s disease (AD) and other types of dementia [1– 4]. With articles before 1996 have been summarized elsewhere in
the increase in public awareness, dementia patients and meta-analyses and reviews [8,9]. We further conducted a
family members as well as the general public now often review of all the genes listed in the ALZGENE website
request genetic testing as part of their assessment of risk of (accessed February 14, 2006) [10] and articles from bibli-
dementia, even in the absence of treatments known to slow ographies of selected studies. Authors’ files were also
or prevent disease progression [5–7]. It is thus expected that scanned. For genetic association studies, we only consid-
these requests will increase over time when such treatments ered those with large sample sizes (⬎300 cases and ⬎300
become realities. The clinician must understand the role of controls), with well-defined clinical diagnostic criteria and
genetic risk factors to provide accurate information to their with findings consistently replicated in four or more inde-
patients and to refer them to genetics clinics when appro- pendent samples. Because we came to the conclusion that
priate. Canadian sites can be found at http://ccmg.medical.org/ APOE is the only genetic risk factor that has been consis-
clinical.html (accessed March 3, 2006). Here we review the tently replicated to date, we further examined studies on
current literature on the genetic risk factors for dementia, APOE and its interaction with other risk factors in relation
summarize the current knowledge, and provide evidence-based to AD. We also looked at single gene mutations studies,
recommendations to clinicians. because within specific families they are known to be causal
and thus represent risk factors for unaffected biologic rela-
*Corresponding author. Tel.: (604) 682-2344 x63459; Fax: (604) 822-7191. tives of demented individuals carrying the specific muta-
E-mail address: hsiung@interchange.ubc.ca tion.
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.010
G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427 419

Table 1
Genes confirmed to be associated with dementia mentioned in this article
Disease Chromosome Gene Penetrance Frequency of Mutation Pathology
Mutation in Families

EOAD 21q21 APP Full penetrance About 18% of familial Missense mutations around Increases A␤42
early-onset AD A␤ portion of APP production from
APP processing
EOAD 14q24.3 PSEN1 Full penetrance Represent up to 78% Mostly missense mutations Promotes cleavage at
of familial early- ␥-secretase site and
onset AD increases A␤
production and
accumulation
EOAD 1q31.42 PSEN2 Incomplete penetrance Rare, ⬃4% of familial Missense mutations Promotes cleavage at
early-onset AD ␤-secretase site
leading to increased
A␤ production and
accumulation
CADASIL 19p13 NOTCH3 Full penetrance 400 families described 95% are missense Functional
worldwide, but mutations consequences of
probably under- mutation still
recognized unclear; probably
affects protein
conformation of
Notch3 product
FTDP-17 17 MAPT Full penetrance 10% to 30% of Missense and splice site Increase ratio of 4-
familial FTD, 5% mutations repeat tau to 3-
of all FTD repeat tau
FTLD-U 17 PGRN High penetrance 23% of familial FTD, Missense, splice site, and Leads to null
5% of all FTD promoter region mutations with
mutations haploinsufficiency

Disease Chromosome Gene Penetrance Allele frequency in Mutation Pathology


population

LOAD 19q13.2 APOE Risk factor, but e2, ⬃10%, e4 genotype, heterozygous Acts as a molecular
neither necessary e3, ⬃75%, increases odds by ⬃3 chaperone to
nor sufficient to e4, ⬃15% and homozygous by ⬃9 promote A␤
cause disease accumulation and
senile plaque
formation

Abbreviations: EOAD, early-onset AD; LOAD, late-onset AD; FTDP-17, FTD with parkinsonism linked to chromosome 17.

3. Results peptide is released, which can undergo further conforma-


tional change into an insoluble form that aggregates in
3.1. Causative mutations for dementia
senile plaques [19 –21]. APP mutations that are causal for
3.1.1. Alzheimer’s disease early-onset familial AD all promote cleavage at the ␤ or ␥
Through linkage analysis of affected pedigrees (posi- sites, leading to an overproduction of the A␤ peptide[20].
tional cloning), mutations in three genes have been found to Two other genes with mutations that cause early-onset fa-
cause early-onset familial AD (Table 1) [1,11]. The first milial AD are presenilin 1 (PSEN1) and presenilin 2
gene identified was the amyloid precursor protein gene (PSEN2), which are located on chromosomes 14 and 1,
(APP) on chromosome 21 [12–16]. Although the physio- respectively [22,23]. Current evidence suggests that both
logic function of the amyloid precursor protein (APP) re- PSEN1 and PSEN2 play an important role in the ␥-secretase
mains unclear, the post-translational processing of APP is complex, and mutations in these genes lead to an excessive
clearly involved in the pathogenesis of AD [17,18]. APP is cleavage at the ␥ site leading to excessive production and
a membrane bound protein that can undergo a series of accumulation of A␤ [17,19]. Taken together, the three
endoproteolytic cleavages by enzymes known as secretases. causal genes identified to date for AD provide strong sup-
When cleaved by ␣-secretase in the middle of the A␤ port for the amyloid cascade hypothesis, in which the ac-
domain within APP, a soluble fragment of the protein is cumulation of A␤ peptide into senile neuritic plaques is
released, and there is no accumulation of the peptide; central to the pathogenesis of AD.
whereas when it is cleaved at the ␤ and ␥ sites, the A␤ To date, although well-documented families exist in
420 G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427

which late-onset AD (onset after age 65 years) appears to be bules. Tau also likely plays a role in microtubule-dependent
inherited in an autosomal dominant manner, no causal mu- axonal transport [40]. Mutations in MAPT causing FTDP-17
tation(s) has been identified. generally reduce the ability of tau to interact with microtu-
bules, leading to faulty microtubule assembly [25,40].
3.1.2. Frontotemporal dementia Nonetheless, the exact mechanism of how tau causes neu-
The use of the term frontotemporal dementia (FTD) has rodegeneration remains unclear and is an area of intense
been inconsistent and is still evolving [4,24 –28]. To clarify, research.
here we use FTD to refer to the clinical presentation of the Recently, causative mutations in the progranulin gene
dementing illness and frontotemporal lobar degeneration (PGRN) have been identified for an autosomal dominant form
(FTLD) to denote the pathologic diagnosis of the disease. of familial FTLD associated with ubiquitinated neuronal in-
FTD is a clinical syndrome of dementia characterized by tranuclear inclusions (FTLD-U) [41]. Mutations in PGRN are
changes in behavior, personality, and social conduct, with responsible for 23% of familial FTD and represent about 5% of
relative preservation of memory function in the early stages all FTD cases encountered in a clinical setting [42]. PGRN is
[4]. Incidence estimates of FTD range from 3.3 per 100,000 a secreted growth factor involved in the regulation of multiple
for 50- to 59-year-olds to 8.9 per 100,000 for 60- to 69- processes including development, wound repair, and inflam-
year-olds [29], representing approximately 5% of all de- mation [43]. Elevated PGRN expression in activated microglia
mentias presenting to specialized Canadian clinics [30]. is found in a number of neurodegenerative diseases including
Pathologically, FTLD is associated with circumscribed de- Creutzfeldt-Jakob disease, amyotrophic lateral sclerosis, and
generation of the prefrontal and anterior temporal lobes with Alzheimer’s disease, suggesting that PGRN might play an
pathologic changes that are distinctly different from AD important role in maintenance of neuronal survival. To date, all
type pathology. The pathology of FTLD is quite varied and identified PGRN mutations leading to disease development are
includes (1) the classic presentation of Pick’s disease with due to null mutations, which likely result in haploinsufficiency
Pick’s bodies, (2) neurofibrillary tangles with tau immuno- [41,44 – 46].
staining, (3) achromatic tau-positive balloon neurons as Mutations in other genes, including CHMP2B and VCP,
seen in corticobasal degeneration, (4) intraneuronal and have also been found in familial FTD, but these appear to be
intranuclear inclusions that are ubiquitin-positive but tau- very rare cases [47,48]. Other genetic loci with significant
negative (FTLD-U), and (5) dementia lacking distinct his- linkage to FTD are actively being investigated.
topathology (DLDH) [25,26,31].
The clinical presentation of FTLD is also highly variable 3.1.3. Cerebral autosomal dominant arteriopathy with
and might comprise (1) FTD, exhibiting early decline in subcortical infarcts and leukoencephalopathy
social interpersonal conduct, loss of insight, emotional Cerebral autosomal dominant arteriopathy with subcor-
blunting, and mental inflexibility; (2) nonfluent progressive tical infarcts and leukoencephalopathy (CADASIL) is cur-
aphasia (also known as primary progressive aphasia), asso- rently the only known inherited form of vascular dementia
ciated with early loss of speech and language function; and [49]. The clinical spectrum includes recurrent transient isch-
(3) semantic dementia, showing loss of ability to name and emic attacks, migraine headaches, psychiatric disorders, and
understand words and to recognize faces and objects. The progressive dementia with associated deep white matter
term frontal variant of FTD (FTD-fv) has been used to lesions visible on brain imaging. Although initially thought
specifically refer to the behavioral syndrome of FTD (FTD- to be a rare disorder, CADASIL is likely underdiagnosed
bv) and temporal variant of FTD (FTD-tv) to denote the because the number of families being identified is continu-
clinical presentation of semantic dementia (26,27,32,33). In ously increasing [34]. The pathologic hallmark is accumu-
some FTD patients, there is coexisting motor neuron dis- lation of electron dense granules in the media of cerebral
ease, and these cases have been termed FTD-MND or FTD- arterioles. To date, more than 100 different mutations in the
ALS. FTD might also present clinically with movement NOTCH3 gene on chromosome 19p13 have been reported
disorders such as parkinsonism, corticobasal degeneration, in CADASIL patients of white and Asian descent [49 –52].
and progressive supranuclear palsy. About 95% are missense point mutations, and the others are
In a prevalence study of FTD from the Netherlands, 43% small deletions and one splice site mutation [3]. However,
have a positive family history [34]. In 1998, mutations in the mechanism by which these mutations become patho-
the microtubule-associated protein tau gene (MAPT) on genic remains unknown and warrants ongoing research.
chromosome 17 were found to be causative for a familial
form of FTD associated with parkinsonism (hence the name 3.1.4. Others
FTDP-17) [35–37]. About 10% to 30% of familial FTD There are a number of other genetic diseases associated
have a mutation in tau [38,39]. Tau is a phosphoprotein with progressive dementia such as Huntington’s disease, some
expressed in both central and peripheral nervous system and forms of spinocerebellar ataxia, hereditary amyloid angiopa-
is found predominantly in neuronal axons. The major phys- thy, fragile X syndrome, and familial prion diseases. Discus-
iologic function of tau is to bind to and stabilize microtu- sion of each of these diseases is beyond the scope of this
G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427 421

review. However, the principles behind genetic counseling and in up to 30 months of follow-up [6]. Nevertheless, all
testing in these disorders are similar to those discussed here. individuals eligible for PGT should be referred to specialists
in this field. These services are available at Canadian Col-
4. Genetic services for patients at risk for a causal lege of Medical Genetics’ accredited clinics listed on http://
mutation for a dementing illness ccmg.medical.org/clinical.html, and laboratories where mo-
lecular genetic tests are performed are listed on http://
Causative mutations refer to single gene mutations lead- ccmg.medical.org/molecular.html.
ing to expression of a disease. In dementias, most currently In principle, prenatal genetic diagnosis is an option for
known causative mutations are autosomal dominant families showing an autosomal dominant dementia with an
[2,10,53,54], and affected individuals are characterized by identified mutation, but this is not standard practice in Can-
an early-onset dementia (usually younger than age of 60 ada. Such concern is not unique to dementias but is faced by
years). Thus, biologic relatives of affected individuals with prospective parents for many other late-onset genetic disor-
a causal mutation are at risk for the mutation on the basis of ders such as familial breast cancer and familial hyperlipid-
the genetic closeness (DNA sharing) of the relative to the emia. Individual cases might need to be discussed with the
affected individual. appropriate medical ethics board [55]. Although there is
Currently, predictive genetic testing (PGT) is possible agreement that a young person carrying a family-specific
only for dementia families in whom a known causal muta- genetic mutation needs genetic counseling about the risks of
tion has been identified (ie, a specific mutation has been passing that mutation to a child, there is great concern
found in at least one affected family member). PGT is an among the genetics community about terminating a preg-
option for unaffected biologic relatives of the affected in- nancy when the disease onset will probably not occur until
dividual with the causal mutation [55]. For example, em- an older age. It is hoped that the progress in understanding
pirically, a full brother or sister of an affected individual the molecular pathogenesis of dementias might realistically
would have a 50% (1 in 2) chance of having the mutation, be expected to lead to effective treatment or ideally preven-
whereas a half-sibling would have a 25% (1 in 4) chance tion within the next few decades. For this reason the present
and a first cousin a 12.5% risk (1 in 8). In these rare families discussion will exclude prenatal genetic diagnosis.
(constituting less than 3% of all dementias), a specific ge-
netic change (mutation) travels through the family with the
5. Genetic counseling for unaffected individuals at risk
disease. These specific mutations can be shared by more
for familial dementia, but no family-specific causal
than one family or can be specific to a single family. In
mutation has been identified
families in whom a causal mutation is suspected but not
known, the options are to enroll in a research study or to pay For families with no known mutation but showing clin-
for testing by commercial laboratories in the USA in which ical presentation of an early-onset dementia with a family
screens are available for some of the more common muta- history compatible with an autosomal dominant mode of
tions. inheritance, genetic risk counseling must follow the predic-
The two most common mutations in AD (APP and tions of the autosomal dominant model for the specific
PSEN1) show virtually 100% penetrance, meaning that an family structure (eg, twin of an affected individual, off-
individual with the family-specific inherited mutation will spring of an affected parent, niece of an affected uncle
almost always develop the disease if he/she lives past the whose mother died well before the age of onset in the
usual age of disease onset for that family [56,57]. Pen- family, etc). It is critical to obtain as much documentation as
etrance of PSEN2 (a rare AD mutation on chromosome 1) possible on reportedly affected individuals to determine the
and tau mutations (for FTDP-17) is more variable, with “best estimate” clinical diagnosis. In large families, this
reports of an octogenarian carrier remaining cognitively might be very labor intensive, and the clinician might opt to
intact while passing on the disease to affected children refer the family to a specialized center, http://ccmg.medi-
[22,58]. However, in contrast to the situation for Hunting- cal.org/clinical.html. It is not uncommon to find that al-
ton’s disease [12], but similar to that for breast cancer [13], though families are initially believed to have familial AD,
an individual for whom PGT for familial dementia is an on follow-up they have heterogeneous etiologies with re-
option must fully understand that the absence of a family- spect to dementia (eg, alcohol-related dementia, Parkinson’s
specific mutation does not “protect” that individual from disease, depression). Such findings would significantly alter
AD or another dementia as a result of causes other than the the risk estimates for genetic counseling [55,59].
family-specific mutation. In other words, the absence of the On rare occasions an unaffected individual might still
mutation simply restores the level of risk to that of the want to be tested even though no DNA is available from an
general population (age and ethnically matched), which for affected family member. This type of genetic testing is not
a common disorder such as AD is intrinsically high. offered as a service in Canada but might be available com-
In several prospective studies of patients receiving PGT mercially. Individuals in these situations might opt for test-
for dementias, no negative emotional effects were observed ing through private companies, where one can screen for the
422 G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427

most common PSEN1 mutations. However, this is not en- current evidence suggests that it is a molecular chaperone
couraged, because proper counseling before and after the that helps promote the deposition of A␤ into amyloid
tests and assistance with interpretation of test results are not plaques [70].
often in place. Although the APOE e4 allele has been confirmed to be a
Because it is believed that novel genes for familial de- genetic susceptibility risk for AD, its use as a diagnostic tool
mentias, including late-onset AD, will be identified in the has been examined in several studies. The consensus was
future, families might have the opportunity to bank tissue that because of its low sensitivity and specificity, it should
material (through autopsy) and DNA (through a blood sam- not be used for general screening of patients at risk or for
ple or a buccal smear) from affected individuals for the diagnosis of AD [71–73]. On the other hand, several pro-
potential future benefit of other family members [57,58]. spective cohort studies suggest that it might significantly
This must of course be done with appropriate ethics ap- add to a predictive model in the assessment of patients
proval and signed informed consents. categorized as mild cognitive impairment (MCI) or cogni-
tively impaired not demented (CIND) whose risk of pro-
gression to dementia is high [69,71,74,75]. There are also
6. Genetic susceptibility factor in AD
three randomized controlled trials demonstrating that APOE
In the previous section we discussed causal genetic mu- genotype can affect therapeutic response to cholinesterase
tations. In contrast, the identification of a genetic suscepti- inhibitors [76 –78]. Therefore, APOE genotype might be
bility factor increases the likelihood (risk) of an individual used, with appropriate counseling, to stratify patients at risk
developing the disease within his/her lifetime, but there is of progression to dementia, and it might potentially have an
also a good chance that this will not occur. Thus, although important role in the assessment of therapeutic interventions
the proportion of individuals who develop the disease is that prevent or delay the progression of AD.
greater among those who have the specific genetic risk A recent randomized clinical trial conducted to examine
factor compared with those who do not, it is still possible for the impact of genetic risk assessment for adult children of
an individual to possess the risk factor but never develop the people with AD shows that revealing their APOE genotype
disease (low sensitivity), whereas some people who do not when presenting their risk of dementia did not increase
possess the risk factor might still develop the disease (low symptoms of depression or anxiety, whereas the genetic
specificity). information helped them make long-term life-planning de-
The literature review described above identified more cisions and motivated their engagement in risk reduction
than 1,000 publications related to the study of genetic risk activities [5]. It must be noted that this study used MSc
factors for AD, with 308 genes implicated [10]. However, to trained genetic counselors to convey the information. A
date, only one gene, APOE, has been consistently confirmed study examining the public perception on predictive testing
as a genetic susceptibility factor for AD. There are three of AD found that not only individuals with family history of
common polymorphisms in APOE, with e3 occurring in AD are interested, and many people without family history
about 75% of chromosomes in white populations, with e2 in of AD are just as interested in their genetic risks [79].
8% and e4 in 15% [60]. The association of the e4 allele in Clinicians must be prepared to discuss, at least in some
APOE with late-onset AD was first reported in 1993 detail, the topics of predictive testing in autosomal domi-
[61,62]. There is an apparent gene-dosing effect according nant families and genetic risk factors such as APOE when
to the actual genotype; the odds of developing AD for raised in the clinical setting.
carriers of one APOE e4 allele is 3.2 (95% confidence
interval [CI], 2.9 to 3.5) compared with e3/e3 individuals, 7. Conclusion
whereas the odds for homozygous e4/e4 carriers is 11.6
(95% CI, 8.9 to 15.4) [8,63]. In a meta-analysis with 5,930 Although more than 300 genes have been implicated in
patients and 8,607 controls, the e2 allele was shown to be the pathogenesis of AD and related dementias, at present
associated with a lower risk of AD (odds ratio, 0.6; 95% CI, only three (APP, PSEN1, PSEN2) have been confirmed to
0.5 to 0.8 in people with e2/e3 genotype in reference to be causatively associated with early-onset AD and two
e3/e3) [9]. Although APOE as a genetic risk factor for AD (MAPT and PGRN) with FTD. Causal genes have also been
has been demonstrated in populations of various ethnicity, identified for some diseases in which dementia is a major
the strength of association appears to be weaker among component of the phenotype, including CADASIL, Hun-
African Americans and Hispanics compared with whites, tington’s disease, and familial Creutzfeldt-Jakob disease.
whereas it is stronger among Japanese [9,64 – 69]. In addi- Genetic testing for diagnostic and predictive purposes is
tion, APOE exhibits interactions with age and gender, with only available as a clinical service for families in whom a
the highest odds occurring between ages 55 and 65 years, causal mutation has already been identified in an affected
and these odds are greater for female ⑀4 carriers compared family member. Predictive testing in minors and prenatal
with male. How APOE e4 actually affects the pathogenesis diagnosis are not within the normal patterns of practice in
of AD remains an area of intense investigation, although Canada, but they are dealt with on a case-by-case basis,
G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427 423

Fig 1. A hypothetical pedigree in which individuals are at risk for inheriting the disease genetic mutation.

