Demetia
Demetia
Demetia
Movement Disorders
in Dementias
123
Movement Disorders in Dementias
Marcelo Merello • Sergio E. Starkstein
Editors
Movement Disorders
in Dementias
Editors
Marcelo Merello, MD, PhD Sergio E. Starkstein, MD, PhD
Neuroscience Department School of Psychiatry
Raul Carrea Institute for and Clinical Neurosciences
Neurological Research (FLENI) University of Western Australia Fremantle
Universidad Catolica Argentina Fremantle, WA
Buenos Aires Australia
Argentina
Neurologists today need to deal in their daily practice with cognitive impairment in
patients with movement disorders, particularly but not limited to those with
Parkinson’s disease. This book offers a somewhat opposite view, which is the occur-
rence of movement disorders in patients in whom dementia is the primary clinical
manifestation. Movement disorders, as currently understood, comprise a large vari-
ety of conditions, not all of which are related to basal ganglia dysfunction. However,
the contribution of the ascending dopaminergic projection to cognitive processing is
well known, and, indeed, the now old-fashioned concept of “subcortical” dementia
originated from observations in patients with progressive supranuclear palsy. On the
other hand, it is now recognized that cognitive impairment very often evolves in
parallel with altered gait and equilibrium and that the pathology of many neurode-
generative diseases is sufficiently widespread to impinge upon several circuits and
brain regions, thus producing multiple clinical combinations. No doubt, dementia
and movement disorders are a current hot topic.
Drs. Merello and Starkstein, therefore, deserve the most sincere and warm con-
gratulation for this initiative and the outcome. This book provides a comprehensive
account of a variety of different movement disorders in the setting of the major
diseases and processes associated with cognitive impairment. The list of authors is
impressive, as experts in the field have written each chapter. Nowadays, the world
of publishing is moving quickly toward electronic editions only in what seems an
inevitable trend. However, Merello and Starkstein’s Movement Disorders in
Dementias is exactly the book one wants to have on the desk to read leisurely and
consult very often when thinking about specific patients with cognitive and motor
impairments. I am looking forward to the smell of a new, just-printed, highly intel-
lectual work!
vii
Preface
Since the end of the twentieth century, the cognitive and psychiatric comorbidities
of movement disorders have received increasing attention. On the other hand, the
study of movement disorders in dementia has been relatively neglected. To our
knowledge, there are no specific books devoted to clarify the variety, mechanism,
and treatment of motor problems in dementia.
It was 14 years ago that we decided to write a book on the non-motor problems
of Parkinson’s disease (PD). By then, research and clinical evidence were rapidly
accumulating regarding the high frequency of specific cognitive deficits, affective
disorders, and anxiety in PD. Apathy was also emerging as a prominent comorbid
condition of PD, and the mechanism and treatment of psychotic symptoms were an
area of intense investigation. We now present a book that inverts that focus, that is,
it examines the motor problems in dementias due to different mechanisms.
An increasing number of studies have demonstrated a high frequency of parkin-
sonism and other movement disorders such as myoclonus, paratonia, and dyskinesia
in Alzheimer’s disease (AD). In addition, the introduction of medication to treat the
cognitive impairment as well as novel antipsychotic drugs contributed to the variety
and severity of movement disorders in AD. The association between dementia and
movement disorders is also evident in the high proportion of patients with fronto-
temporal dementia and comorbid parkinsonism and the association between cogni-
tive deficits and a variety of movement disorders such as the “alien hand syndrome”
and “psychomotor” symptoms such as apraxia in corticobasal syndrome.
Cognitive dysfunction and parkinsonism are closely related, and regardless of
which of them is the primary problem, the other is invariably present. For many
years, dementias and parkinsonism have been separately addressed, through differ-
ent specialties, by different researchers, and in different books and journals.
However, emerging concepts on topics such as neurodegeneration with synucle-
inopathies, tauopathies, and amyloid deposit mechanisms have generated a trend to
lump these disorders together.
In his masterpiece The History of Mental Symptoms, German Berrios asks what
he considers the crucial question in the history of PD: why did it take so long for
cognitive and psychiatric symptoms to be considered part and parcel of PD? The
ix
x Preface
answer to this question is that patients did not live long enough to show the non-
motor comorbidities or that, following James Parkinson’s description, neurologists
refused to accept their presence. Perhaps a similar process occurs in dementia, with
most of the focus being given to the cognitive aspects of this disorder, while the
motor aspects are not so well attended. It is interesting that in his seminal paper “On
the relationship between senile cerebral atrophy and aphasia,” Arnold Pick already
described motor problems in patients with dementia. The patient Augustus H had
brisk knee reflexes and a fast clonus, a 52-year-old man developed progressive
weakness of the right extremities and speech disturbance, and the third patient com-
plained of pain in the right leg “gradually losing the use of it.”