often involving local medical ethics committees (http://cc- for determination of the best estimate clinical diagnosis
mg.medical.org/clinical.html.) (Grade A, Level 2). Testing for specific mutations might be
Commercial services for genetic testing are not endorsed arranged by the specialist through accredited genetic centers
because of the apparent need for longitudinal counseling of (http://ccmg.medical.org/clinical.html). In situations in
individuals having such tests with respect to both the inter- which a family-specific mutation has been identified, failure
pretation of the results and the reaction and adjustment after to find evidence for this mutation in another clearly affected
revelation of the findings. family member can be interpreted as being (1) phenocopy,
For AD the only confirmed genetic susceptibility risk (2) dementia as a result of other causes, (3) non-paternity,
factor is the APOE e4 allele. Nevertheless, because of its (4) non-maternity (eg, when the patient does not know about
low specificity and sensitivity, APOE genotyping should not an adoption).
be used as either a screening or a diagnostic tool. However,
in patients with mild cognitive impairment, research has 8.2. Predictive genetic testing for asymptomatic at-risk
shown that APOE genotype might be used to stratify pa- individuals with an apparent autosomal dominant
tients at risk of progression and to predict response to inheritance, and a family-specific mutation has been
therapy. Research is ongoing to identify novel genetic fac- identified
tors important in the risk of developing dementia.
8.2.1. Preamble
Gene mutations for dementia or neurodegenerative dis-
8. Summary of recommendations ease with dementia tend to be inherited in an autosomal
dominant pattern. The majority of mutations identified to
8.1. Genetic testing for confirmation of the etiology of a date show high penetrance and expressivity.
dementia
8.2.2. Recommendation 2
8.1.1. Preamble
With appropriate pre-testing and post-testing counseling,
Genetic mutations have been identified in certain types
PGT can be offered to at-risk individuals (Grade B, Level
of familial dementia, as discussed in this article.
2). Examples are (1) first-degree relatives of an affected
8.1.2. Recommendation 1 individual with the mutation (eg, children and siblings;
Symptomatic patients with a clear family history of a Figure 1, individuals III-2, III-3, and II-6); (2) first cousins
dementia should be referred to specialized dementia clinics of an affected individual if the common ancestors (parents
424 G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427

who were siblings) died before the average age of onset of 3). The ethical implications are complex and need to be
dementia in the family (eg, individual II-7 in Figure 1); (3) explored further. Requests might be considered on a case-
nieces and nephews of affected individuals whose parent by-case basis by the relevant medical ethics committee(s).
(sibling of the affected individual) died well before the
average age of onset of dementia in the family (eg, individ- 8.4. Ethical issues in genetic testing
ual III-4 in Figure 1); (4) PGT in minors is not generally 8.4.1. Preamble
offered in Canada, but it occasionally might be considered Guidelines still need to be developed for genetic testing
on a case-by-case basis by the relevant medical ethics com- in an affected individual who is demented and not cogni-
mittee(s) (eg, individual III-6 in Figure 1); (5) individuals tively competent to give consent. Such testing probably will
who are not at risk for the inherited disease do not require not benefit the individual directly but could impact on future
testing (eg, individual III-1 in Figure 1). generations if a known mutation is identified, or if new
8.2.3. Preamble mutations are discovered in the future through ongoing
De novo mutations can arise in an early-onset dementia research.
patient with a negative family history. 8.4.2. Recommendation 6
8.2.4. Recommendation 3 After careful genetic counseling with family members,
In young persons (60 years or younger) presenting with if it is decided that genetic testing and/or banking of
an early-onset dementia, it is sometimes worthwhile to test DNA (or autopsy material) for future studies is in the best
for the most common mutations on the basis of the best interest of family members, this might be done even
estimate diagnosis (eg, in early-onset AD, one might test for without the consent or assent of the affected individual
the most common mutations in PSEN1, APP) (Grade B, who is not cognitively competent, if consent from the
Level 2). If a mutation is identified, it would have direct family is given (Grade B, Level 2). However, extreme
implications for offspring of the individual (if a de novo care must be taken to minimize any distress the patient
mutation is assumed). It would also be important to test might experience while the sample is being obtained. In
other family members such as parents and siblings for cases in which family members, after extensive counsel-
possible non-penetrance of a mutation. ing, cannot agree on a plan, the case might have to go
before a medical ethics committee.
8.2.5. Preamble
Genetic counseling can be offered on the basis of the 8.4.3. Preamble
assumptions of an autosomal dominant model of inheritance A relatively rare but certainly real concern with ge-
in some families in whom affected individuals occur in at netic testing is the situation in which testing one family
least 2 generations, or there are at least 3 cases in the member can identify an “obligate carrier” who perhaps
collateral generation (siblings, first cousins) with insuffi- does not want this information. For example, in a family
cient information available on the previous generation, even with AD because of PS1 with a mean age of onset of 50
in the absence of an identified mutation. years, testing the daughter could in fact (assuming correct
parentage) identify the unaffected parent as an obligate
8.2.6. Recommendation 4
carrier (Figure 1, II-6 and III-7). The ethical dilemma is
Careful review of all available documentation (examina-
the right of the daughter to know versus the right of the
tion, clinical records, autopsy reports, etc) on reportedly
parent not to know.
affected relatives is essential to rule out the heterogeneous
causes of dementia in the elderly, including depression, 8.4.4. Recommendation 7
alcoholism, vascular dementia. If appropriate after review, Concerned family members should have appropriate pre-
genetic counseling should include the risks associated with test and post-test counseling available to be able to make
an autosomal dominant mode of inheritance as the upper- informed decisions (Grade B, Level 2). In cases of conflict
most risk. For the benefit of future generations, it is advis- among family members, medical ethics committees might
able to bank DNA and/or autopsy material from affected become involved.
individuals in such families in case novel gene mutations
might be discovered in the future (Grade B, Level 2). 8.5. Genetic susceptibility risk factors
8.3. Prenatal genetic testing 8.5.1. Preamble
APOE e4 increases risk of progression to AD in patients
8.3.1. Recommendation 5 with MCI/CIND. It also affects response to cholinesterase
Although prenatal testing for a known family mutation inhibitors.
associated with adult-onset dementia is technically possible
(eg, individual III-5 in Figure 1), this is not generally of- 8.5.2. Recommendation 8
fered in Canada. Currently, there is insufficient evidence to Genetic screening with APOE genotype in asymptomatic
recommend prenatal testing for dementia (Grade C, Level individuals in the general population is not recommended
G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427 425

because of the low specificity and sensitivity (Grade E, other inherited dementias. Am J Alzheimers Dis Other Demen
Level 2). 2005;20:233– 8.
[7] Evans GA. The Human Genome Project: applications in the diagnosis
8.5.3. Recommendation 9 and treatment of neurologic disease. Arch Neurol 1998;55:1287–90.
Genetic testing with APOE genotype is not recom- [8] Rubinsztein DC, Easton DF. Apolipoprotein E genetic variation and
Alzheimer’s disease. a meta-analysis. Dement Geriatr Cogn Disord
mended for the purpose of diagnosing AD because the
1999;10:199 –209.
positive and negative predictive values are low (Grade E, [9] Farrer LA, Cupples LA, Haines JL, Hyman B, Kukull WA, Mayeux
Level 2). R, et al. Effects of age, sex, and ethnicity on the association between
apolipoprotein E genotype and Alzheimer disease: a meta-analysis—
8.5.4. Recommendation 10 APOE and Alzheimer Disease Meta Analysis Consortium. JAMA
Although APOE e4 cannot be used as a diagnostic or a 1997;278:1349 –56.
screening test for AD, it has been consistently shown to be [10] Bertram L, McQueen M, Mullin K, Blacker D, Tanzi R. The AlzGene
a significant risk factor for progression from MCI/CIND to Database: Alzheimer Research Forum. Available at: http://www.alz-
AD (Grade B, Level 2). It might be used to stratify MCI/ gene.org. Accessed Feb 14, 2006.
CIND patients at risk for progression, and it might also be [11] Rocchi A, Pellegrini S, Siciliano G, Murri L. Causative and suscep-
tibility genes for Alzheimer’s disease: a review. Brain Res Bull
important in the assessment of therapeutic efficacy in clin- 2003;61:1–24.
ical research (Grade B, Level 1). [12] Tanzi RE, Hyman BT. Studies of amyloid beta-protein precursor
Note: Recommendations 1 and 10 received only weak expression in Alzheimer’s disease. Ann N Y Acad Sci 1991;640:
consensus (76% each), whereas recommendations 2 to 9 149 –54.
received strong consensus (greater than 80%). [13] Selkoe DJ, Podlisny MB, Joachim CL, Vickers EA, Lee G, Fritz LC,
et al. Beta-amyloid precursor protein of Alzheimer disease occurs as
110- to 135-kilodalton membrane-associated proteins in neural and
Author Disclosures nonneural tissues. Proc Natl Acad Sci U S A 1988;85:7341–5.
[14] Schellenberg GD, Pericak-Vance MA, Wijsman EM, Moore DK,
Ging-Yuek Robin Hsiung has received support from Gaskell PC Jr, Yamaoka LA, et al. Linkage analysis of familial
Novartis (Speaker fees), and Pfizer ⫹ Ono (Site Investigator Alzheimer disease, using chromosome 21 markers. Am J Hum Genet
on clinical trials). 1991;48:563– 83.
A. Dessa Sadovnick has received support from Biogen [15] Goate A, Chartier-Harlin MC, Mullan M, Brown J, Crawford F,
Fidani L, et al. Segregation of a missense mutation in the amyloid
Idec (Speaker, Meeting Participant), Serono (Speaker, precursor protein gene with familial Alzheimer’s disease. Nature
Meeting Participant), Teva Neuroscience (Funding for mul- 1991;349:704 – 6.
tiple sclerosis research, Speaker, Meeting Participant), and [16] Kang J, Lemaire HG, Unterbeck A, Salbaum JM, Masters CL, Grz-
Berlex Canada (Funding for multiple sclerosis research, eschik KH, et al. The precursor of Alzheimer’s disease amyloid A4
Speaker, Meeting Participant). protein resembles a cell-surface receptor. Nature 1987;325:733– 6.
[17] Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease:
progress and problems on the road to therapeutics. Science 2002;297:
Acknowledgments 353– 6.
[18] Selkoe DJ, Schenk D. Alzheimer’s disease: molecular understanding
A. Dessa Sadovnick is a recipient of a Michael Smith predicts amyloid-based therapeutics. Annu Rev Pharmacol Toxicol
Distinguished Scholar Award. 2003;43:545– 84.
Ging-Yuek Robin Hsiung is supported by funds from the [19] Brunkan AL, Goate AM. Presenilin function and gamma-secretase
activity. J Neurochem 2005;93:769 –92.
St. Paul’s Hospital Foundation.
[20] Selkoe DJ. The cell biology of beta-amyloid precursor protein and
presenilin in Alzheimer’s disease. Trends Cell Biol 1998;8:447–53.
References [21] Sudoh S, Kawamura Y, Sato S, Wang R, Saido TC, Oyama F, et al.
Presenilin 1 mutations linked to familial Alzheimer’s disease increase
the intracellular levels of amyloid beta-protein 1-42 and its N-termi-
[1] Bertram L, Tanzi RE. The current status of Alzheimer’s disease
nally truncated variant(s) which are generated at distinct sites. J Neu-
genetics: what do we tell the patients? Pharmacol Res 2004;50:385–
96. rochem 1998;71:1535– 43.
[2] Pastor P, Goate AM. Molecular genetics of Alzheimer’s disease. Curr [22] Rogaev EI, Sherrington R, Rogaeva EA, Levesque G, Ikeda M, Liang
Psychiatry Rep 2004;6:125–33. Y, et al. Familial Alzheimer’s disease in kindreds with missense
[3] Federico A, Bianchi S, Dotti MT. The spectrum of mutations for mutations in a gene on chromosome 1 related to the Alzheimer’s
CADASIL diagnosis. Neurol Sci 2005;26:117–24. disease type 3 gene. Nature 1995;376:775– 8.
[4] Neary D, Snowden J, Mann D. Frontotemporal dementia. Lancet [23] Sherrington R, Rogaev EI, Liang Y, Rogaeva EA, Levesque G, Ikeda
Neurol 2005;4:771– 80. M, et al. Cloning of a gene bearing missense mutations in early-onset
[5] Roberts JS, Cupples LA, Relkin N, Whitehouse P, Green RC, Group familial Alzheimer’s disease. Nature 1995;375:754 – 60.
ftRS. Genetic risk assessment for adult children of people with Alz- [24] Bird T, Knopman D, VanSwieten J, Rosso S, Feldman H, Tanabe H,
heimer’s disease: the Risk EValuation and Education for ALzhei- et al. Epidemiology and genetics of frontotemporal dementia/Pick’s
mer’s Disease (REVEAL) Study. J Geriatr Psychiatry Neurol 2005; disease. Ann Neurol 2003;54(Suppl 5):S29 –31.
18:250 –5. [25] Munoz DG, Dickson DW, Bergeron C, Mackenzie IR, Delacourte A,
[6] Molinuevo JL, Pintor L, Peri JM, Lleo A, Oliva R, Marcos T, et al. Zhukareva V. The neuropathology and biochemistry of frontotempo-
Emotional reactions to predictive testing in Alzheimer’s disease and ral dementia. Ann Neurol 2003;54(Suppl 5):S24 – 8.
426 G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427

[26] Kertesz A, McMonagle P, Blair M, Davidson W, Munoz DG. The temporal dementia linked to chromosome 17q21. Nature 2006;442:
evolution and pathology of frontotemporal dementia. Brain 2005; 920 – 4.
128(Pt 9):1996 –2005. [46] Mukherjee O, Pastor P, Cairns NJ, Chakraverty S, Kauwe JS, Shears S,
[27] Kertesz A. Pick complex: an integrative approach to frontotemporal et al. HDDD2 is a familial frontotemporal lobar degeneration with ubiq-
dementia—primary progressive aphasia, corticobasal degeneration, uitin-positive, tau-negative inclusions caused by a missense mutation in
and progressive supranuclear palsy. Neurologist 2003;9:311–7. the signal peptide of progranulin. Ann Neurol 2006;60:314 –22.
[28] Kertesz A, Munoz DG, Hillis A. Preferred terminology. Ann Neurol [47] Guyant-Marechal L, Laquerriere A, Duyckaerts C, Dumanchin C,
2003;54)Suppl 5):S3– 6. Bou J, Dugny F, et al. Valosin-containing protein gene mutations:
[29] Knopman DS, Petersen RC, Edland SD, Cha RH, Rocca WA. The clinical and neuropathologic features. Neurology 2006;67:644 –51.
incidence of frontotemporal lobar degeneration in Rochester, Minne- [48] Momeni P, Rogaeva E, Van Deerlin V, Yuan W, Grafman J, Tierney
sota, 1990 through 1994. Neurology 2004;62:506 – 8. M, et al. Genetic variability in CHMP2B and frontotemporal demen-
[30] Feldman H, Levy AR, Hsiung GY, Peters KR, Donald A, Black SE, tia. Neurodegener Dis 2006;3:129 –33.
et al. A Canadian cohort study of cognitive impairment and related [49] Joutel A, Corpechot C, Ducros A, Vahedi K, Chabriat H, Mouton P,
dementias (ACCORD): study methods and baseline results. Neuro- et al. Notch3 mutations in cerebral autosomal dominant arteriopathy
epidemiology 2003;22:265–74. with subcortical infarcts and leukoencephalopathy (CADASIL), a
[31] Mackenzie IR, Shi J, Shaw CL, Duplessis D, Neary D, Snowden JS, mendelian condition causing stroke and vascular dementia. Ann N Y
et al. Dementia lacking distinctive histology (DLDH) revisited. Acta Acad Sci 1997;826:213–7.
Neuropathol (Berl) 2006;112:551–9. [50] Suwanwela N, Srikiatkhachorn A, Tangwongchai S, Phanthumchina
[32] Perry RJ, Hodges JR. Differentiating frontal and temporal variant K. Mutation of the Notch 3 gene in a Thai cerebral autosomal
frontotemporal dementia from Alzheimer’s disease. Neurology 2000; dominant arteriopathy with subcortical infarcts and leukoencephalop-
54:2277– 84. athy family. J Med Assoc Thai 2003;86:178 – 82.
[33] Bozeat S, Gregory CA, Ralph MA, Hodges JR. Which neuropsychi- [51] Thomas NJ, Morris CM, Scaravilli F, Johansson J, Rossor M, De
atric and behavioural features distinguish frontal and temporal vari- Lange R, et al. Hereditary vascular dementia linked to notch 3
ants of frontotemporal dementia from Alzheimer’s disease? J Neurol mutations: CADASIL in British families. Ann N Y Acad Sci 2000;
Neurosurg Psychiatry 2000;69:178 – 86. 903:293– 8.
[52] Uyama E, Tokunaga M, Suenaga A, Kotorii S, Kamimura K, Taka-
[34] Rosso SM, Donker Kaat L, Baks T, Joosse M, de Koning I, Pijnen-
hashi K, et al. Arg133Cys mutation of Notch3 in two unrelated
burg Y, et al. Frontotemporal dementia in The Netherlands: patient
Japanese families with CADASIL. Intern Med 2000;39:732–7.
characteristics and prevalence estimates from a population-based
[53] Bird TD. Genetic factors in Alzheimer’s disease. N Engl J Med
study. Brain 2003;126(Pt 9):2016 –22.
2005;352:862– 4.
[35] Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, et
[54] Kalaria RN, Viitanen M, Kalimo H, Dichgans M, Tabira T. The
al. Association of missense and 5’-splice-site mutations in tau with
pathogenesis of CADASIL: an update. J Neurol Sci 2004;226:35–9.
the inherited dementia FTDP-17. Nature 1998;393:702–5.
[55] Sadovnick A, editor. Genetic counselling in dementia. Oxford: Black-
[36] Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson
well, 2002.
L, et al. Tau is a candidate gene for chromosome 17 frontotemporal
[56] Rogaeva E, Tandon A, St George-Hyslop PH. Genetic markers in the
dementia. Ann Neurol 1998;43:815–25.
diagnosis of Alzheimer’s disease. J Alzheimers Dis 2001;3:293–304.
[37] Spillantini MG, Murrell JR, Goedert M, Farlow MR, Klug A, Ghetti
[57] Rogaeva EA, Fafel KC, Song YQ, Medeiros H, Sato C, Liang Y, et
B. Mutation in the tau gene in familial multiple system tauopathy with
al. Screening for PS1 mutations in a referral-based series of AD cases:
presenile dementia. Proc Natl Acad Sci U S A 1998;95:7737– 41.
21 novel mutations. Neurology 2001;57:621–5.
[38] Stanford PM, Brooks WS, Teber ET, Hallupp M, McLean C, Halliday [58] Bird TD, Sumi SM, Nemens EJ, Nochlin D, Schellenberg G, Lampe
GM, et al. Frequency of tau mutations in familial and sporadic TH, et al. Phenotypic heterogeneity in familial Alzheimer’s disease:
frontotemporal dementia and other tauopathies. J Neurol 2004;251: a study of 24 kindreds. Ann Neurol 1989;25:12–25.
1098 –104. [59] Sadovnick AD. Genetic counselling and genetic screening for Alz-
[39] Poorkaj P, Grossman M, Steinbart E, Payami H, Sadovnick A, Noch- heimer’s disease and other dementias. Can J Neurol Sci 2001;
lin D, et al. Frequency of tau gene mutations in familial and sporadic 28(Suppl 1):S52–5.
cases of non-Alzheimer dementia. Arch Neurol 2001;58:383–7. [60] Zannis VI, Kardassis D, Zanni EE. Genetic mutations affecting hu-
[40] Iqbal K, Alonso Adel C, Chen S, Chohan MO, El-Akkad E, Gong man lipoproteins, their receptors, and their enzymes. Adv Hum Genet
CX, et al. Tau pathology in Alzheimer disease and other tauopathies. 1993;21:145–319.
Biochim Biophys Acta 2005;1739:198 –210. [61] Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M,
[41] Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity
R, Lindholm C, et al. Mutations in progranulin cause tau-negative binding to beta-amyloid and increased frequency of type 4 allele in
frontotemporal dementia linked to chromosome 17. Nature 2006;442: late-onset familial Alzheimer disease. Proc Natl Acad Sci U S A
916 –9. 1993;90:1977– 81.
[42] Gass J, Cannon A, Mackenzie IR, Boeve B, Baker M, Adamson J, et [62] Saunders AM, Roses AD. Apolipoprotein E4 allele frequency, isch-
al. Mutations in progranulin are a major cause of ubiquitin-positive emic cerebrovascular disease, and Alzheimer’s disease. Stroke 1993;
frontotemporal lobar degeneration. Hum Mol Genet 2006;15:2988 – 24:1416 –7.
3001. [63] Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell
[43] He Z, Bateman A. Progranulin (granulin-epithelin precursor, PC-cell- PC, Small GW, et al. Gene dose of apolipoprotein E type 4 allele and
derived growth factor, acrogranin) mediates tissue repair and tumor- the risk of Alzheimer’s disease in late onset families. Science 1993;
igenesis. J Mol Med 2003;81:600 –12. 261:921–3.
[44] Masellis M, Momeni P, Meschino W, Heffner R Jr, Elder J, Sato C, [64] Nishimura AL, Oliveira JR, Mitne-Neto M, Guindalini C, Nitrini
et al. Novel splicing mutation in the progranulin gene causing familial R, Bahia VS, et al. Lack of association between the brain-derived
corticobasal syndrome. Brain 2006;129(Pt 11):3115–23. neurotrophin factor (C-270T) polymorphism and late-onset Alz-
[45] Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, heimer’s disease (LOAD) in Brazilian patients. J Mol Neurosci
et al. Null mutations in progranulin cause ubiquitin-positive fronto- 2004;22:257– 60.
G-Y.R. Hsiung and A.D. Sadovnick / Alzheimer’s & Dementia 3 (2007) 418 – 427 427

[65] Raygani AV, Zahrai M, Doosti M, Javadi E, Rezaei M, Pourmot- [73] Tierney MC, Szalai JP, Snow WG, Fisher RH, Tsuda T, Chi H, et al.
abbed T. Association between apolipoprotein E polymorphism and A prospective study of the clinical utility of ApoE genotype in the
Alzheimer disease in Tehran, Iran. Neurosci Lett 2005;375:1– 6. prediction of outcome in patients with memory impairment. Neurol-
[66] Lai CL, Tai CT, Lin SR, Lin RT, Yang YH, Liu CK. Apolipoprotein ogy 1996;46:149 –54.
E in Taiwan Chinese patients with dementia. Dement Geriatr Cogn [74] Petersen RC, Smith GE, Ivnik RJ, Tangalos EG, Schaid DJ, Thi-
Disord 2003;16:208 –11. bodeau SN, et al. Apolipoprotein E status as a predictor of the
[67] Katzman R, Zhang MY, Chen PJ, Gu N, Jiang S, Saitoh T, et al. development of Alzheimer’s disease in memory-impaired individu-
Effects of apolipoprotein E on dementia and aging in the Shanghai als. JAMA 1995;273:1274 – 8.
Survey of Dementia. Neurology 1997;49:779 – 85. [75] Devanand DP, Pelton GH, Zamora D, Liu X, Tabert MH, Goodkind
[68] Tang MX, Stern Y, Marder K, Bell K, Gurland B, Lantigua R, et al. M, et al. Predictive utility of apolipoprotein E genotype for Alzheimer
The APOE-epsilon4 allele and the risk of Alzheimer disease among disease in outpatients with mild cognitive impairment. Arch Neurol
African Americans, whites, and Hispanics. JAMA 1998;279:751–5. 2005;62:975– 80.
[69] Hsiung GY, Sadovnick AD, Feldman H. Apolipoprotein E epsilon4 [76] Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R, Ferris
genotype as a risk factor for cognitive decline and dementia: data S, et al. Vitamin E and donepezil for the treatment of mild cognitive
from the Canadian Study of Health and Aging. CMAJ 2004;171: impairment. N Engl J Med 2005;352:2379 – 88.
863–7. [77] Bizzarro A, Marra C, Acciarri A, Valenza A, Tiziano FD, Brahe C, et
[70] Shobab LA, Hsiung GY, Feldman HH. Cholesterol in Alzheimer’s al. Apolipoprotein E epsilon4 allele differentiates the clinical re-
disease. Lancet Neurol 2005;4:841–52. sponse to donepezil in Alzheimer’s disease. Dement Geriatr Cogn
[71] Aggarwal NT, Wilson RS, Beck TL, Bienias JL, Berry-Kravis E, Disord 2005;20:254 – 61.
Bennett DA. The apolipoprotein E epsilon4 allele and incident Alz- [78] Poirier J, Delisle MC, Quirion R, Aubert I, Farlow M, Lahiri D, et al.
heimer’s disease in persons with mild cognitive impairment. Neuro- Apolipoprotein E4 allele as a predictor of cholinergic deficits and
case 2005;11:3–7. treatment outcome in Alzheimer disease. Proc Natl Acad Sci U S A
[72] Mayeux R, Saunders AM, Shea S, Mirra S, Evans D, Roses AD, et al. 1995;92:12260 – 4.
Utility of the apolipoprotein E genotype in the diagnosis of Alzheimer’s [79] Cutler SJ, Hodgson LG. To test or not to test: interest in genetic
disease: Alzheimer’s Disease Centers Consortium on Apolipoprotein E testing for Alzheimer’s disease among middle-aged adults. Am J
and Alzheimer’s Disease. N Engl J Med 1998;338:506 –11. Alzheimers Dis Other Demen 2003;18:9 –20.
Alzheimer’s & Dementia 3 (2007) 428 – 440

Toward a revision of criteria for the dementias


Kenneth Rockwooda,*, Rémi W Bouchardb, Richard Camiciolic, Gabriel Légerd
a
Department of Medicine (Geriatric Medicine & Neurology), Dalhousie University, Halifax, Nova Scotia, Canada
b
Department of Neurology, Laval University, Québeec, Québec, Canada
c
Department of Medicine (Neurology), University of Alberta, Edmonton, Alberta, Canada
d
Department of Medicine, University of Montreal, Montréal, Québec, Canada

Abstract This article critically considers current diagnostic criteria for dementia and reports recommen-
dations approved by at least 80% of experts attending the Third Canadian Consensus Conference on
the Diagnosis and Treatment of Dementia. There was consensus that many of the features proposed
as essential to a diagnosis of dementia in the 1980s no longer are relevant (for example, the
requirements for memory impairment, electroencephalogram and cerebrospinal fluid studies, age-
specific exclusions). In addition, other syndromes such as frontotemporal dementia have been
recognized and need to inform new dementia criteria. It is also recognized that a diagnosis of
depression need not exclude a dementia diagnosis. Other proposals, such as neuropathology
should be considered as additional evidence and not as a gold standard or that some people with
dementia have prolonged plateaus so that progressive decline need not be a criterion for
Alzheimer’s disease, were more controversial and did not receive similar support. Given the
evidence of the last three decades, there is merit in reconsidering the criteria by which dementia and
Alzheimer’s disease are diagnosed.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: Dementia; Alzheimer’s disease; Diagnostic criteria; Neuropathology; Frontotemporal dementia; Depression

1. Introduction: What can we know with certainty represent Grade B recommendations. Unless otherwise in-
about dementia diagnosis? dicated, the recommendations presented here were ap-
proved by at least 80% of the votes cast by the 45 experts
This report constitutes a background article for the Third
who attended the CCCDTD3, representing the experts on
Canadian Consensus Conference on the Diagnosis and
dementia in Canada from Geriatric Medicine, Neurology,
Treatment of Dementia (CCCDTD3) held in March 2006.
Psychiatry, Neuropsychology, and Family Practice. Be-
This article differs from the other 17 articles preceding it in
cause these recommendations were all debated and reached
that the goal was not so much to deliver recommendations
broad consensus within the entire Canadian academic com-
for current physician management of dementing diseases as
to point the way toward revising and improving the diag- munity, we hope that these proposals will initiate a serious
nostic criteria for different forms of dementia being used debate on revision of criteria during the coming years.
around the world. Because much of this knowledge is pro- As people age, most notice changes in their cognitive
posive, there is no possibility for randomized controlled functioning. Some changes are beneficial. Expert learners,
trials or even clinical-pathologic studies that compare sub- for example, have vast resources of prior information on
jects diagnosed according to different criteria. In keeping which to draw, thus allowing them readily to contextualize
with the fashion of evidence grading, these recommenda- and interpret complex information. But for most people, the
tions would all be rated as Level 2 or 3 evidence and would perceptible differences that occur with age are declines in
function, such as less efficient word finding or less efficient
execution of mental calculation.
*Corresponding author. Tel.: (902) 473-8687; Fax: (902) 473-1050. Until about the late 1960s, the differential diagnosis of
E-mail address: kenneth.rockwood@dal.ca memory-related complaints could operate with reasonable
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.07.014
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 429

certainty. It was recognized, for example, that although 2. Where have we come from in dementia diagnosis?
age-related changes in cognitive functioning were common,
2.1. Overview
these were not, in themselves, a disease. A clear separation
was made between such age-related benign changes and the There are many ways to define and classify the demen-
malignant changes of the dementias, especially Alzheimer’s tias. For example, 10 years ago, a comprehensive Cana-
disease (AD). These differences could be validated against dian review of the nosology of dementia noted four types
autopsy, which represented a gold standard for definitive of classification [1]: (1) histologic/neuropathologic (eg,
diagnosis. DLB); (2) etiologic (eg, vascular, infectious, but also
Since the late 1960s, tremendous progress has been made genetic); (3) brain region involved (eg, the focal atro-
in understanding dementia. Quite apart from progress in phies); and (4) clinical profile (eg, dementia with motor
molecular neurobiology, which is likely to impact how we signs, early-onset, rapidly progressive).
think about dementia in years to come, whole new dementia For the syndromic diagnosis, it is necessary to begin
syndromes have been identified (eg, frontotemporal demen- broadly. Dementia is typically defined as a clinical syn-
tia with relative memory sparing) as have new diagnostic drome of cognitive (historically memory is here privileged)
entities, such as dementia with Lewy bodies (DLB), or decline that is sufficiently severe to interfere with social or
human immunodeficiency virus–related dementia. occupational functioning. Routine clinical practice shows
Some of the progress in understanding cognition has that these cognitive and functional changes typically are
undermined prior certainties. Importantly, the comforting accompanied by changes in behavior and in personality, but
notion of the gold standard no longer seems valid. Although these have had variable prominence in diagnostic criteria.
neuropathology has an important contribution to make, the Dementia remains a clinical diagnosis, in which labora-
recognition that people with apparently identical lesions can tory or imaging tests as yet provide only supportive roles.
differ widely in their cognitive function and that differing Although some view the development of biomarkers as
sets of neuropathologic criteria yield differing estimates of essential for advancing the field [2], experience from other
dementias in the same brains each suggest that its role, like fields shows that unless there is very clear clinical correla-
that of other measures, best provides construct (cf. criterion) tion, the effect is to substitute the problem of the seeming
arbitrariness of clinical markers with the irrelevance of
validity. Similarly, the sharp line between normal aging and
biologic ones [3]. As such, the diagnosis is based on a
pathology is not as clear as it once was. It does not seem
careful history. In specialist practice, this is typically done
plausible that we should chiefly look only for single causes
by using a semistructured interview with an informant, eg,
of dementia; on a population basis, dementias with more
the Clinical Dementia Rating (CDR) [4] or the Brief Cog-
than one cause occur commonly, and our criteria must
nitive Rating Scale (BCRS) [5], and a detailed medical and
reflect this.
neurologic examination. The CDR and BCRS each incor-
In general, there are two approaches to handling concep-
porate neurocognitive testing, which otherwise can be
tual uncertainty. One is to clear out all the underbrush and
added by using instruments such as the Behavioural Neu-
find a firm ground on which new structures can be built,
rology Assessment [6]. At the syndromic level, the differ-
such as a new gold standard, and thus new, definitive cri- ential diagnosis classically includes the cognitive disorders
teria. A second approach is to recognize that conceptual as of delirium and depression, as well as the possibility of
well as instrumental uncertainty is part of this stage of our having no cognitive impairment. Since the proposal for
endeavors. Gold standards are unlikely to be widely avail- Benign Senescent Forgetfulness [7] the possibility of a
able, multiple causes of dementia will be a reality, and any diagnosis that falls short of dementia but cannot be regarded
new consensus criteria will need to anticipate revision. In as normal cognition must also be considered. In the Cana-
consequence, it will be necessary to build in methods of dian context, during the last dozen years this would include
information handling that allow the contribution of individ- terms such as cognitive impairment, no dementia (CIND)
ual items to be evaluated. [8 –10] and mild cognitive impairment (MCI) and its sub-
Our approach conforms most closely to the second, and types [11,12] and questionable and very early Alzheimer’s
thus we have chosen to recognize uncertainty rather than to disease [13].
effect a consensus that will assume it away. In what follows, To construct a differential diagnosis of the cause of the
we will briefly recapitulate the state of the art in dementia dementia, the starting point is typically the clinical presen-
diagnosis, enumerate the chief challenges that new data tation. In memory clinics, with the predominance of AD, the
have provided, and propose how to proceed in a way that usual question is whether the presentation conforms to the
allows pragmatic clinical decisions to be made without usual pattern seen with that illness. Otherwise, the usual
adopting a false sense of security. This is a philosophical considerations of the nature of onset, the duration of symp-
decision, not an evidence-based one. So too would be the toms, the degree of progression, and the presence of medical
decision to pursue any other path. and neurologic signs allow characteristic syndromes to be
430 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

identified. As detailed in the last Canadian consensus con- 3.1. Recommendation


ference on the assessment of dementia [14], many condi-
tions can confound the diagnosis of dementia including 3. An acute onset should not necessarily exclude a di-
communication disorders, comorbid psychiatric and medi- agnosis of AD. (This recommendation received weak
cal illnesses including developmental disorders. In addition, consensus support only.)
conditions such as transient global amnesia, subclinical ep-
ileptic syndrome, and medication toxicity challenge clini- The NINCDS-ADRDA [22] criteria for probable AD
cians to be alert to their possibility when evaluating patients have also been used for research purposes worldwide and
with cognitive complaints. A careful history, examination, remain applicable for clinical diagnosis in practice. They are
and longitudinal follow-up will elucidate whether an under- summarized as follows: dementia based on clinical exami-
lying or coexisting dementing process exists, so that rational nation and neuropsychological tests, with onset between
investigation and treatment can be carried out. ages 40 and 90 years; deficits in two or more areas of
cognition; progressive worsening of memory and other cog-
2.2. Current general criteria for dementia nitive functions; normal consciousness; supportive features
Even though the DSM IIIR criteria [15] have notionally include progressive deterioration of language, praxis, and
been displaced by DSM IV [16] and DSM-IV-TR, the most gnosis, impaired activities of daily living and behavior,
recent version [17], each remains in wide use. DSM-IV-TR positive familial history, normal cerebrospinal fluid (CSF),
requires impairment of memory and at least one of the nonspecific electroencephalogram (EEG), and progressive
following domains: language, praxis, gnosis, executive cerebral atrophy with time on neuroimaging. Other possible
functioning. These impairments need to be sufficiently se- features include some plateaus in the course, seizures in late
vere to impair social or professional life and must not occur stage, some neuropsychiatric and extrapyramidal symp-
as a consequence of a delirium or by another medical, toms, and normal computed tomography or magnetic reso-
neurologic, or psychiatric condition. Memory impairment, nance imaging (MRI) scans for age. Although CSF and
although present in most people with dementia, is not an EEG studies would not be part of a usual evaluation, they
essential requirement; instead, at least two cognitive do- sometimes can be indicated clinically. Neuroimaging that is
mains must be impaired, and of course the other criteria normal for age can be seen in established disease, especially
must be met. The presence of dementia without a dominant in its early stages. The discussion of EEG and CSF studies
memory complaint has been demonstrated in large cohorts as being “supportive” likely reflects an earlier era in which
of patients with dementia [18,19]. AD was believed to be a diagnosis of exclusion; therefore
these tests are no longer appropriate as considerations in
2.2.1. Recommendations usual practice. Similarly, there is no reason a priori to
1. Although memory impairment is an important part of exclude patients younger than 40 or older than 90.
most dementias, there are some dementias (subcorti- 3.2. Recommendations
cal ischemic dementia, primary progressive aphasia,
some other types of frontotemporal dementia [FTD]) 4. There is no need to suggest that routine EEG and CSF
in which the requirement for memory impairment studies help exclude a diagnosis of Alzheimer’s disease.
limits the sensitivity of a dementia diagnosis. The 5. There is no reason a priori to exclude patients younger
requirement for memory impairment should be dropped than 40 or older than 90.
from the criteria for dementia in favor of impairment in
at least two domains of cognitive function. Although the NINDS-ADRDA criteria are widely used,
2. The new onset of a mood disorder or behavioral when interpreted strictly they select so-called pure AD pa-
disturbances, in the face of changes in cognition, tients. In consequence, they exclude AD patients with
should be seen as supportive of a diagnosis of mixed disorders. Broader use of the DSM-IV-TR category
dementia. of “Dementia due to multiple etiologies” needs to be en-
couraged, with specification of the diseases contributing to
the dementia routinely spelled out.
3. Alzheimer’s disease
3.3. Recommendation
The diagnosis of AD rests on the standard criteria for
dementia being met and typically are seen with insidious
6. Broader use of the DSM-IV-TR category of “Demen-
onset and progressive decline. This approach generally is
tia due to multiple etiologies” needs to be encouraged,
nonproblematic, especially because memory is a predomi-
with specification of the diseases contributing to the
nant symptom in AD. The requirement for an insidious
dementia routinely spelled out.
onset should be seen as supportive but not absolute; uncom-
plicated AD can have an acute onset, for example, as with At present, standard diagnostic criteria for dementia do
coming on after a delirium [20,21]. not adequately recognize focal presentations. Although un-
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 431

common, focal presentations of dementia occur and can be rapid decline) can also occur. (In consequence, the
recognized in expert hands. One example is progressive visual requirement for continued cognitive decline should be
impairment [23,24]. At present, standard diagnostic criteria for dropped from the criteria for AD in clinical practice.)
dementia do not adequately recognize focal presentations. (Note: This recommendation by the authors was con-
sidered controversial and did not reach consensus at
3.4. Recommendation
the meeting.).
7. Revision of AD criteria should recognize the possi- In 1984 the AD consensus criteria proposed that neuro-
bility of focal presentations. pathology serve as the gold standard in the diagnosis of AD
and other causes of dementia. Much data since then suggest
In mild dementia, depression is common [25]. In contrast that such a view is naive. Instead, it is reasonable to view
to an earlier era in which it was thought to be important to the status of neuropathology not in providing criterion val-
rule out depression before making a diagnosis of AD, there idation but as another factor to consider, in short, in pro-
is now recognition of symptom overlap, perhaps reflecting viding construct validity. In 1996 Stuss and Levine, citing
shared pathophysiology. work that showed that some elderly people met neuropatho-
3.5. Recommendation logic criteria for dementia but in fact were not demented
[36,37], asked whether neuropathology alone could be con-
8. Depressive features should be recognized as concom- sidered as the absolute criterion for the classification of
itant in patients with AD and should not exclude a dementia. The question remains [38]. In addition, although
diagnosis. many studies have demonstrated a good correlation between
clinical severity and Braak’s neuropathologic staging, stud-
Much of the misery that arises from a diagnosis of AD ies in very elderly people suggest more that Braak’s staging
has less to do with the patient’s current state than with the represents a broad concept of the evolution of neurofibril-
prospect of disease progression. Should meaningful decline lary tangles rather than a precise hierarchical model of
be operationalized in the criteria? It is well-accepted that cognitive decline abilities near the upper limit of life [39].
patients with vascular dementia can have periods in which No area of scientific inquiry is without controversy, so the
their illness plateaus [26]. This has been less well-accepted fact that neuropathologists disagree about the neuropatho-
with AD, where the experience of relentless progression is logic diagnosis of dementia does not disqualify it as a useful
usual. Indeed, despite occasional suggestions that “the measure [40]. Still, although the classic criteria for AD have
course of dementia can be progressive, static or remittent” a high degree of accuracy (usually reported at ⫾90% [41–
[27], in neurodegenerative dementias, the idea of progres- 45], this has been disputed when referral and work-up bias
sion of cognitive impairment has been characterized as are taken into account [46].
“obvious and essential” [28]. Nevertheless, several groups
have observed that a significant minority of people with AD 3.7. Recommendation
(about 15%) [29 –31] exhibit comparatively little disease
progression. Two broad possibilities suggest themselves to 10. The classification of neuropathology as providing a
account for apparent stability of supposedly progressive definitive diagnosis of AD (or other cause of demen-
disorders, falsely negative progression as a result of insen- tia) should be dropped. Instead, semiquantitative and
sitivity of the measures to detect change over time or true quantitative criteria should be applied to pathologic
variability in progression [32,33]. Studies of the latter pos- findings to model neurodegenerative diseases in a
sibility have generally focused on people with more rapid manner that goes beyond simplistic dichotomous
progression (eg, as has been suggested in some studies of categories. (Note: This recommendation by the au-
DLB in comparison with AD without concomitant DLB thors was considered controversial and did not reach
[32]). In short, although dementia is typically considered to consensus at the meeting.).
be a progressive illness, relative stabilization in vascular
cognitive impairment (VCI), fluctuation in DLB, and vari- 4. Frontotemporal dementia
ability in the rates of progression of AD, with both very
rapid [34] and slowly progressive forms, need to be recog- People who are not dementia specialists could be for-
nized [35]. Each variation contributes to the nuance that given for feeling somewhat at sea when getting to grips with
operational criteria require to capture the variable phenom- FTD. There is particular heterogeneity in the frontal lobe
ena encountered in clinical practice [34]. dementias, both clinically and neuropathologically, and it
has led to many attempts at classification. None has yet
3.6. Recommendation proved to be entirely satisfactory. A 2001 consensus con-
ference popularized a three-part account [47]. First, the
9. Although most people with AD have a progressive account proposed an easy to recognize frontolobar degen-
cognitive decline, periods of relative stability (or of eration, which consists of difficulty in adapting to social
432 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