This book will mainly focus on extrapyramidal signs and symptoms in the most
common or novel types of dementia and will address the issue of the artificial
boundary between dementia and parkinsonism, the two most common degenerative
disorders.
Recognized specialists in the field of movement disorders provided chapters on
topics generally restricted to dementia experts. The first chapters address important
general aspects on the relationship between motor disorders and dementia, such as
the association between medications and motor problems, and motor disorders
common to many types of dementia, such as gait disorders, falls, and motor mani-
festations of psychiatric complications of dementia. The following chapters provide
an in-depth analysis on the relationship between motor and cognitive symptoms,
addressing their common pathogenesis and specific treatments.
The book was timely written in 1 year, which warrants up-to-date information
and views. We hope we have covered the topic widely enough, so that the book will
appeal to a wide readership, including general practitioners, gerontologists, and
neurologists. We expect this book to become the main reference in the field for
years to come.
xi
xii Contents
xiii
xiv Contributors
Introduction
In 1942, the great Argentine writer Jorge Luis Borges wrote an essay entitled “El
idioma analítico de John Wilkins” (The Analytical Language of John Wilkins) in
which he laid out the challenges of human attempts to classify the world (Borges
2001). To illustrate his argument, Borges reproduced a classification of animals
purportedly found in “a certain Chinese encyclopedia entitled “Celestial
[Emporium]” of Benevolent Knowledge.” Borges observed that “it is clear that there
is no classification of the Universe not being arbitrary and full of conjectures.” It
may appear odd that Borges should see conjectures in classifications, which are by
definition rather explicit. This is because he considered that a full understanding of
things to be classified and of their mutual relationships is necessary for the classifi-
cation to make sense (Borges 2001).
Over 100 neurodegenerative disorders affect humans, among which many over-
lap either clinically or pathologically, rendering their practical classification most
challenging. The issue is further complicated by the fact that different combinations
of lesions can give rise to different clinical conditions (Luheshi and Dobson 2009;
Burn and Jaros 2001). Furthermore, the same neurodegenerative processes, espe-
cially at the beginning, can affect different areas of the brain, thus varying signifi-
cantly at different stages of the neurodegenerative process.
Classic categorization of neurodegenerative disorders is based on predominant
lesion topography. Thus, they may be grouped, for instance, as cortical, of the basal
ganglia, cerebellum, or as motoneuron disorders. Or diseases may be classified
based on main clinical features into dementias, movement disorders, motoneuron
disease, or ataxias. However, topographical classification of the disorders does not
coincide with expected clinical manifestations. So neurodegeneration predomi-
nantly affecting the cerebral cortex may induce dementia or other non-dementing
conditions, whereas diseases that predominantly involve the basal ganglia in
1 Neurodegenerative Disorders 3
The complexity linked to classifying the majority of patients seen in clinical prac-
tice testifies the extent to which the boundaries between different disorders may in
reality be less distinct than previously believed and have inspired some researchers
to suggest that neurodegenerative diseases characterized by abnormal protein
deposits should be viewed as existing along a continuum of symptoms and patholo-
gies, rather than as discrete entities. For instance, Armstrong et al, in an elegant
review (Armstrong 2012), have postulated that different models of neurodegenera-
tive disorders could be argued. They hypothesize that although distinct diseases
may exist (“discrete” model), they may also exhibit overlapping features (“overlap”
model), and the most challenging concept they put forward is that distinct diseases
do not really exist, but form part of a “continuum” in which there is constant varia-
tion in clinical/pathological features from one case to another (“continuum” model).
This last one would infer that many different pathways may exist through which
different individuals ultimately present the same process, abnormal protein deposi-
tion. It is worth noting that the “hallmark molecular findings” themselves are not
necessarily the cause of the underlying disease and clinical symptoms. Instead they
may be a response to the disease processes, although at some stage the “hallmark
4 M. Merello and M. Rossi
Clinical Classification
Parkinson´s plus disorders is a term proposed by Stanley Fahn and refers to dis-
orders with features of parkinsonism in addition to other neurologic feature, such as
ophthalmoplegia, ataxia, dysautonomia, amyotrophy, cortical signs, cerebellar
signs, or dementia. This group presents multiple system atrophies (parkinsonian,
cerebellar, and mixed forms), including corticobasal degeneration, PSP, parkinson-
ism, and motor neuron disease. Combination of parkinsonism with dementia such as
in Guamanian complex, Creutzfeldt–Jakob, Alzheimer’s, and Pick, as well as vari-
ous hereditary diseases in which parkinsonism and dementia can be present together,
such as Wilson, NBIA, Huntington’s, and spinocerebellar-nigral degenerations, are
also included in the classification. Other non-degenerative disorders such as normal
pressure hydrocephalus also correspond to this category. Parkinsonism occurring in
dopa-responsive dystonia, also known as benign parkinsonism, which can have its
onset in adults but is also seen in children, completes the group (Table 1.2).