situations, with disinhibition, impulsivity, loss of insight, not recommended for primary clinical practice. Neverthe-
and other aspects of impaired social conduct. In addition, less, many recently published studies regarding the clinical
two language-dominant forms were suggested, primary pro- differential profile of these progressive aphasic disorders
gressive aphasia according to the classic description of have led to some degree of consensus for the diagnostic
Mesulam [48] and semantic dementia. The idea that each criteria that might be useful for research and clinical trials
involves a frontal/insular/anterior cingulate circuitry, with a [41,46,50,55,56].
left lateralized degeneration in the language disorders, is
attractive and has some empirical support [49,50]. Still, the 4.1. Recommendation
tripartite approach does not go without many notations. For
example, it does not say enough about the movement dis- 11. Specific frontal temporal lobar degeneration criteria
orders that commonly accompany FTD or about the syn- should be used, including criteria that recognize
drome of frontal lobe dementia with motor neuron disease. aphasic disorders and including primary progressive
Moreover, it is silent on cases in which memory problems are aphasia and semantic dementia, when these disor-
an early feature, such as very old adults. In such cases, differ- ders are considered. At present, no one set of FTD
entiation from atypical AD can be especially challenging. criteria captures all the variants encountered in prac-
One classification describes frontotemporal lobar degen- tice. The DSM-IV-TR category of “dementia due to
eration [41] with these core features: insidious onset and Pick’s disease” specifically is inadequate.
gradual progression, early impairment in social and per-
sonal conduct, early emotional blunting, and loss of insight;
5. Dementia with Lewy bodies
supportive features include examples of behavioral changes,
speech and language impairment, primitive reflexes, and It is vitally important that physicians have a good work-
extrapyramidal sign. Imaging studies are said to show pre- ing knowledge of DLB, because it is one dementia in which
dominant atrophy in the frontal and/or temporal region, common treatments can often be either especially bene-
supported by frontal lobe abnormalities on neuropsycholog- ficial (eg, the dramatic improvement sometimes seen with
ical testing. The latter can also show relative sparing of cholinesterase inhibitors [57]) or markedly detrimental
memory, spatial orientation, and perception. (the dramatic, even fatal worsening sometimes seen with
A Canadian contribution describes a set of disorders that neuroleptics [58]).
can be called Pick’s complex [51]. This classification em- The description of DLB has thus been an essential con-
phasizes the overlap that can be seen with the syndromes, tribution to modern dementia practice. Still, the initial two
both clinically and pathologically, and also draws attention sets of consensus criteria [58,59], while offering good spec-
to an akinetic rigid syndrome, which can be a common and ificity, were believed to be inadequately sensitive to the
disabling part of the evolution of these disorders and which protean manifestations of DLB, so that in late 2005 a third
is not always given its due. set of criteria were proposed [60]. These criteria added new
Before these more recent efforts, the Lund and Manches- clinical features and reclassified previously supportive fea-
ter criteria [52,53] were widely used, but they increasingly tures into two categories. Features are now separated into
appear to be displaced by more recent classifications. For those with a higher frequency of occurrence compared with
the present purposes, the elucidation of the clinical profile of other dementia syndromes (suggestive features) or those
FTD makes evident the need not to privilege memory over with a common occurrence but lower specificity (supportive
other cognitive disorders in defining dementia [54]. This features). For example, the former now includes severe
experience reinforces recommendation 1. neuroleptic sensitivity and rapid eye movement sleep be-
The rest of the criteria are variations on the DSM-IV-TR havior disorder (which can be manifested by occasionally
[17] approach and include the notion of impairment in dramatic verbal and motor outbursts that both often pre-date
social or occupational functioning, a decline compared with the dementia by years). In addition, imaging studies that
the previous level of functioning, a gradual onset and pro- suggest reduced striatal dopamine uptake are now seen as
gressive decline, and the deficits do not occur only during substantially supporting the diagnosis of DLB. Further-
delirium and cannot be explained by any other medical, more, new supportive features include early autonomic
neurologic, or psychiatric disorder. dysfunction, characteristic metaiodobenzylguanidine car-
It is important to note that primary progressive aphasia diac scintigraphy, and structural imaging that shows pres-
and semantic aphasia (also called semantic dementia) might ervation of the medial temporal lobe structures. The cri-
initially not meet dementia criteria. Initially those syn- teria also noted the common difficulty of appropriately
dromes are primarily aphasic disorders and will progress to quantifying fluctuation by history alone and suggested ways
a dementia syndrome, often with a dysexecutive syndrome to operationalize this common but sometimes clinically
and/or apraxia. Considering the initial focality of these challenging part of the diagnosis of DLB.
syndromes and the possibility of many underlying etiolo- The diagnosis of DLB in routine practice often varies
gies, specific operationalized criteria for these subgroups are according to the clinical service on which it is made [61].
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 433

Stereotypically, psychiatrists will more often see the phe- the cortex; those with striatal LBs first will have PD. For
notype in which hallucinations are common, neurologists now, PDD has yet to benefit from validated clinical criteria
where movement disorders predominate, and geriatricians for its diagnosis, although it is clear that dementia occurs
where loss of autonomy is especially a problem. Operation- commonly in advanced PD [65] and in older patients [66],
ally, this challenge is readily dealt with, but the conceptual so that about 30% to 40% of PD patents, on a population
issue of how to handle overlap, here between DLB and other basis, have dementia [65– 68].
disorders, again must be faced. The promoters of criteria for One additional consideration is the overlap between
any disorder that aim to select “pure” patients for research DLB/PDD and AD. Clinicopathologic reports of the extent
purposes need to recognize that this is not inherently more of overlap with AD yield variable estimates [69,70], but
“scientific” but rather a decision to trade off internal validity there is no reason to expect that PDD will be exempt from
for generalizability. Of note, the presence of a single sug- the mixed pathology seen with other dementias (eg, AD and
gestive feature in the absence of any of the core features vascular contribution) [71–74]. Nevertheless, PDD is a dif-
now allows a diagnosis of possible DLB. ferent clinical syndrome in the spectrum of DLB. In the
A technological approach to DLB diagnosis, which uses setting of DLB, it is particularly important to consider how
single photon emission computed tomography (SPECT) im- to deal with the relationship between DLB, AD, and PDD.
aging with a dopamine transporter scan (DaTScan) [62], is It seems reasonable to suggest that patients with predomi-
gaining popularity. In a multi-center European study, the nantly neocortical LBs will have the classic triad of hallu-
accuracy of classification between probable DLB, possible cinations, fluctuation, and parkinsonism, whereas striatal
DLB, and non-DLB (chiefly AD) was between 84% and LBs will give rise to the motor disorder first. In addition, the
87% [63]. Although impressive, the real value added by more that coincident AD pathology exists, the less likely it
such an approach would be its ability to distinguish not is that the presentation will be classic. There is a greater loss
between patients with clinically evident DLB and clinically of nigral DA neurons in PDD versus DLB, reflecting dif-
evident AD but to help diagnosis in cases that clinically are ferential initial focal involvement of the LBs. The single LB
rated as possible DLB. In such cases the distribution of scan disease model is probably best for neurobiologic studies,
results was bimodal, with roughly half having abnormal but the DLB/PDD distinction has advantages for classifying
scans and half not. In consequence, the role of DaTScans in patients. The 2005 DLB consensus criteria used an arbitrary
aiding usual practice is not yet clear. In terms of neuropsy- 1-year window for parkinsonism symptoms to precede cog-
chology, the clinical diagnosis is aided by recognizing that nitive changes in patients to be labeled DLB [60], so that a
visuoconstructional tasks best distinguish DLB from AD. delay beyond 1 year would suggest PDD. Note too that
On the ubiquitous Mini-Mental State Examination (MMSE) cognitive decline often can be detected with early PD but
[64] it has been suggested that copying a pentagon incor- well before dementia [75,76]. Because some patients with
rectly is more characteristic of DLB than AD, but it is PDD [77] as with DLB [78] respond well to cholinesterase
somewhat insensitive. In general, the more complex the inhibitors, in clinical practice it would seem important not to
visual task, the more sensitive it is for DLB, but of course withhold a treatment trial on the basis of deficiencies in the
specificity is traded off, because AD patients will also have current criteria. A full review of therapy for these conditions
trouble. is outside the mandate of this article.
5.1. Recommendation 6.1. Recommendation

12. Because of the present uncertainty with criteria for 13. Criteria for PDD are needed and likely are best
DLB, use of the broader 2005 consensus criteria will incorporated in new criteria that update those for
require additional data to test the hypothesis that dementia with LBs.
they have better accuracy. Pending new confirma-
tory studies, it is reasonable to continue to use the
1996 consensus criteria in routine clinical practice. 7. Vascular dementia and vascular
cognitive impairment

6. Parkinsonian dementia Vascular dementia (VaD) occurring with AD is common,


but “pure” vascular dementia appears to be uncommon [79].
As noted, parkinsonism is an important aspect of the A full review of the management of dementia with a cere-
clinical diagnosis of DLB. Obviously too, parkinsonism will brovascular component is presented in article by Bocti et al
be present in the dementia seen with Parkinson’s disease (this issue). In consequence, there are both conceptual and
(PD), which is termed Parkinson’s disease dementia (PDD). pragmatic difficulties in trying to portray VaD as a distinct
It seems likely that the difference between DLB and PDD entity. Of the criteria now in use (the NINDS-AIREN [80]
reflects where LBs first accumulate. Patients with halluci- and ADTCC criteria [81] have been especially influential),
nations, inattention, and cognitive impairment have them in no set is perfect. In practice, most are insensitive [82], and
434 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

different sets of criteria give differing estimates of who has Recent consensus guidelines published in 2005 focused
VaD [82– 86]. Vascular lesions are found in many dementia on clinical documentation of one or more of the character-
patients, including those with otherwise classic AD, where istic symptoms of idiopathic NPH in combination with a
they are often detected only by routine neuroimaging brain imaging study demonstrating nonobstructive ventric-
[87]. On the other hand, patients with only vascular ular enlargement disproportionate to cerebral atrophy. It
pathology as the cause of their dementia have been un- was suggested that probable, possible, and unlikely NPH
common in many series [44,87]. Most patients have classifications be used. Adjunct tests, involving transient
mixed pathology, so that patients with vascular lesions removal of CSF via lumbar puncture or lumbar drain, were
commonly have evidence of other neurodegenerative dis- advocated for the dual purpose of adding to diagnostic
orders [38,71,72]. It is also important to note that many certainty and assisting in prognostication about response to
patients with positive imaging do not have a stroke his- treatment [95].
tory [82,88] and vice versa [89]. A common clinical scenario is the patient referred for
The NINDS-AIREN criteria for VaD are problematic, evaluation for NPH not on the basis of the characteristic
because they adhere to a multiple infarction model and also clinical triad but on the basis of the neuroimaging that has
privilege memory impairment among cognitive deficits, been interpreted as compatible with it and the suggestion
when in fact executive dysfunction often predominates [90]. that NPH be ruled out. The presence of central atrophy,
Although the requirement for a temporal relationship be- associated white matter changes, the extent of sulcal efface-
tween dementia and stroke makes sense for cases of post- ment and the pattern of the gyri in relation to these features
stroke dementia, it does not apply to the cases (which in each offer clues from the scan in the clinically nonevident
memory clinics typically will be the majority [82]) in which case. Such patients require careful follow-up.
cognitive deterioration might be slowly progressive rather Typically, the differential diagnosis of NPH includes
than stepwise [91]. Moreover, it does not acknowledge the obstructive hydrocephalus, multiple systems atrophy (asso-
common incidence of silent infarctions [92]. ciated with ataxia and incontinence), vascular gait impair-
Against this background, the proposal to expand the ment, and AD with gait impairment. Where the clinical triad
concept to one of VCI makes sense [93]. Still, the broad is the starting point, the finding of hydrocephalus on imag-
concept has meant that subtypes are necessary for clinical ing is predictive of shunt responders [96]. Other clinical
recognition. Three subtypes in particular are recognized: features that suggest a good response include a shorter
VCI–no dementia (VCI-ND), a subcortical vascular demen- duration of illness, lack of prominent cortical cognitive
tia with a prominent dysexecutive profile and white matter deficits, and good response to a removal of CSF (by external
changes on neuroimaging, and VaD with multiple or single or internal lumbar drainage or by single or repeated lumbar
strategic infarcts. These profiles appear to be readily recog- puncture) [97]. As with other dementias, mixed pathologies
nizable in memory clinic practices [82,89]. are not uncommon. Thus neuropsychological demonstration
of anomia or marked medial temporal lobe atrophy on MRI
might suggest AD and a poorer potential for response to
8. Recommendation shunting. Although longer duration of cognitive impairment
is a predictor of poor response, patients might respond
14. The proposal to develop new criteria for VCI has despite the presence of negative prognostic indicators [98].
merit. The criteria should move away from the mod- Ancillary tests are less helpful in predicting response. In
els of post-stroke dementia and multi-infarct demen- particular, intracranial pressure monitoring and ventricular
tia. Moreover, they should emphasize opportunities cisternography are not recommended routinely [95].
for prevention of impairment that arise as a conse-
quence of potentially modifiable cerebrovascular 9.1. Recommendation
disease.
15. The diagnosis of dementia caused by NPH should
use the recent 2005 consensus criteria, recognizing
9. Normal pressure hydrocephalus their limitations. In uncertain cases referral to a
Normal pressure hydrocephalus (NPH) is not a particu- specialized clinic is recommended.
larly common cause of dementia, but as a potentially treat-
able syndrome, it has special importance. Classically, it is
10. Dementias associated with infectious diseases
characterized as dementia seen with gait impairment and
with urinary urgency or incontinence [94]. NPH can be Several infectious diseases might be associated with a
idiopathic or secondary, usually as a result of hemorrhage progressive dementia. Human immunodeficiency virus pro-
or other brain injury. It remains relatively understudied, duces a so-called subcortical dementia that can sometimes
making the development of evidence-based guidelines occur in the absence of a history of known human immu-
difficult [95]. nodeficiency virus or acquired immune deficiency syn-
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 435

drome [99]. Neurosyphilis, although now rare, is potentially able sensitivity and lack of specificity [104]. Diffusion-
treatable [100]. Acute viral encephalitis can leave patients weighted MRI might have superior sensitivity compared
with dementia and can sometimes present in an indolent with EEG and superior specificity compared with CSF ex-
fashion. Clues to acute and subacute central nervous system amination [105]. In general, sporadic CJD is inexorably
infection include headache, fever, seizures, systemic com- progressive, resulting in death within a year; however, the
plaints, infection of peripheral tissue, rapid progression, course of sporadic CJD can be considerably longer [106]. A
focal neurologic features (in the setting of abscesses with long duration is common in the autosomal dominant prion
parenchymal involvement), and travel to endemic areas. disease, Gerstmann-Straussler-Scheinker syndrome. Variant
Immunosuppression predisposes to infections in general, CJD is a progressive neuropsychiatric disorder ultimately
and specific disorders, such as progressive multifocal leu- leading to ataxia, dementia, and myoclonus (or chorea)
koencephalopathy, are important to consider in this setting. without the typical EEG appearance of CJD or the propor-
Worldwide tuberculosis and neurocysticercosis are common tion with elevation in the 14-3-3 protein [107]. Young-onset
infections that have a predilection for the central nervous sporadic CJD cases share features of a long neuropsychiat-
system. Lyme disease is an infection that should be consid- ric prodrome with variant CJD [108].
ered in individuals with appropriate symptoms, such as a Spongiform changes are also found in variant CJD,
rash and polyarthritis, from an endemic area. Creutzfeldt- where they are found in the pulvinar in 70% of cases [107].
Jakob disease (CJD), Whipple’s disease, and other infec- It has been proposed that MRI be included among diagnos-
tions associated with dementia generally offer clues on the tic criteria, given reasonable sensitivity and specificity in
clinical examination as noted above. Other infections have some series [109]. Periodic sharp complexes on EEG
abnormalities on CSF examination, including elevated pro- changes are characteristic but can occur transiently or late in
tein, pleocytosis, and evidence for microorganisms on ex- the course of disease and are thus insensitive [110]. False
amination or culture. The polymerase chain reaction is use- positives include cases of DLB, AD, and VaD.
ful for amplifying genomic material for specific infections,
including herpes simplex and Whipple’s disease. 11.1. Recommendation

10.1. Recommendation 17. Documentation of rapid progression should raise the


possibility of CJD and suggests prompt referral.
16. In young patients or those with atypical presenta- MRI should be done in the appropriate clinical
tions, systemic features, or relevant risk factors, in- setting.
fectious causes should be borne in mind in the dif-
ferential diagnosis of dementia. Focal cognitive
deficits raise this possibility, even when they do not 12. Other dementias with specific etiologies
meet traditional dementia criteria. Such features Nondegenerative dementias caused by acquired systemic
should prompt specialist referral. or metabolic derangements and genetic conditions can be
diagnosed if they otherwise meet DSM-IVTR criteria and
11. Creutzfeldt-Jakob disease are often associated with a specific etiology. Genetic con-
ditions are rare, tend to occur in younger individuals, and
CJD is a rapidly progressive dementia that can only be are often inherited in an autosomal recessive fashion. Hun-
definitively diagnosed by a brain tissue examination that tington’s disease is common among young-onset dementias
shows prion proteins with associated spongiform changes. and is associated with an autosomal dominant family his-
CJD can be sporadic or familial (associated with mutations tory. Cerebral autosomal dominant arteriopathy with sub-
in gene coding for the prion protein) or transmissible. Trans- cortical infarcts and leukoencephalopathy (CADASIL) is an
missible forms of CJD include iatrogenic and variant CJD. example of an autosomal dominant VaD that illustrates the
Criteria for probable CJD have been proposed (http://www. overlap between genetic and vascular conditions [111].
eurocjd.ed.ac.uk/def.htm) and include typical EEG features Acquired conditions of acute onset might be consistent
with at least two of the following clinical features: myoclonus, with vascular causes of dementia, including cerebrovascular
visual or cerebellar signs, pyramidal/extrapyramidal signs, or disease [112], vasculitis, and amyloid angiopathy (with or
akinetic mutism. The clinical diagnosis can also be made without hemorrhage) [112,113]. Nonvascular causes can
with a history of a rapidly progressive dementia with dura- also give rise to an acute-onset dementia [20]. Although
tion of less than 2 years and at least two of the above clinical delirium can occur in the absence of dementia, they com-
features, with a positive 14-3-3 test suggested in recent monly co-occur; dementia is a risk factor for delirium and
iterations of the criteria. The clinical criteria might be in- vice versa [114,115]. Rapid progression of symptoms can
sensitive in early cases [101] that have had pathologic be seen in AD and DLB [116,117]. Dementia associated
confirmation with accepted criteria [102,103]. The utility of with motor neuron disease can also have a rapid course
the 14-3-3 test has come into question because of its vari- [118]. Dramatic fluctuations in the level of consciousness
436 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

can mimic an acute stroke-like onset and are characteristic Award Holder), and the Nova Scotia Department of Health
of DLB and can be seen in PDD [119]. Other syndromes (Consultant).
that should be considered in the differential diagnosis of a Rémi W. Bouchard has received support from Pfizer
rapidly progressive dementia include viral encephalitis, Canada (Consultant, Advisory Board Member, Speaker,
paraneoplastic (limbic) encephalitis, central nervous system Lecturer, Clinical Trials), Novartis (Consultant, Advisory
cancer, Hashimoto’s encephalitis, other autoimmune disor- Board Member, Speaker, Lecturer, Clinical Trials), Janssen
ders (including anti-phospholipid syndrome, systemic lupus Ortho Inc. (Consultant, Advisory Board Member, Speaker,
erythematosus, sarcoidosis, and non-vasculitic autoimmune Lecturer, Clinical Trials), and Lundbeck Canada (Consul-
meningoencephalitis) [120,121], infections, and metabolic tant, Advisory Board Member, Speaker, Lecturer, Clinical
disorders. Neuroimaging and other specific studies can be Trials).
helpful in identifying ischemic changes and in evaluating Richard Camicioli has received support from Novartis
other brain lesions. (Grant, Speaker), Pfizer (Grant, Medical Advisory Board
Other dementias for which broad experience is lacking, Member, Speaker), Janssen-Ortho (Medical Advisory
which have variably valid diagnostic criteria, include argyr- Board Member, Speaker), and GlaxoSmithKline (Medical
ophilic grain disease [122], neurofilament inclusion disease Advisory Board Member).
[123], late-life hippocampal sclerosis [124 –126], corticoba- Gabriel Léger has received support from Janssen Ortho
salganglionic degeneration, progressive supranuclear palsy, Inc. (Meeting Participant), Novartis (Meeting Participant,
Whipple’s disease [127], Huntington’s disease, pantothe- Speaker), and Pfizer Canada (Meeting Participant, Speaker).
nate kinase–associated neurodegeneration [128], autoim-
mune channelopathies [129], leukodystrophies [130], and
Acknowledgments
superficial siderosis [131].
Kenneth Rockwood is supported by an Investigator
13. Concluding comments Award from the Canadian Institutes of Health Research
(CIHR) and the Dalhousie Medical Research Foundation as
Some of what is needed to advance the diagnosis of the Kathryn Allen Weldon Professor of Alzheimer’s Re-
dementia is consensus not on a final set of criteria but on search. He receives research support from the CIHR and
recording enough information that will allow future criteria from the Alzheimer Society of Canada.
to be developed on the basis of data—to yield “evidence- Richard Camicioli is supported by an operating grant
based not eminence-based” recommendations. In Canada from the CIHR.
this has been the rationale behind the ACCORD [19] and
CIVIC [82] studies.
References
Similarly, clinical pathologic correlation studies should
examine the pathologic markers as quantitative traits that [1] Stuss DT, Levine B. The dementias: nosological and clinical factors
might have a dose-response and topographic-response rela- related to diagnosis. Brain Cogn 1996;31:99 –113.
tionship to various aspects of the dementia syndrome. For [2] Wiltfang J, Lewczuk P, Riederer P, Grunblatt E, Hock C, Scheltens
example, many disputes about neuropathologic markers P, et al. Consensus paper of the WFSBP Task Force on Biological
now cast them as either/or [132], so that the possibility of Markers of Dementia: the role of CSF and blood analysis in the early
and differential diagnosis of dementia. World J Biol Psychiatry
their use in the aggregate, for example, as an index variable 2005;6:69 – 84.
[133], has been less well pursued. [3] Rothwell PM. External validity of randomised controlled trials: “to
Our emphasis is on refining criteria as we go and on whom do the results of this trial apply?” Lancet 2005;365:82–93.
anticipating future developments that will change the way [4] Morris JC. The Clinical Dementia Rating (CDR): current version
we think about cognitive disorders. This strikes us as ap- and scoring rules. Neurology 1993;43:2412– 4.
[5] Reisberg B, Ferris SH. Brief Cognitive Rating Scale (BCRS). Psy-
propriate to the optimism inspired by the last 10 years of chopharmacol Bull 1988;24:629 –36.
progress. [6] Darvesh S, Leach L, Black SE, Kaplan E, Freedman M. The behav-
ioural neurology assessment. Can J Neurol Sci 2005;32:167–77.
[7] Kral VA. Senescent forgetfulness: benign and malignant. Can Med
Author Disclosures Assoc J 1962;86:257– 60.
[8] Ebly EM, Hogan DB, Parhad I. Cognitive impairment in the non-
Kenneth Rockwood has received support from Pfizer demented elderly: results from the Canadian Study of Health and
Canada (Consultant, Medical Advisory Board Member, Aging. Arch Neurol 1995;52:612–9.
Speaker), Janssen Ortho Inc. (Consultant, Medical Advisory [9] Graham JE, Rockwood K, Beattie BL, Eastwood R, Gauthier S,
Board Member, Research Grant Holder), Novartis (Speaker, Tuokko H, et al. Prevalence and severity of cognitive impairment
Consultant), Lundbeck Canada (Advisory Board Member), with and without dementia in an elderly population. Lancet 1997;
349:1793– 6.
Merck Frosst (Speaker), CIHR (Research and Career Award [10] Tuokko H, Frerichs R, Graham J, Rockwood K, Kristjansson B, Fisk
Holder), Alzheimer Society of Canada (Research Grant J, et al. Five-year follow-up of cognitive impairment with no dementia.
Holder), Dalhousie Medical Research Foundation (Career Arch Neurol 2003;60:577– 82.
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 437

[11] Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins PV, [31] Holmes C, Lovestone S. Long-term cognitive and functional decline
et al. Current concepts in mild cognitive impairment. Arch Neurol in late onset Alzheimer’s disease: therapeutic implications. Age
2001;58:1985–92. Ageing 2003;32:200 – 4.
[12] Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund [32] Galasko DR, Gould RL, Abramson IS, Salmon DP. Measuring
LO, et al. Mild cognitive impairment– beyond controversies, to- cognitive change in a cohort of patients with Alzheimer’s disease.
wards a consensus: report of the International Working Group on Stat Med 2000;19:1421–32.
Mild Cognitive Impairment. J Intern Med 2004;256:240 – 6. [33] Ashford JW, Schmitt FA. Modeling the time-course of Alzheimer
[13] Morris JC, Storandt M, Miller JP, McKeel DW, Price JL, Rubin EH, dementia. Curr Psychiatry Rep 2001;3:20 – 8.
et al. Mild cognitive impairment represents early-stage Alzheimer [34] Soto ME, Gillette-Guyonnet S, Vellas B; The REAL.FR group.
disease. Arch Neurol 2001;58:397– 405. Rapid cognitive decline: searching for a definition and predictive
[14] Patterson CJ, Gauthier S, Bergman H, Cohen CA, Feightner JW, factors among elderly with Alzheimer’s disease. J Nutr Health
Feldman H, et al. The recognition, assessment and management of Aging 2005;9:158 – 61.
dementing disorders: conclusions from the Canadian Consensus [35] Fisk JD, Rockwood K. Outcomes of incident mild cognitive impair-
Conference on Dementia. CMAJ 1999;160:S1–15. ment in relation to case definition. J Neurol Neurosurg Psychiatry
[15] American Psychiatric Association, Committee on Nomenclature and 2005;76:1175–7.
Statistics. Diagnostic and statistical manual of mental disorders, 3rd [36] Katzman R, Terry R, DeTeresa R, Brown T, Davies P, Fuld P, et al.
ed, revised. Washington, DC: American Psychiatric Association; Clinical, pathological, and neurochemical changes in dementia: A
1987. subgroup with preserved mental status and numerous neocortical
[16] American Psychiatric Association. Diagnostic and statistical manual plaques. Ann Neurol 1988;23:138 – 44.
of mental disorders. 4th ed. Washington, DC: American Psychiatric [37] Crystal H, Dickson D, Fuld P, Masur D, Scott R, Mehler M, et al.
Association; 1994. Clinico-pathologic studies in dementia: nondemented subjects with
[17] American Psychiatric Association. Diagnostic and statistical manual pathologically confirmed Alzheimer’s disease. Neurology 1988;38:
of mental disorders (IV-TR). Washington, DC: American Psychiat- 1682– 87.
ric Association; 2000. [38] Neuropathology Group. Medical Research Council Cognitive Func-
[18] Lopez OL, Becker JT, Kaufer DI, Hamilton RL, Sweet RA, Klunk tion and Aging Study: athological correlates of late-onset dementia
W, et al. Research evaluation and diagnosis of probable Alzheimer’s
in a multicentre, community-based population in England and
disease over the last two decades: pt 1. Neurology 2000;55:1854 –
Wales—Neuropathology Group of the Medical Research Council
62.
Cognitive Function and Ageing Study (MRC CFAS). Lancet 2001;
[19] Feldman H, Levy AR, Hsiung GY, Peters KR, Donald A, Black SE,
357:169 –75.
et al. A Canadian cohort study of cognitive impairment and related
[39] Gold G, Bouras C, Kövari E, Canuto A, Glaría BG, Malky A, et al.
dementias (ACCORD): study methods and baseline results. Neuro-
Clinical validity of Braak neuropathological staging in the oldest-
epidemiology 2003;22:265–74.
old. Acta Neuropathol (Berl) 2000;99(5):579 – 82.
[20] King P, Davichand P, Rockwood K. Dementia of acute onset in the
[40] Nagy Z, Esiri MM, Hindley NJ, Joachim C, Morris JH, King EM,
Canadian Study of Health and Aging. Int Psychogeriatr 2005;17:
et al. Accuracy of clinical operational diagnostic criteria for Alz-
451–9.
heimer’s disease in relation to different pathological diagnostic
[21] King P, Wang Y, Rockwood K. Cognitive impairment of acute
protocols. Dement Geriatr Cogn Disord 1998;9:219 –26.
onset. Am J Geriatr Psychiatry 2006;14:893– 6.
[41] Blacker D, Albert MS, Bassett SS, Go RC, Harrell LE, Folstein MF.
[22] McKhann G, Drachman D, Folstein M, Katzman R, Price D,
Reliability and validity of NINCDS-ADRDA criteria for Alzhei-
Stadlan EM. Clinical diagnosis of Alzheimer’s disease: Rrport of
the NINCDS-ADRDA Work Group under the auspices of De- mer’s disease: the National Institute of Mental Health Genetics
partment of Health and Human Services Task Force on Alzhei- Initiative. Arch Neurol 1994;51:1198 –204.
mer’s Disease. Neurology 1984;34:939 – 44. [42] Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S,
[23] Renner JA, Burns JM, Hou CE, McKeel DW Jr, Storandt M, Morris et al. Frontotemporal lobar degeneration: a consensus on clinical
JC. Progressive posterior cortical dysfunction: a clinicopathologic diagnostic criteria. Neurology 1998;51:1546 –54.
series. Neurology 2004;63:1175– 80. [43] Rockwood K, Howard K, Thomas VS, Mallery L, MacKnight C,
[24] Tang-Wai DF, Graff-Radford NR, Boeve BF, Dickson DW, Parisi Sangalang V, et al. Retrospective diagnosis of dementia using an
JE, Crook R, et al. Clinical, genetic and neuropathological charac- informant interview based on the Brief Cognitive Rating Scale. Int
teristics of posterior cortical atrophy. Neurology 2004;63:1168 –74. Psychogeriatr 1998;10:53– 60.
[25] Visser PJ, Verhey FR, Ponds RW, Kester A, Jolles J. Distinction [44] Holmes C, Cairns N, Lantos P, Mann A. Validity of current clinical
between preclinical Alzheimer’s disease and depression. J Am Geri- criteria for Alzheimer’s disease, vascular dementia and dementia
atr Soc 2000;48:479 – 84. with Lewy bodies. Br J Psychiatry 1999;174:45–50.
[26] Hachinski VC, Iliff LD, Zilhka E, Du Boulay GH, McAllister VL, [45] Knopman DS, DeKosky ST, Cummings JL, Chui H, Corey-Bloom
Marshall J, et al. Cerebral blood flow in dementia. Arch Neurol J, Relkin N, et al. Practice parameter: diagnosis of dementia (an
1975;32:632–7. evidence-based review)—report of the Quality Standards Subcom-
[27] Gustafson L. What is dementia? Acta Neurol Scand Suppl 1996; mittee of the American Academy of Neurology, Neurology 2001;
168:22– 4. 56:1143–53.
[28] Graeber M, Muller U. Dementia with Lewy bodies: disease concept [46] Bowler JV, Munoz DG, Merskey H, Hachinski V. Fallacies in the
and genetics. Neurogenetics 2003;4:157– 62. pathological confirmation of the diagnosis of Alzheimer’s disease.
[29] Bowler JV, Munoz DG, Merskey H, Hachinski V. Factors affecting J Neurol Neurosurg Psychiatry 1998;64:18 –24.
the age of onset and rate of progression of Alzheimer’s disease. [47] McKhann GM, Albert MS, Grossman M, Miller B, Dickson D,
J Neurol Neurosurg Psychiatry 1998;65:184 –90. Trojanowski JQ, et al. Clinical and pathological diagnosis of fron-
[30] Perrault A, Wolfson C, Egan M, Rockwood K, Hogan DB. Prog- totemporal dementia: report of the Work Group on Frontotemporal
nostic factors for functional independence in older adults with mild Dementia and Pick’s Disease. Arch Neurol 2001;58:1803–9.
dementia: results from the Canadian Study of Health and Aging. [48] Mesulam MM. Primary progressive aphasia: differentiation from
Alzheimer Dis Assoc Disord 2002;16:239 – 47. Alzheimer’s disease. Ann Neurol 1987;22:533– 4.
438 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

[49] Seeley WW, Bauer AM, Miller BL, Gorno-Tempini ML, Kramer [71] Erisi MM, Wilcock GK, Morris JH. Neuropathological assessment
JH, Weiner M, et al. The natural history of temporal variant fron- of the lesions of significance in vascular dementia. J Neurol Neu-
totemporal dementia. Neurology 2005;64:1384 –90. rosurg Psychiatry 1997;63:749 –53.
[50] Rosen HJ, Gorno-Tempini ML, Goldman WP, Perry RJ, Schuff N, [72] Tomlinson BE, Blessed G, Roth M. Observations on the brain of
Weiner M, et al. Patterns of brain atrophy in frontotemporal demen- demented old people. J Neurol Sci 1970;11:205– 42.
tia and semantic dementia. Neurology 2002;58:198 –208. [73] Hansen LA, Samuel W. Criteria for Alzheimer’s disease and the
[51] Kertesz A, Munoz DG. Primary progressive aphasia and Pick com- nosology of dementia with Lewy bodies. Neurology 1997;48:
plex. J Neurol Sci 2003;206:97–107. 126 –32.
[52] The Lund Manchester Groups. Clinical and neuropathological crir- [74] Hughes AJ, Daniel SE, Blankson S, Lees AJ. A clinicopathologic
teria for frontotemporal dementia. J Neurol Neurosurg Psychiatry study of 100 cases of Parkinson’s disease. Arch Neurol 1993;50:
1994;57:416 – 8. 140 – 8.
[53] Miller BL, Ikonte C, Ponton M, Levy M, Boone K, Darby A, et al. [75] Merdes AR, Hansen LA, Jeste DV, Galasko D, Hofstetter CR, Ho
A study of the Lund-Manchester research criteria for frontotemporal GJ, et al. Influence of Alzheimer pathology on clinical diagnostic
dementia: clinical and single-photon emission CT correlations. Neu- accuracy in dementia with Lewy bodies. Neurology 2003;60:1586 –
rology 1997;46:937– 42. 90.
[54] Pasquier F, Petit H. Frontotemporal dementia: its rediscovery. Eur [76] Srikanth S, Nagaraja AV, Ratnavalli E. Neuropsychiatric symptoms
Neurol 1997;38:1– 6. in dementia-frequency, relationship to dementia severity and com-
[55] Grosman M. Frontotemporal dementia: a review. J Int Neuropsychol parison in Alzheimer’s disease, vascular dementia and frontotem-
Soc 2002;8:566 – 83. poral dementia. J Neurol Sci 2005;236:43– 8.
[56] Gorno-Tempini ML, Dronkers NF, Rankin KP, Ogar JM, Phen- [77] Emre M, Aarsland D, Albanese A, Byrne EJ, Deuschl G, De Deyn
grasamy L, Rosen HJ, et al. Cognition and anatomy in three variants PP, et al. Rivastigmine for dementia associated with Parkinson’s
of primary progressive aphasia. Ann Neurol 2004;55:335– 46. disease. N Engl J Med. 2004;351:2509 –18.
[57] Shea C, MacKnight C, Rockwood K. Donepezil for treatment of [78] McKeith I, Del Ser T, Spano P, Emre M, Wesnes K, Anand R, et al.
dementia with Lewy bodies: a case series of nine patients. Int Efficacy of rivastigmine in dementia with Lewy bodies: a random-
Psychogeriatr 1998;10:229 –38. ised, double-blind, placebo-controlled international study. Lancet
[58] McKeith IG, Perry RH, Fairbairn AF, Jabeen S, Perry EK. Opera- 2000;16;356:2031– 6.
tional criteria for senile dementia of Lewy body type (SDLT). [79] O’Brien JT, Erkinjuntti T, Reisberg B, Roman G, Sawada T, Pantoni
Psychol Med 1992;22:911–22. L, et al. Vascular cognitive impairment. Lancet Neurol 2003;2:89 –
[59] McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen 98.
LA, et al. Consensus guidelines for the clinical and pathologic diag- [80] Roman G, Tatemichi TK, Erkinjuntti T, the NINDS-AIREN Work-
nosis of dementia with Lewy bodies (DLB): report of the consortium on group. Vascular dementia: diagnostic criteria for research studies—
DLB international workshop. Neurology 1996;47:1113–24. report of the NINDS-AIREN International workshop. Neurology
[60] McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman 1993;43:250 – 60.
H, et al. Diagnosis and management of dementia with Lewy bodies: [81] Chui HC, Victoroff JI, Margolin D, Jagust W, Shankle R, Katzman
third report of the DLB consortium. Neurology 2005;65:1863–72. R. Criteria for the diagnosis of ischemic vascular dementia proposed
[61] Lopez OL, Litvan I, Catt KE, Stowe R, Klunk W, Kaufer DI, et al. by the State of California Alzheimer’s Disease Diagnostic and
Accuracy of four diagnostic criteria for the diagnosis of neurode- Treatment Centers. Neurology 1992;42:473– 80.
generative dementias. Neurology 1999;53:1292–9. [82] Rockwood K, Davis H, MacKnight C, Vandorpe R, Gauthier S,
[62] Soret M, Koulibaly PM, Darcourt J, Buvat I. Partial volume effect Guzman A. The Consortium to Investigate Vascular Impairment of
correction in SPECT for striatal uptake measurements in patients Cognition: methods and first findings. Can J Neurol Sci 2003;30:
with neurodegenerative diseases. impact upon patient classification. 237– 43.
Eur J Nucl Med Mol Imaging 2006 [Epub ahead of print]. [83] Lopez OL, Kuller LH, Becker JT, Jagust WJ, DeKosky ST, Fitz-
[63] O’Brien JT, Colloby S, Fenwick J, Williams ED, Firbank M, Burn patrick A, et al. Classification of vascular dementia in the Cardiovas-
D, et al. Dopamine transporter loss visualized with FP-CIT SPECT cular Health Study Cognition Study. Neurology 2005;64:1539 – 47.
in the differential diagnosis of dementia with Lewy bodies. Arch [84] Verhey FR, Lodder J, Rozendaal N, Jolles J. Comparison of seven
Neurol 2004;61:919 –25. sets of criteria used for the diagnosis of vascular dementia. Neuro-
[64] Folstein MF, Folstein SE, McHugh PR.“Mini-mental state”: A prac- epidemiology 1996;15:166 –72.
tical method for grading the cognitive state of patients for the [85] Pohjasvaara T, Mantyla R, Ylikoski R, Kaste M, Erkinjuntti T.
clinician. J Psychiatr Res 1975;12:189 –98. Comparison of different clinical criteria (DSM-III, ADDTC, ICD-10,
[65] Korczyn AD. The complex nosological concept of vascular demen- NINDS-AIREN, DSM-IV) for the diagnosis of vascular dementia:
tia. J Neurol Sci 2002;203-204:3– 6. National Institute of Neurological Disorders and Stroke-Association
[66] Aarsland D, Tandberg E, Larsen JP, Cummings JL. Frequency of Internationale pour la Recherche et l’Enseignement en Neurosciences.
dementia in Parkinson’s disease. Arch Neurol 1996;53:538 – 42. Stroke 2000;31:2952–7.
[67] Aarsland D, Andersen K, Larsen JP, Lolk A, Nielsen H, Kragh- [86] Gold G, Bouras C, Canuto A, Bergallo MF, Herrmann FR, Hof PR,
Sorensen P. Risk of dementia in Parkinson’s disease: a community- et al. Clinicopathological validation study of four sets of clinical
based, prospective study. Neurology 2001;56:730 – 6. criteria for vascular dementia. Am J Psychiatry 2002;159:82–7.
[68] Emre M. Dementia associated with Parkinson’s disease. Lancet [87] Hulette C, Nochlin D, McKeel D, Morris JC, Mirra SS, Sumi SM,
Neurol 2003;2:229 –37. et al. Clinical-neuropathologic findings in multi-infarct dementia: a
[69] Boller F, Mizutani T, Roessmann U, Gambetti P. Parkinson disease, report of six autopsied cases. Neurology 1997;48:668 –72.
dementia, and Alzheimer’s disease: clinicopathological correlations. [88] Knopman DS, Rocca WA, Cha RH, Edland SD, Kokmen E. Inci-
Ann Neurol 1980;7:329 –35. dence of vascular dementia in Rochester, Minn, 1985–1989. Arch
[70] Aarsland D, Perry R, Brown A, Larsen JP, Ballard C. Neuropathol- Neurol 2002;59:1605–10.
ogy of dementia in Parkinson’s disease: a prospective, community- [89] Rockwood K, Black SE, Song X, Hogan DB, Gauthier S, MacKnight
based study. Ann Neurol 2005;58:773– 6. C, et al. Clinical and radiographic subtypes of vascular cognitive
K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440 439

impairment in a clinic-based cohort study. J Neurol Sci tivity and specificity for diagnosis. Am J Neuroradiol 2005;
2006;15:7–14. 26:1551– 62.
[90] Erkinjuntti T, Inzitari D, Pantoni L, Wallin A, Scheltens P, Rock- [110] Steinhoff BJ, Zerr I, Glatting M, Schulz-Schaeffer W, Poser S,
wood K, et al. Research criteria for subcortical vascular dementia in Kretzschmar HA. Diagnostic value of periodic complexes in
clinical trials. J Neural Transm Suppl 2000;59:23–30. Creutzfeldt-Jakob disease. Ann Neurol 2004;56:702– 8.
[91] Prins ND, van Dijk EJ, den Heijer T, Vermeer SE, Jolles J, Koud- [111] Dong Y, Hassan A, Zhang Z, Huber D, Dalageorgou C, Markus HS.
staal PJ, et al. Cerebral small-vessel disease and decline in infor- Yield of screening for CADASIL mutations in lacunar stroke and
mation processing speed, executive function and memory. Brain leukoaraiosis. Stroke 2003;34:203–5.
2005;128(Pt 9):2034 – 41. [112] Greenberg SM, Vonsattel JP, Stakes JW, Gruber M, Finklestein SP.
[92] Vermeer SE, Prins ND, den Heijer T, Hofman A, Koudstaal PJ, The clinical spectrum of cerebral amyloid angiopathy: presentations
Breteler MM. Silent brain infarcts and the risk of dementia and without lobar hemorrhage. Neurology 1993;43:2073–9.
cognitive decline. N Engl J Med 2003;348:1215–22. [113] Harkness KA, Coles A, Pohl U, Xuereb JH, Baron JC, Lennox GG.
[93] Hachinski V. Preventable senility: a call for action against the Rapidly reversible dementia in cerebral amyloid inflammatory vas-
vascular dementias. Lancet. 1992;340:645– 8. culopathy. Eur J Neurol 2004;11:59 – 62.
[94] Hebb AO, Cusimano MD. Idiopathic normal pressure hydrocepha- [114] McCusker J, Cole M, Dendukuri N, Belzile E, Primeau F. Delirium
lus: a systematic review of diagnosis and outcome. Neurosurgery in older medical inpatients and subsequent cognitive and functional
2001;49:1166 – 86. status: a prospective study. CMAJ 2001;165:575– 83.
[95] Relkin N, Marmarou A, Klinge P, Black PM. Diagnosing idiopathic [115] Rockwood K, Cosway S, Carver D, Jarrett P, Stadnyk K, Fisk J.
normal pressure hydrocephalus. Neurosurgery 2005;57(Suppl):S4 –16. The risk of dementia and death after delirium. Age Ageing
[96] Vanneste J, Augustijn P, Tan WF, Dirven C. Shunting normal 1999;28:551– 6.
pressure hydrocephalus: the predictive value of combined clinical [116] Tschampa HJ, Neumann M, Zerr I, Henkel K, Schröter A, Schulz-
and CT data. J Neurol Neurosurg Psychiatry 1993;56:251– 6. Schaeffer WJ, et al. Patients with Alzheimer’s disease and dementia
[97] Gallia GL, Rigamonti D, Williams MA. The diagnosis and treatment with Lewy bodies mistaken for Creutzfeldt-Jakob disease. J Neurol
of idiopathic normal pressure hydrocephalus. Nat Clin Pract Neurol Neurosurg Psychiatry 2001;71:33–9.
2006;2:375– 81. [117] Reinwald S, Westner IM, Niedermaier N. Rapidly progressive Alz-
[98] Poca MA, Mataró M, Matarín M, Arikan F, Junqué C, Sahuquillo J.
heimer’s disease mimicking Creutzfeldt-Jakob disease. J Neurol
Good outcome in patients with normal-pressure hydrocephalus and
2004;251:1020 –2.
factors indicating poor prognosis. J Neurosurg 2005;103:455– 63.
[118] Catani M, Piccirilli M, Geloso MC, Cherubini A, Finali G, Pellic-
[99] McArthur JC, Brew BJ, Nath A. Neurological complications of HIV
cioli G, et al. Rapidly progressive aphasic dementia with motor
infection. Lancet Neurol 2005;4:543–55.
neuron disease: a distinctive clinical entity. Dement Geriatr Cogn
[100] Timmermans M, Carr J. Neurosyphilis in the modern era. J Neurol
Disord 2004;17:21– 8.
Neurosurg Psychiatry 2004;75:1727–30.
[119] Ballard CG, Aarsland D, McKeith I, O’Brien J, Gray A, Cormack F,
[101] Rabinovici GD, Wang PN, Levin J, Cook L, Pravdin M, Davis J,
et al. Fluctuations in attention: PD dementia vs DLB with parkin-
et al. First symptom in sporadic Creutzfeldt-Jakob disease. Neurol-
sonism. Neurology 2002;59:1714 –20.
ogy 2006;66:286 –7.
[120] Caselli RJ, Boeve BF, Scheithauer BW, O’Duffy JD, Hunder GG.
[102] Masters CL, Harris JO, Gajdusek DC, Gibbs CJ Jr, Bernoulli C,
Nonvasculitic autoimmune inflammatory meningoencephalitis
Asher DM. Creutzfeldt–Jakob disease: patterns of worldwide occur-
(NAIM): a reversible form of encephalopathy. Neurology 1999;53:
rence and the significance of familial and sporadic clustering. Ann
1579 – 81.
Neurol 1979;5:177– 88.
[103] WHO Global Surveillance. Diagnosis and therapy of human trans- [121] Gómez-Puerta JA, Cervera R, Calvo LM, Gómez-Ansón B, Espi-
missible spongiform encephalopathies: report of a WHO consulta- nosa G, Claver G, et al. Dementia associated with the antiphospho-
tion. In: World Health Organization: emerging and other communi- lipid syndrome: clinical and radiological characteristics of 30 pa-
cable diseases, surveillance and control. Geneva, Switzerland: tients. Rheumatology (Oxford) 2005;44:95–9.
World Health Organization; 1998. [122] Tolnay M, Clavaguera F. Argyrophilic grain disease: a late-onset
[104] Geschwind MD, Martindale J, Miller D, DeArmond SJ, Uyehara- dementia with distinctive features among tauopathies. Neuropathol-
Lock J, Gaskin D, et al. Challenging the clinical utility of the 14-33 ogy 2004;24:269 – 83.
protein for the diagnosis of sporadic Creutzfeldt-Jakob disease. Arch [123] Cairns NJ, Grossman M, Arnold SE, Burn DJ, Jaros E, Perry RH,
Neurol 2003;60:813– 6. et al. Clinical and neuropathologic variation in neuronal intermedi-
[105] Tschampa HJ, Kallenberg K, Urbach H, Meissner B, Nicolay C, ate filament inclusion disease. Neurology 2004;63:1376 – 84.
Kretzschmar HA, et al. MRI in the diagnosis of sporadic [124] Blass DM, Hatanpaa KJ, Brandt J, Rao V, Steinberg M, Troncoso
Creutzfeldt-Jakob disease: a study on inter-observer agreement. JC, et al. Dementia in hippocampal sclerosis resembles frontotem-
Brain 2005;128:2026 –33. poral dementia more than Alzheimer disease. Neurology 2004;63:
[106] Brown P, Rodgers-Johnson P, Cathala F, Gibbs CJ Jr, Gajdusek DC. 492–7.
Creutzfeldt-Jakob disease of long duration: clinicopathological char- [125] Hatanpaa KJ, Blass DM, Pletnikova O, Crain BJ, Bigio EH, Hedreen
acteristics, transmissibility, and differential diagnosis. Ann Neurol JC, et al. Most cases of dementia with hippocampal sclerosis may
1984;16:295–304. represent frontotemporal dementia. Neurology 2004;63:538 – 42.
[107] Will RG, Zeidler M, Stewart GE, Macleod MA, Ironside JW, [126] Lippa CF, Dickson DW. Hippocampal sclerosis dementia: expand-
Cousens SN, et al. Diagnosis of new variant Creutzfeldt-Jakob ing the phenotypes of frontotemporal dementias? Neurology 2004;
disease. Ann Neurol 2000;47:575– 82. 63:414 –5.
[108] Boesenberg C, Schulz-Schaeffer WJ, Meissner B, Kallenberg K, [127] Louis ED, Lynch T, Kaufmann P, Fahn S, Odel J. Diagnostic
Bartl M, Heinemann U, et al. Clinical course in young patients with guidelines in central nervous system whipple’s disease. Ann Neurol
sporadic Creutzfeldt-Jakob disease. Ann Neurol 2005;58:533– 43. 1996;40:561– 8.
[109] Young GS, Geschwind MD, Fischbein NJ, Martindale JL, Henry [128] Houlden H, Baker M, McGowan E, Lewis P, Hutton M, Crook R, et
RG, Liu S, et al. Diffusion-weighted and fluid-attenuated inver- al. Variant Alzheimer’s disease with spastic paraparesis: clinical
sion recovery imaging in Creutzfeldt-Jakob disease: high sensi- characterization. Neurology 2000;54:1103–9.
440 K. Rockwood et al. / Alzheimer’s & Dementia 3 (2007) 428 – 440

[129] Graves TD, Hanna MG. Neurological channelopathies. Postgrad [132] Gold G, Kövari E, Corte G, Herrmann FR, Canuto A, Bussière T, et al.
Med J 2005;81:20 –32. Clinical validity of A beta-protein deposition staging in brain aging and
[130] Lyon G, Fattal-Valevski A, Kolodny EH. Leukodystrophies: clinical Alzheimer disease. J Neuropathol Exp Neurol 2001;60:946 –52.
and genetic aspects. Top Magn Reson Imaging. 2006;17:219 – 42. [133] Song X, Mitnitski A, Rockwood K. Index variables for studying
[131] Kumar N. Superficial siderosis: associations and therapeutic impli- outcomes in vascular cognitive impairment. Neuroepidemiology
cations. Arch Neurol 2007;64:491– 6. 2005;25:196 –204.
Alzheimer’s & Dementia 3 (2007) 441– 443

Perspectives

Commentary on “The Third Canadian Consensus Conference on the


Diagnosis and Treatment of Dementia:”
Clinical guidelines are not enough—System-wide, population-based
programs are needed to improve the care of patients with Alzheimer’s
disease and related dementias
Howard Fillit*
Alzheimer’s Drug Discovery Foundation and the Institute for the Study of Aging, New York, New York

This volume truly represents a timely, expert, well- clinical practice and patient outcomes.” Chertkow states,
organized synopsis of recent developments in the treatment “In conclusion, the CCCDTD3 was a time-consuming but
and care of patients with Alzheimer’s disease, particularly rewarding enterprise for all those who participated. Only
from the Canadian perspective, but based on worldwide time will tell, though, if we will improve both the care
data. The purpose of this effort is to create a consensus of provided by Canadian physicians and the outcomes of pa-
recommendations on relevant issues based on new knowl- tients with a dementia.”
edge. The consensus process is rigorous and clearly defined, We are all happy that the committee had a “rewarding”
and has a strong base of evidence. The papers and recom- experience. But demonstrating the impact of these recom-
mendations presented here cover many critical and relevant mendations on clinical care and outcomes is the most im-
areas of recent advances in the field. Relevant clinical topics portant, difficult, and complex task. I have not practiced
range from early detection to treatment and management of medicine in Canada, but I have practiced geriatric medicine
care. Ultimately, 215 recommendations were considered, in the United States for almost 30 years. I have also had the
and 149 recommendations were approved with strong con- opportunity to practice “population-based” medicine in the
sensus by the Canadian academic community involved in United States through my tenure as a corporate medical
clinical research and care. I will not attempt to comment director for a national Medicare managed-care organization
specifically on these many recommendations that were so that served over 125,000 elderly individuals throughout the
carefully and thoughtfully evaluated by my esteemed col- country. From these experiences at bedside and in the ad-
leagues, though in general, I certainly agree with them. ministrative health system office, I know that while the goal
Rather, my perspective will address the real value of this to improve the quality of care for dementia is certainly
extensive, expensive, and time-consuming work. It is criti- laudable and critical, there are many, many challenges along
cally important to note that, quoting Dr. Chertkow in his the way.
paper, “Translating, disseminating, and implementing our In the trenches, physicians throughout the world are
recommendations received particular emphasis.” An imple- under stress. In the United States, the average primary-care
mentation committee was created to address these “knowl- office visit is quite brief, generally less than 10 minutes, and
edge translation” issues, with a specific work plan that little time is available to provide extensive counseling in our
included publications, tool kits, and practice aids for distri- national health system for the elderly (called Medicare,
bution to Canadian family practitioners, course material, which provides extensive and comprehensive health bene-
and “assessment of the impact of . . . recommendations on fits, including prescription drug coverage, to over 95% of
the population). One-hour visits for counseling are not well-
paid. Primary-care physicians, who see the majority of de-
*Corresponding author. mented persons in the United States (and in most other parts
E-mail address: hfillit@alzdiscovery.org of the world) [1], are ill-equipped to provide cognitive
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.09.003
442 H. Fillit / Alzheimer’s & Dementia 3 (2007) 441– 443

assessments, and have little experience with dementia pa- the Centers for Medicare and Medicaid Services in the
tients, dementia care management strategies, and the use of United States have adopted many principles of managed
antidementia treatments. From a “back of the envelope” care into their fee-for-service program because the lessons
perspective, in a typical internal-medicine or family practice learned from managed care have been valuable as an ex-
(as opposed to an academic neurology clinic), about 50% of perimental laboratory to improve our health system with
the patients are elderly. If 5% of these patients over 65 have regard to specific diseases in a population-based manner.
dementia, that amounts to 2.5% of the total population that I emphasize this because the individual physician alone
the physician sees. If the physician has a practice of 1,500 cannot be the target for improving the quality of care for
active patients, that amounts to 750 elderly, or about 20 dementia patients. Individual physicians, or even groups of
dementia patients in the entire practice. If each dementia physicians, do not have the resources, incentives, position in
patient was seen four times a year, the physician would have the health system, or capacity to change the system of care.
a total of 80 dementia visits a year, or a little more than one The distribution of guidelines, recommendations, practice
patient with dementia per week: not really quite enough to aids, and “kits” to physicians has relatively little impact if
get the doctor’s attention, or focus on issues such as care- not linked to an interventional program that can change
management strategies and quality-of-care measures. physician or patient behavior by adding value and resources
The same arithmetic holds true for health systems and to the health system itself [20 –22]. These programs often
society, but here we must address not only clinical and offer additional system-wide services to physicians that
quality issues, but also cost issues. To get the attention of can enable and ensure quality of care and adherence to
the national systems which ultimately pay for care and have guideline-recommended care. To implement effectively the
a societal perspective in the distribution of finite resources 149 recommendations for dementia presented in this vol-
and effort with multiple diseases competing for attention, ume in clinical practice is a great challenge, particularly
and to get these systems to invest in improving the baseline because many of them do not have convincing “grade A”
of care for a particular condition, we must be able to dem- recommendations that would warrant the financial and re-
onstrate to these organizations that the dementias are prev- source investment in the programs, systems, and services
alent and costly, and that something proven effective can be necessary to ensure their adoption and adherence.
done to improve care. Getting the attention of health sys- From the recommendations of the Canadians, as well as
tems throughout the world has been difficult with regard to of others, we probably know what we need to do to improve
dementia for a variety of reasons, including under-reporting, care for Alzheimer’s disease [8,9,23–25], and we have made
undercoding, underdiagnosis [2]. Also, payers often lack an some progress in changing the system. We have developed
understanding of the impact of dementias on individuals and telephonic approaches to cognitive assessment [26,27] and
society. They do not see the dementias as traditional med- Alzheimer’s disease management programs [11–16]. In the
ical illnesses, and the direct relationship between “losing United States and elsewhere, nonprofit organizations such
your keys” and costly hospitalizations and other direct med- as the Alzheimer’s Association (http://www.alz.org/we_can_
ical costs is not always self-evident [3,4]. Thus, at least in help_24_7_helpline.asp) and the Alzheimer’s Foundation of
the United States, Medicare certainly “knows” about the America (AFA) provide system-wide telephonic services
dementias, and as much as we in the field think it is so directly to patients to promote early detection, support groups,
important, compared with congestive heart failure and dia- and telephonic care management. The AFA also conducts a
betes, Alzheimer’s disease is not really on the radar of community-based memory screening program. Some dem-
Medicare. onstration programs and research reports demonstrated that
Studies show that changing the way that care is provided Alzheimer’s disease care management can have an impact
for any disease by the distribution or publication of educa- on outcomes of importance and relevance (http://www.
tional materials and consensus opinions is not enough [5]. nationalmemoryscreening.org/). The United States Veteran’s
Guidelines for the elderly present many challenges, espe- Administration health system, one of the largest health systems
cially to busy physicians [6]. System-wide, programmatic in the world, has also adopted certain aspects of assessment
interventions that change the way the system of care oper- and care management for Alzheimer’s disease (http://www1.
ates with regard to the particular disease state are needed va.gov/geriatricsshg/docs/VADementiaActUpdate1_05.doc).
[7–9]. One approach has involved screening, collaborative Why is all this important in terms of this volume of
care, and care-management, disease-management interven- papers? To really change clinical practice and promote
tions [10 –17]. Another involves systematic approaches to quality of care requires more than just the publication of
quality improvement [18], such as Medicare’s QIC pro- well-written academic guidelines and the consensus opin-
grams (http://www.ahqa.org/pub/media/159_766_2687.cfm). ions of experts. Apart from academic specialists, the busy
Few of these, to my knowledge, include programs related to practicing physicians in the community, their patients, and
dementia. A third recent approach is pay-for-performance the medical managers of health systems will not have the
[19], which is being explored and implemented in the time, focus, interest, incentives, or mechanisms to read this
United States, the United Kingdom, and elsewhere. Thus, entire volume, let alone consider, remember, and utilize the
H. Fillit / Alzheimer’s & Dementia 3 (2007) 441– 443 443

149 recommendations during their business day and imple- [9] Fillit H, Knopman D, Cummings J, Appel F. Opportunities for im-
ment them. Health systems, including national health sys- proving managed care for individuals with dementia: part 1—the
issues. Am J Manag Care 1999;5:309 –15.
tems, managed-care organizations, and other payers, will [10] Boustani M, Callahan CM, Unverzagt FW, et al. Implementing a
not adopt these guidelines, promote system-wide changes, screening and diagnosis program for dementia in primary care. J Gen
and implement programs without clear demonstrations that Intern Med 2005;20:572–7.
there will be societal or population benefits. This is good [11] Charlson M, Charlson RE, Briggs W, Hollenberg J. Can disease
thing, because before we commit large sums of money and management target patients most likely to generate high costs? The
impact of comorbidity. J Gen Intern Med 2007;22:464 –9.
resources to improve the health of populations with Alzhei- [12] Dancha L, Martini EM. Using data in disease management. Oppor-
mer’s disease and related dementias, we need to be sure tunities and challenges. Med Group Manage J 1998;45:20 – 8.
about what we are doing, and how we will do it. [13] Iraqi A. Disease management intervention on quality and outcomes of
Therefore I applaud the extensive efforts of my esteemed dementia care. Ann Intern Med 2007;147:69.
Canadian neighbors to begin the process of improving the [14] Cherry DL, Vickrey BG, Schwankovsky L, Heck E, Plauchm M, Yep
R. Interventions to improve quality of care: the Kaiser Permanente-
population-wide health of patients with dementias in North Alzheimer’s Association Dementia Care Project. Am J Manag Care
America. The Canadians have a unique and effective 2004;10:553– 60.
national health system that should be able to implement [15] Clark PA, Bass DM, Looman WJ, McCarthy CA, Eckert S. Outcomes
system-wide changes that, together with greater knowledge for patients with dementia from the Cleveland Alzheimer’s Managed
on the part of physicians, can make a difference, if there is Care Demonstration. Aging Ment Health 2004;8:40 –51.
[16] Mittelman MS, Ferris SH, Shulman E, Steinberg G, Levin B. A
the will and the money. Their efforts could serve as a model family intervention to delay nursing home placement of patients with
for health systems throughout the world, both public and Alzheimer disease. A randomized controlled trial. JAMA 1996;276:
private, to make changes that would have real effects on the 1725–31.
quality, efficiency, effectiveness, and cost of care for Alz- [17] Callahan CM, Boustani MA, Unverzagt FW, et al. Effectiveness of
heimer’s disease and related dementias. They have set the collaborative care for older adults with Alzheimer disease in primary
care: a randomized controlled trial. JAMA 2006;295:2148 –57.
challenge for themselves, but to paraphrase Dr. Chertkow [18] Diamond LH. Local implementation of clinical practice guidelines
again, only time will tell if the appropriate commitment of and continuous quality improvement: challenges and opportunities.
resources and innovations will systematically improve both Semin Dial 2000;13:364 – 8.
the care provided by Canadian physicians and the outcomes [19] Boyd CM, Darer J, Boult C, Fried LP, Boult L, Wu AW. Clinical
of patients with a dementia in Canada. practice guidelines and quality of care for older patients with multiple
comorbid diseases: implications for pay for performance. JAMA
2005;294:716 –24.
[20] Gandhi TK, Sequist TD, Poon EG, Karson AS, Murff H, Fairchild
References D,et al. Primary care clinician attitudes towards electronic clinical
reminders and clinical practice guidelines. AMIA Annu Symp Proc
[1] Wilkinson D, Stave C, Keohane D, Vincenzino O. The role of general 2003;848.
practitioners in the diagnosis and treatment of Alzheimer’s disease: a [21] Gerhardt WE, Schoettker PJ, Donovan EF, Kotagal UR, Muething
multinational survey. J Int Med Res 2004;32:149 –59. SE. Putting evidence-based clinical practice guidelines into practice:
[2] Fillit H, Geldmacher DS, Welter RT, Maslow K, Fraser M. Optimiz- an academic pediatric center’s experience. Jt Comm J Qual Patient
ing coding and reimbursement to improve management of Alzhei- Saf 2007;33:226 –35.
mer’s disease and related dementias. J Am Geriatr Soc 2002;50: [22] Green CJ, Fortin P, Maclure M, Macgregor A, Robinson S. Informa-
1871– 8. tion system support as a critical success factor for chronic disease
[3] Wilkinson D. Is there a double standard when it comes to dementia management: necessary but not sufficient. Int J Med Inform 2006;75:
care? Int J Clin Pract 2005;146(Suppl).3–7. 818 –28.
[4] Fillit H, Hill JW, Futterman R. Health care utilization and costs of [23] Cummings JL, Frank JC, Cherry D, et al. Guidelines for managing
Alzheimer’s disease: the role of co-morbid conditions, disease stage, Alzheimer’s disease: part I. Assessment. Am Fam Physician 2002;
and pharmacotherapy. Fam Med 2002;34:528 –35. 65:2263–72.
[5] Cochrane LJ, Olson CA, Murray S, Dupuis M, Tooman T, Hayes S. [24] Cummings JL, Frank JC, Cherry D, et al. Guidelines for managing
Gaps between knowing and doing: understanding and assessing the Alzheimer’s disease: part II. Treatment. Am Fam Physician 2002;65:
barriers to optimal health care. J Contin Educ Health Prof 2007;27: 2525–34.
94 –102. [25] Doody RS, Stevens JC, Beck C, et al. Practice parameter: manage-
[6] Tinetti ME, Bogardus ST Jr, Agostini JV. Potential pitfalls of disease- ment of dementia (an evidence-based review). Report of the Quality
specific guidelines for patients with multiple conditions. N Engl Standards Subcommittee of the American Academy of Neurology.
J Med 2004;351:2870 – 4. Neurology 2001;56:1154 – 66.
[7] Epping-Jordan JE, Pruitt SD, Bengoa R, Wagner EH. Improving the [26] Hill J, McVay JM, Walter-Ginzburg A, et al. Validation of a brief
quality of health care for chronic conditions. Qual Saf Health Care screen for cognitive impairment (BSCI) administered by telephone
2004;13:299 –305. for use in the Medicare population. Dis Manag 2005;8:223–34.
[8] Fillit H, Knopman D, Cummings J, Appel F. Opportunities for im- [27] de Jager CA, Budge MM, Clarke R. Utility of TICS-M for the
proving managed care for individuals with dementia: part 2—a assessment of cognitive function in older adults. Int J Geriatr Psy-
framework for care. Am J Manag Care 1999;5:317–24. chiatry 2003;18:318 –24.
Alzheimer’s & Dementia 3 (2007) 444 – 445

Commentary on “The Third Canadian Consensus Conference on the


Diagnosis and Treatment of Dementia”
Bruno Vellasa,*, Emma Reynisha, Philippe Robertb
a
Alzheimer’s Disease Research and Clinical Center, INSERM U 558, Gerontopole, Toulouse, France
b
Memory and Alzheimer’s Disease Center, CM2R CHU, University of Nice, Nice, France

Abstract A comprehensive and useful initiative has been performed by our Canadian colleagues. The subjects
presented are under active discussion in many countries, with the goal of direct applications in daily
clinical practice. The review provided by this special issue of the Journal will therefore be very
useful for all. In this commentary we will not discuss all papers, but will underline only some
important points concerning mild cognitive impairment and the diagnosis and management of
Alzheimer’s disease.
© 2007 The Alzheimer’s Association. All rights reserved.
Keywords: ●●●

1. Mild cognitive impairment loss. To increase our knowledge, research on cognitive


intervention, physical training, or psychological interven-
In the future, the place for mild cognitive impairment tions is needed, and must be performed in different coun-
(MCI) will be found between the predementia stage of the tries to take into account potential cultural differences.
disease [1] and the population targeted in the primary pre-
vention of Alzheimer’s disease (AD). The biological and
pathophysiological process of Alzheimer’s disease starts
decades before clinical symptoms appear, and large preven- 2. Diagnosis of Alzheimer’ disease
tive trials are presently planned or underway for patients in The diagnosis of AD is generally easy to make clinically
the pre-MCI stage of cognitive decline. However, MCI in mild to moderate stages of the disease, with its typical
patients are commonly seen in our memory clinics and need forms of progressive memory loss and decline in the activ-
specific assessment, follow-up, and intervention. Certainly a ities of daily living. The more extensive investigations in-
comprehensive multidomain intervention for a healthy life, volving complex neuropsychological testing, neuroimaging,
including physical and cognitive exercise, nutrition, social and cerebrospinal fluid biomarkers should perhaps be used
activities, control of vascular and metabolic risk factors, and initially for more atypical cases. However, surrogate mark-
control of hearing and visual impairments, seems to make ers, including biomarkers, will be very useful for therapeu-
sense, and will shortly be tested in both the primary and tic research, as mentioned recently [2].
secondary prevention domains. In this field, the information Recommendations concerning the revision of criteria for
indicating that physical training may be protective against the diagnosis of AD perhaps need further discussion. Rock-
cognitive decline appears robust enough for clinical recom- wood et al. in particular indicated the importance of depres-
mendations. There is evidence that physicians and therapists sion in mild dementia, where it is common. In contrast to an
should promote activity at an intensity level that is adapted earlier era, when it was felt important to rule out depression
to a person’s overall physical capacities, as part of a healthy before making a diagnosis of AD, there is now recognition
lifestyle for older individuals with and without memory of symptom overlap, perhaps reflecting a shared pathophys-
iology. In addition, it is important to underline that apathy is
*Corresponding author. Tel.: 33 (5) 61 77 76 49; Fax: 33 (5) 61 49 71 09. even more frequent than depressive symptoms in MCI and
E-mail address: brunovellas@aol.com mild AD [3,4], and these symptoms are frequently falsely
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.09.001
B. Vellas et al. / Alzheimer’s & Dementia 3 (2007) 444 – 445 445

regarded as depressive symptoms, potentially leading to passes a range of disabilities in each of three domains, i.e.,
psychotropic misuse. cognition, behavior, and function. In particular, BPSD treat-
ment is a key point in the management of severe AD.
Herrmann and Gauthier, in their recommendation 9, indi-
3. The management of dementia
cate that “approaches that may be useful for severe AD
The management of dementia, from very early to severe include behavioural management for depression, and
stages of AD, is still a difficult area. Recommendations and caregivers/staff education programs for a variety of behav-
possible complications are well-documented, but further iors.” Taking into account that the most problematic BPSDs
information is required about continuing care: how fre- are agitation, aggressiveness, oppositional behavior, and
quently should patients be followed, what assessment of psychotic disturbances, it is also important to dedicate spe-
associated clinical disorders should be made and how cific guidelines for their management [5].
should they be managed, when should treatment be contin- Finally, here as well as in the guidelines published in the
ued or withdrawn, and what is the most appropriate man- last decade, it is indicated that for BPSDs, nonpharmaco-
agement of endstage disease? These issues all require some logical treatment should be initiated first. Protocols involv-
thought. ing such interventions are more complex methodologically
The Plasa Study (see www.clinical trial.gov), a 2-year than those for pharmacological trials, but we feel that it is
randomized, clinical study on a specific care plan versus now time to go beyond good intentions, and to develop
usual care, incorporates a complete neuropsychological and more systematic research in this field. The consensus
geriatric assessment every 6 months in a specialized mem- reached by our Canadians colleagues is a very good exam-
ory clinic among 60 hospitals in France. This periodicity ple for all of us, and represents an important achievement in
must not be assumed to be fixed, but is adaptable in frail, the field of AD.
elderly patients, in those with the aggressive form of the
disease, and in those with a milder evolution of AD. Weight
loss and mobility disorders occur often in Alzheimer’s pa-
References
tients, and must be taken into account in the care plan, as
must behavioral and psychological symptoms of dementia [1] Dubois B, Feldman HH, Jacova C, et al. Research criteria for the
(BPSDs). Instauration and cessation of treatment are still diagnosis of Alzheimer’s disease: revising the NINCDS-ADRDA cri-
difficult questions to resolve in each individual situation. teria. Lancet Neurol 2007;6:734 – 46.
There is an obvious need to focus major research efforts on [2] Lovestone S. Biomarkers in Alzheimer’s disease. J Nutr Health Aging
2006;10:118 –22.
the biological basis, diagnosis, and treatment of early AD. It
[3] Robert PH, Berr C, Volteau M, et al. Apathy in patients with mild
is also our duty to take care of patients who are or shortly cognitive impairment and the risk of developing dementia of Alzhei-
will be at the most severe stage of the disease, and who will mer’s disease: a one-year follow-up study. Clin Neurol Neurosurg
probably not be able to benefit from new treatments. Even 2006;108:733– 6.
if there are no current, widely accepted consensus criteria [4] Robert PH, Berr C, Volteau M, et al. Neuropsychological performance
in mild cognitive impairment with and without apathy. Dement Geriatr
for severe AD, it is important to underline, in accordance
Cogn Disord 2006;21:192–7.
with the European Dementia Consensus, that the dementia [5] Benoit M, Robert PH, Staccini P, et al. One-year longitudinal evalu-
syndrome is inadequately defined by criteria which only ation of neuropsychiatric symptoms in Alzheimer’s disease. The
include the domains of cognition. Severe dementia encom- REAL.FR Study. J Nutr Health Aging 2005;9:95–9.
Alzheimer’s & Dementia 3 (2007) 446 – 448

Commentary on “The Third Canadian Consensus Conference on the


Diagnosis and Treatment of Dementia”—An appraisal
Paul S. Aisen*
●●●

Since the second series of consensus documents from mentation in mild-to-moderate AD (conducted by
Canadian Alzheimer’s disease (AD) experts was published the Alzheimer’s Disease Cooperative Study), with
in 1999, the field has undergone major changes in the areas overall negative results [1].
of genetics, risk and prevention, prodromal stages, diagno- 4. The discussion of various diagnostic labels for con-
sis, and management. The methodology utilized by this ditions between the extremes of normal cognition
Canadian group is well-constructed, and has yielded a new and dementia (Chertkow et al.) is very clear and
collection of papers that represents a major resource to useful. Though the consensus group did not favor
students, general practitioners, specialists, and research sci- Cognitive Impairment, No Dementia (CIND), or
entists. It was a large effort, but the payoff is substantial. Mild Cognitive Impairment (MCI) as optimal terms,
Most important, though, will be the efforts to assure optimal outside of Canada, and particularly within the realm
translation of the recommendations into clinical practice. of therapeutic trials, MCI prevails. But controversy
I offer comments on a few specific areas within this very continues, ranging from the issue of whether amnes-
broad project. tic forms of MCI should simply be referred to as
very mild AD [2], to the optimal memory test to
1. There continue to be many areas of confusion sur-
define the category. Most likely, useful criteria in
rounding the interpretation of observational studies
primary-care practice will differ from those in ther-
of modifiable risk factors of dementia (Patterson et
apeutic trials that must serve the purpose of identi-
al.), such as blood pressure, cholesterol, smoking,
fying a cohort with a predictable rate of progression.
and depression. This probably reflects the very dif-
There is reason for some optimism that this area will
ficult issue of bias in nonrandomized studies; it is a
be clarified by the application of biomarker studies.
major problem with no ready solution.
Thus, an individual with objectively confirmed epi-
2. With regard to the genetics of susceptibility to AD
sodic memory impairment that by history is progres-
(Hsiung and Sadovick), the apolipoprotein E geno-
sive, coupled with a marker of amyloid accumula-
type is unquestionably important, but it remains un-
tion such as low cerebrospinal fluid abeta [3] or an
certain whether any other genetic risk factor will
increased signal on amyloid positron emission to-
have much impact.
mography imaging [4], is likely to have an early
3. The relationship between homocysteine, B vitamins,
stage of AD (whether it is referred to as amnestic
and cognitive impairment and dementia (Garcia) is
MCI or very early AD).
given a balanced review. Whereas homocysteine el-
5. Few randomized prevention trials have aimed to reduce
evation is linked to both cerebrovascular disease
the risk of AD (Patterson et al.). This serves as a reflection
and AD, there is insufficient evidence to justify
of the cost and time necessary to conduct such trials, as
testing for or treating homocysteine in the older
well as uncertainty regarding the interventions that should
population with or without cognitive dysfunction.
be tested. The results of the interrupted Alzheimer’s Dis-
We recently presented the results of a randomized,
ease Anti-inflammatory Prevention Trial suggest that non-
controlled trial of high-dose B vitamin supple-
steroidal, anti-inflammatory drugs may, if anything, in-
crease the risk of cognitive decline and dementia [5],
*Corresponding author. and the Women’s Health Initiative indicated an in-
E-mail address: psa@georgetown.edu creased risk in women assigned to take hormones
1552-5260/07/$ – see front matter © 2007 The Alzheimer’s Association. All rights reserved.
doi:10.1016/j.jalz.2007.09.002
P.S. Aisen / Alzheimer’s & Dementia 3 (2007) 446 – 448 447

[6]. Results of the Ginkgo Evaluation of Memory ipation. To preclude research in consideration of an
Trial [7], assessing the preventive effect of ginkgo incomplete capacity to consent may slow or halt
biloba, are expected soon. therapeutic advances.
6. While important controversies continue to surround 10. The consensus recommendations on the diagnosis of
reimbursement issues, a consensus has developed dementia (Rockwell et al., and Robillard) recognize
regarding optimal pharmacotherapy for the primary the varied presentations in terms of cognitive do-
cognitive symptoms of AD. Hogan et al. provide a mains as well as time course, the limited value of
particularly thorough update on treatment, covering some ancillary procedures such as electroencepha-
primary and secondary symptoms, and important is- lograms and routine cerebrospinal fluid studies, and
sues such as driving safety and caregiver support. Cho- the frequent contributions of multiple etiologies. Neu-
linesterase inhibitors are modestly effective, and are ropsychological assessment (Jacova et al.) should be
generally appropriate therapy for mild to moderate AD. used selectively to clarify cognitive impairment in MCI
As reviewed by Herman and Gauthier, donepezil and and dementia. Although a huge number of such tests
presumably other cholinesterase inhibitors are also use- were described and demonstrated to be useful, a few
ful in the treatment of severe AD. Memantine, alone or straightforward tests that are brief and require little
in combination with a cholinesterase inhibitor, is training to administer and interpret may be sufficient
modestly effective for moderate to severe AD. in most clinical settings: the delayed paragraph re-
While Hogan et al. conclude that vitamin E is not call test, the MMSE, and clock-drawing. In some
recommended in the treatment of AD, this is open to cases, referral for comprehensive neuropsychologi-
question. The only randomized, controlled trial of cal testing may be appropriate. Although dementia
vitamin E in AD showed benefits [8], and this must and AD will continue to be clinical diagnoses rely-
be weighed against evidence from meta-analyses ing primarily on historical information provided by
and randomized trials in other populations that sug- knowledgeable informants, in coming years, there
gest the possibility of risk of congestive heart failure may be increased attention to the role of amyloid-
[9] or death [10]. In the case of vitamin E, recom- related biomarkers, from biochemical assessments in
mendations should be individualized (considering, cerebrospinal fluid to amyloid imaging, in the diag-
for example, cardiac risk profiles). Evidence regard- nosis of AD.
ing the efficacy of nonpharmacological approaches,
such as cognitive training, is still insufficient to This methodically researched and carefully written
support recommendations. series of review papers represents a highly useful sum-
7. The review of the literature by Bocti et al. suggests mary of the state of the art in AD diagnosis and treat-
that, in addition to attention to vascular risk factors, ment. All papers in this series offer valuable, practical
patients with dementia and cerebrovascular disease guidance to clinicians. Huge progress has been made
benefit to some degree from galantamine and per- since the first series from this group, and the field now
haps other cholinesterase inhibitors. Evidence for appears to be on the verge of additional major therapeutic
the benefits of Memantine is weaker. advances. The authors of this series have done a substan-
8. Fisk et al. review the difficult issues surrounding tial service to primary care and to specialty clinicians in
discussions of the diagnosis of dementia with pa- providing guidance today, and laying the groundwork for
tients at various stages of impairment, and with their advances to come.
families. They reasonably advocate progressive
disclosure during the process of evaluation, and an
References
individualized approach that considers the needs,
wishes, and expectations of patients, and they [1] Aisen PS, Jin S, Thomas RG, Sano M, Diaz-Arrastia R, Thal L.
emphasize the need for adequate time and, of ADCS Homocysteine Trial. Alzheimers Dementia 2007;3(Suppl.):
course, empathy and respect. S199.
9. The review of ethical issues surrounding consent-to- [2] Morris JC. Mild cognitive impairment is early-stage Alzheimer dis-
treatment and research participation (Fisk et al.) pro- ease: time to revise diagnostic criteria. Arch Neurol 2006;63:15– 6.
[3] Fagan AM, Roe CM, Xiong C, Mintun MA, Morris JC, Holtzman
vides an excellent summary of the complexity of DM. Cerebrospinal fluid tau/beta-amyloid(42) ratio as a prediction of
capacity determination, the role of proxy decision- cognitive decline in nondemented older adults. Arch Neurol 2007;64:
making, and caregiver/family involvement in this 343–9.
difficult process. Principles of autonomy and benef- [4] Forsberg A, Engler H, Almkvist O, et al. PET imaging of amyloid
icence must guide the consent process in this most deposition in patients with mild cognitive impairment. Neurobiol
Aging 2007.
vulnerable of populations. It is also essential to con- [5] Group AR, Lyketsos CG, Breitner JC, et al. Naproxen and celecoxib
sider that therapy for dementia, including severe do not prevent AD in early results from a randomized controlled trial.
stages, cannot be optimized without research partic- Neurology 2007;68:1800 – 8.
448 P.S. Aisen / Alzheimer’s & Dementia 3 (2007) 446 – 448

[6] Shumaker SA, Legault C, Thal L, et al. Estrogen plus progestin and [8] Sano M, Ernesto C, Thomas RG, et al. A controlled trial of selegiline,
the incidence of dementia and mild cognitive impairment in alpha-tocopherol, or both as treatment for Alzheimer’s disease.
postmenopausal women: the Women’s Health Initiative Mem- N Engl J Med 1997;336:1216 –22.
ory Study: a randomized controlled trial. JAMA 2003;289: [9] Lonn E, Bosch J, Yusuf S, et al. Effects of long-term vitamin E
2651– 62. supplementation on cardiovascular events and cancer: a randomized
[7] DeKosky ST, Fitzpatrick A, Ives DG, et al. The Ginkgo Evaluation of controlled trial. JAMA 2005;293:1338 – 47.
Memory (GEM) Study: design and baseline data of a randomized trial [10] Miller ER III, Pastor-Barriuso R, Dalal D, Riemersma RA, Appel LJ,
of ginkgo biloba extract in prevention of dementia. Contemp Clin Guallar E. Meta-analysis. high-dosage vitamin E supplementation
Trials 2006;27:238 –53. may increase all-cause mortality. Ann Intern Med. 2004.
449

Volume 3 / Issue 4 / October 2007

Spring Research Roundtable equipped to meet the code of federal regulations


Spurs Wide-Ranging Discussion (CFR) requirements for data acquisition.
● In general, academic researchers tend to be some-
Among Industry and Academic what naïve about the drug development process. Ac-
Thought Leaders ademics need education about Food and Drug Ad-
ministration (FDA) policies and procedures, but
The May 30 –31 meeting of the Alzheimer’s Association industry should not be the sole source of this
Research Roundtable convened more than 90 members education.
and invited guests in Washington, D.C., to explore the
Academic/Industry Interface for Alzheimer’s Drug Dis- Despite the challenges complicating the current rela-
covery. Historically, academic laboratories with deep ex- tionship, industry scientists emphasized their desire to
pertise in basic science led the field during the first wave work successfully with academic colleagues. Areas pro-
of accelerating discovery in the 1970s and 1980s. Those posed as niches academia might most successfully fill to
efforts focused on understanding the basic biology of the complement rather than compete with industry efforts in-
brain and what goes wrong in Alzheimer’s disease. In- clude the following:
sights gained identified a growing number of potential
points of intervention to treat symptoms or modify under- ● Develop better animal models. Current models are
lying disease processes. Work leading to development of good predictors of drug safety, with more than 70
tacrine (Cognex), approved in 1993 as the first drug to percent of adverse events picked up in animal stud-
treat cognitive symptoms, took place chiefly in academic ies. But available models fall woefully short in doc-
labs.
umenting therapeutic impact and behavioral effects.
Attracted by the steadily increasing number of potential
● Bring the needs and perspectives of patients and
therapeutic targets, pharmaceutical companies during the
caregivers to bear on clinical trial design, a function
1990s began to develop major Alzheimer drug discovery
academic investigators who work as both clinicians
initiatives. Now, there is a rapidly expanding industry-
and researchers are ideally positioned to fulfill.
based clinical research enterprise, with new compounds
entering human clinical testing at an unprecedented rate. ● Continue to identify novel targets and elucidate
The Research Roundtable Academic/Industry Interface basic disease processes. Regardless of the success or
meeting explored how these two arenas can forge better failure of current studies testing the amyloid hypoth-
working relationships in the current drug discovery cli- esis, achieving a mature clinical environment will
mate to speed movement of potentially valuable discover- require additional therapeutic strategies. Possible ap-
ies from basic academic science laboratories to further proaches include cytoskeletal stabilizers, novel anti-
development and clinical testing under the aegis of the inflammatories, metabolic enhancers, and neuropro-
pharmaceutical companies. Although there are some aca- tectants. In an academic environment, there is really
demic labs that excel in collecting and packaging basic no “bad target.” Even if a molecule turns out to lack
science data in ways that dovetail well with industry pro- potential as a drug, new data on its behavior may
grams, too often the two arenas operate as parallel uni- produce publishable insights into key disease
verses filled with mismatched expectations and missed processes.
opportunities.
Key issues raised by industry representatives included Industry scientists cited the following as elements of an
the following: ideal basic science “data package” with the potential to
pique the interest of a pharmaceutical company in further
● Academic researchers tend to overvalue molecules drug development:
that emerge very early in the target identification
process. One industry scientist emphasized that “tar- ● Complete characterization of the compound. Various
get identification does not equal druggable target speakers described the “ideal molecule” as smallest
identification.” in its class, non-lipophilic, low propensity to form
● Once drug discovery progresses to stages governed active intermediates or to induce or inhibit liver
by the regulatory framework of good laboratory enzymes, close to soluble, and biased toward CNS
practices (GLPs), academic laboratories are seldom penetrance.
450 Alzheimer’s Association Update / Alzheimer’s & Dementia 3 (2007) 449 – 452

● As much risk and safety data as possible, especially Dementia Prevention Conference:
information about structure-activity relationships Emerging Treatments, Diagnostic
(SARs) and detail about the compound’s biology
beyond the immediate indication. One example Tools and Risk Management
cited is the work, primarily from academic labs, Strategies Could Transform the
showing the need to modulate gamma-secretase ac- Clinical Landscape
tivity to avoid the Notch substrate.
● Preclinical studies that involve enough mice (at least Current treatment options for Alzheimer’s disease are ex-
20 –30) and last long enough to provide meaningful tremely limited, creating a huge unmet need for the 5.1 million
statistical power. Americans living with the disease, their families, and the phy-
● Animal data should include such neuronal endpoints sicians who care for them. At the Alzheimer’s Association
as whether synaptic density or expected levels of cell International Conference on Prevention of Dementia, held June
death changed. 9⫺12 in Washington, D.C., more than 1,000 attendees learned
● In addition to traveling with complete and compelling how that bleak clinical landscape could change as a result of
data, an attractive compound fits well into a compa- accelerating progress toward effective treatments, earlier diag-
ny’s current portfolio and pipeline, and competes well nosis, and risk management.
with the entire potential universe of other candidates Consumer health surveys consistently show that avoid-
that might be pursued. ing cognitive decline is a top concern of older adults. Ex-
tensive media coverage of the Prevention Conference sug-
One recurring theme focused on the urgent need for gests general public interest in this issue is growing. In
validated biomarkers. A speaker cited the case of natali- 2011, 78 million American baby boomers begin turning 65,
zumab (Tysrabi), a multiple sclerosis drug whose clinical reaching the age when risk for Alzheimer’s disease rises
data demonstrated the reliability of gadolinium-enhancing sharply. Expectations and values among this historically
lesions as an MS biomarker. With the advent of this biomar- activist group will drive a growing demand for health care
ker, which is capable of giving a good read on efficacy over that encompasses cognitive well-being and provides access
a 6-month trial, more than 30 companies are now looking at to the best available information and interventions.
MS drug discovery programs. Based on unmet medical need and a potentially robust
Several speakers urged colleagues to consider pooling return on investment, many pharmaceutical companies have
placebo group data from clinical studies in Alzheimer’s embarked on Alzheimer drug discovery programs. Several
disease and mild cognitive impairment (MCI) as one drugs that may slow or stop progression of the disease have
strategy to provide an enhanced benchmark for docu- reached Phase III clinical trials. Phase III is the final stage
menting the effects of potentially disease-modifying in a three-part human testing sequence mandated by the
drugs. An FDA spokesman cited an academic re- Food and Drug Administration (FDA) to demonstrate safety
searcher’s role in taking the initiative to collect all the and effectiveness of a new drug.
placebo data from every monotherapy epilepsy trial every The first convincing clinical data showing that Alzhei-
mer’s can be slowed or stopped will spur further drug
conducted. Despite its long-standing reluctance to accept
development. This proof-of-concept data will also acceler-
“historical” controls, the FDA decided to accept the
ate the search for biological markers that can detect the
pooled epilepsy placebo data as the comparison group for
disease in its earliest stages and monitor effectiveness of
any investigational new epilepsy drug, and adopt an of-
experimental drugs. The quest for biomarkers is already
ficial position that use of a placebo in an epilepsy trial is
under way, with current efforts focusing on brain imaging,
now unethical.
signature proteins in spinal fluid or blood, or characteristic
Hosted by the Alzheimer’s Association, the Research
patterns in protein or gene expression profiles.
Roundtable currently includes 13 members from the phar-
maceutical, imaging, and diagnostic testing industries.
Topic-oriented meetings held each spring and fall have
become two of the most respected events in Alzheimer The Treatment Horizon
research. Watch for a paper providing further details about
the Academic/Industry meeting in a forthcoming issue of The next-generation treatments most advanced in clinical
Alzheimer’s & Dementia: The Journal of the Alzheimer’s testing target beta-amyloid, a protein fragment considered a
Association. prime suspect in Alzheimer’s disease. According to a lead-
New members are always welcomed to the roundtable. ing theory, all individuals produce beta-amyloid, but those
For further information about participation, please contact who develop Alzheimer’s generate abnormally large quan-
Jay Thompson, Alzheimer’s Association Corporate Initia- tities or have reduced ability to clear it from the brain. The
tives, at jay.thomspon@alz.org. “chemically sticky” fragments build up into the plaques
Alzheimer’s Association Update / Alzheimer’s & Dementia 3 (2007) 449 – 452 451

considered one pathological hallmark of the disease, and amyloid. Monoclonal antibodies, produced in the lab-
trigger a cascade of other abnormalities contributing to oratory, represent a “passive immunization” approach
cognitive decline. that does not trigger an immune response or persist in
Prevention Conference sessions provided updates on the the body after drug administration stops.
following amyloid-targeting experimental drugs:
Prevention Conference sessions also highlighted experi-
● Alzhemed (tramiprosate) blocks aggregation of beta- mental drugs targeting other mechanisms:
amyloid into plaques. It is the first anti-amyloid drug ● Dimebon is an antihistamine developed in Russia,
to complete a Phase III clinical trial. Based on an selected for further development as an Alzheimer drug
earlier Phase II study suggesting Alzhemed reduced by the Russian Academy of Sciences after it showed
beta-amyloid levels in cerebrospinal fluid and stabi- several potential avenues of effectiveness. New 12-
lized patients with the mildest Alzheimer’s disease, month data from a Russian Phase II study directed by
Phase III results were highly anticipated. Conference two prominent American clinical trialists showed sig-
attendees were disappointed to learn that no results nificant benefit in cognition, global function, and be-
were yet available because unusually high data varia- havior. Developer Medivation is considering a U.S.
tions among trial sites invalidated the statistical Phase III trial.
model. Alzhemed developer Neurochem is working ● Ketasyn (AC-1202), taken in a modified nutritional
with the FDA to develop a new model reducing the supplement “milkshake,” may correct metabolic defi-
impact of site variations. Possible factors responsible ciencies in the Alzheimer brain by providing ketone
for the variation include differences in the number of bodies, an alternate energy source for declining cells
participants taking memantine, cholinesterase inhibi- that have lost the ability to use glucose. Phase II
tors, antidepressants or vitamin E. According to Neu- studies suggest Ketasyn improves memory in people
rochem, trial results could be announced soon. with Alzheimer’s who do not have a common risk
● Flurizan (tarenflurbil) reduces beta-amyloid by gene for the disease. Developer Accera is considering
changing the activity of an enzyme involved in its a Phase III study.
production. A conference session reported results of a ● Avandia (rosiglitazone), a PPAR-gamma receptor ag-
12-month follow-on study to a year-long Phase II trial. onist currently FDA-approved for Type 2 diabetes, is
After 24 months of treatment, patients in the mildest under investigation in several Phase III trials for its
Alzheimer stages who received the highest drug dose potential to improve brain glucose utilization. A con-
showed significant stabilization or improvement in ference session summarized early-stage Alzheimer
cognition and overall function. Two large Phase III trial results and reviewed recent data linking Avandia
studies sponsored by developer Myriad are now under to an increased risk of heart attack and stroke.
way testing that highest Phase II dose in early-stage ● Lipitor (atorvastatin) is being tested as a treatment for
patients. mild-to-moderate Alzheimer’s in a large Phase III
● LY450139 blocks activity of the same amyloid- study under way at 97 sites in 10 countries, with
generating enzyme as Flurizan. The latest Phase II results expected in 2008. Statins may impact Alzhei-
results showed LY450139 decreased beta-amyloid mer’s disease through several mechanisms, including
levels in both blood and spinal fluid. Developer Lilly general benefit for brain blood vessels and anti-in-
plans to launch a Phase III trial in 2008. flammatory effects. Some statins, including Lipitor,
● AN-1792, the first drug in the anti-amyloid class ever may also influence beta-amyloid production.
to reach clinical trials, was a “vaccine” that mobilized
the immune system to produce beta-amyloid antibod-
ies. A Phase II trial was halted in 2002 after 6 percent
of recipients developed encephalitis, but AN-1792 de- Improved Diagnosis and Monitoring
velopers Elan and Wyeth continued to monitor partic-
ipants. In follow-up, vaccine recipients who devel- As demonstrated by early-stage data on Alzhemed, Fluri-
oped the highest levels of anti-amyloid antibodies zan, and other trials reported elsewhere, evidence suggests
have consistently shown encouraging hints of benefit. that drugs aimed at fundamental Alzheimer pathology may
The new four-year data shows trends toward less de- work best when taken as early as possible. The current
cline in memory and overall function; greater likeli- mainstay of Alzheimer diagnosis is cognitive evaluation,
hood of living at home rather than in long-term care; often with the mini-mental status examination (MMSE).
and no further cases of encephalitis or difference in The MMSE is imprecise and insensitive to the first signs of
survival. Elan and Wyeth announced in May 2007 that cognitive change. Extensive neuropsychiatric test batteries
they would move forward in 2008 with a Phase III can detect more subtle deficits, but they are not a practical
trial of AAB-001, a monoclonal antibody against beta- alternative in general practice as they are time-consuming,
452 Alzheimer’s Association Update / Alzheimer’s & Dementia 3 (2007) 449 – 452

unpopular with patients, and often denied reimbursement by characteristic pattern may distinguish people at in-
third-party payers. creased risk or in the earliest stages of Alzheimer’s.
Better approaches to diagnosis and monitoring are
clearly needed. Two of the most promising avenues are
brain imaging and biomarkers in spinal fluid or blood. Emerging Prevention Strategies
A packed Prevention Conference session brought attend-
ees up to date on the Alzheimer’s Disease Neuroimaging Early Alzheimer trial data and experience with other dis-
Initiative (ADNI). This nationwide study aims to standard- eases suggest primary prevention may be the most effective
ize brain imaging protocols for cognitive change and vali- intervention. A conference keynote address by Julie Gerb-
date their usefulness in early diagnosis and monitoring. erding, M.D., M.P.H., director of the Centers for Disease
Both magnetic resonance imaging (MRI) and positron emis- Control and Prevention (CDC), highlighted these leading
sion tomography (PET) are under investigation. ADNI is potentially modifiable risk factors for cognitive decline:
also collecting blood and spinal fluid samples to explore the ● Physical inactivity
accuracy of biomarkers. All ADNI data will be accessible to ● Cardiovascular risk factors, including elevated blood
any qualified investigator. pressure and cholesterol, insulin resistance and Type 2
Although ADNI is funded chiefly by the National Insti- diabetes, smoking, and excess weight
tute on Aging (NIA), a number of commercial interests and ● Psychosocial issues (engagement, anxiety, sense of
organizations, including the Alzheimer’s Association, are control over life)
also supporting the initiative. Alzheimer’s Association
The CDC has launched The Healthy Brain Initiative, a
funding includes support for evaluation of PET scans using first-ever road map to make cognitive well-being part of our
Pittsburgh compound B (PIB) as a strategy to measure brain overall national public health goals. CDC will collaborate
beta-amyloid levels. Initial development of PIB was funded with the Alzheimer’s Association to develop a pilot com-
by the Alzheimer’s Association. The Association is also munity-level intervention program over the next few years.
funding an effort to bring brain imaging and biomarker
studies in progress in Europe and Asia into compliance with
ADNI protocols, broadening the power and scope of the Honoring Leon Thal: The Tomorrow’s
initiative.
It is unlikely any imaging technology or biomarker will Leaders Prize
be widely accepted until ADNI results are available in Dementia research lost one of its most widely admired
about five years. In the meantime, individual investigators leaders when Leon J. Thal, M.D., died in a January 2007
and companies continue to report promising results with private plane crash. Thal, a visionary in designing clinical
specific approaches. Conference sessions highlighted bi- trials, led the Alzheimer’s Disease Cooperative Study
omarker strategies focusing on: (ADCS), a federally funded consortium conducting clinical
studies, including ADNI. He was also a major force in
● Signature proteins in cerebrospinal fluid (CSF). creation and scientific leadership of the Prevention Confer-
Results from research settings suggest these measures ence, so the event provided a fitting venue to announce a
may be the most sensitive and specific current options. new prize honoring the legacies of Thal and George Glen-
However, experts acknowledge that the lumbar punc- ner, who isolated and identified beta-amyloid as the chief
ture (LP) needed to evaluate CSF status is unlikely to component of plaques. The Tomorrow’s Leaders in Alzhei-
achieve general acceptance in the United States. mer’s Disease Research Prize, jointly sponsored by the
● Signature proteins in blood. Poor chances of LP Alzheimer’s Association, the Cure Alzheimer’s Fund, and
acceptance have spurred efforts to correlate blood the Lou Ruvo Brain Institute, will award annual prizes of
levels with brain levels of key proteins. Although $100,000 to talented younger scientists with a record of
these correlations are less well characterized than CSF extraordinary current achievement and outstanding potential
benchmarks, some strategies are generating promising for important future work. Modeled on other “genius”
data. grants, awardees can use the prize funds for any purpose
● “Biomarker barcodes” in blood. These approaches they wish. More details about the Tomorrow’s Leaders
focus on multi-component protein or gene expression Prize and application procedures will be available soon
profiles in which no single element is predictive, but a from the sponsoring organizations.

You might also like