Dementia is a clinical syndrome characterized by acquired loss of cognitive and
emotional abilities, severe enough to interfere with daily functioning and quality of
life. Several dementia classifications have been attempted from the clinical point of
view (Table 1.3). Neuropsychological profiles of dementia reflect the impact of dis-
ease on distinctive neuroanatomic networks associated with complex cognitive
domains. For example, prominent amnesia is associated with medial temporal dys-
function, whereas aphasia is a consequence of left perisylvian dysfunction. The rela-
tionship between clinical symptoms and underlying neuropathology, however, is
less straightforward, as indicated by the multiple neuropathological diagnoses asso-
ciated with the various clinical dementia syndromes (Mesulam 2000). It has been
6 M. Merello and M. Rossi
long said from both the clinical (Pillon et al. 1993) and etiological perspectives
(Darvesh and Freedman 1996) that dementia can be split into cortical and subcorti-
cal types. The typical clinical findings of cortical dementing processes include
prominent memory lost, dyscalculia, dysphasias, dyspraxias, and agnosias. The dif-
ferentiating features of subcortical dementia were said to be a profound slowing of
cognition, milder memory disturbances, frontal executive dysfunction, and changes
in personality and affect in the absence of aphasias, apraxias, and agnosias
(Cummings 1986). While this has been initially generally accepted, other authors
have highlighted the difficulties with the distinction, arguing that the neuropsycho-
logical profiles of cortical and subcortical cases are not sufficiently dissimilar
(Brown and Marsden 1988). Furthermore, cortical symptomatology can often occur
in the so-called subcortical disease and vice versa (Hughes et al. 1993). Perhaps
constant redefinition and reclassification of the frontotemporal syndromes represent
the best example of overlap and lack of boundaries between cortical and subcortical
motor and cognitive disorders. The psychiatric manifestations of subcortical disease
come primarily in the form of personality changes and affective disorders. Apathy
and irritability are particularly common (Aarsland and Karlsen 1999), and depres-
sion is said to be significantly more common in subcortical disorders such as
Parkinson’s disease than in Alzheimer’s (Aarsland and Karlsen 1999). While amnes-
tic symptoms of dementia have the highest likelihood of being associated with
dementia of AD type (DAT) pathology, early aphasia, progressive visuospatial defi-
cits, and changes in personality are usually seen in primary progressive aphasias,
semantic dementia, or behavioral forms of frontotemporal dementia, which can also
be associated with AD neuropathology (Figs. 1.1 and 1.2). As dementia progresses
from early to late stages, symptom domain boundaries become blurred, and distinc-
tive profiles disappear, with dementia becoming a single terminal clinical entity.
1 Neurodegenerative Disorders 7
FTD-
AD DLBD
Pick complex
PPA
Nonfluent Semantic Logopenic AD
variant variant variant variant
of dlbd
PAOS
Fig. 1.1 Clinical and molecular relationship within the main types of dementia (circles) and
between them and the major movement disorders syndromes (squares). FTP frontotemporal
dementia, AD Alzheimer’s disease, DLBD dementia with Lewy bodies disease, PPA primary pro-
gessive aphasia, PAOS progressive apraxia of speech, CBS vorticobasal syndrome, PSP progres-
sive supranuclear palsy, IPD idiopathic Parkinson’s disease
Dementia
Early cerebellar/
Mainaly cognitive Early behavioural/ pyramidal signs/
Early Parkinsonism language alterations akinetic mutism/
alteration
seizures
DLBD
PSP FTD
AD CBD CJD
PPA
CJD
Molecular Classification
β-Amyloid derives from the amyloid precursor protein (APP). APP undergoes proteo-
lytic cleavages by β- and γ-secretases to generate β-amyloid (Kayed et al. 2003). Since
secretases are linked to presenilin genes, presenilins are implicated in the proteolytic
Tau: Tauopathies
The major proteins, but not the only ones, that accumulate in the most common
forms of FTD and ALS are RNA–DNA binding proteins and mutations in the TDP-
43 gene, found in both disorders (Neumann et al. 2006). Another member of the
family, the FUS-TLP, has been found mutated and accumulated in neuronal inclu-
sions in rare forms of FTD and ALS (Mackenzie and Rademakers 2007).
α-Synuclein: Synucleinopathies
Since α-synuclein was identified as the major structural component of Lewy bod-
ies, Parkinson’s disease was raised to the level of major class disease.α-Synuclein
is a phosphoprotein. The physiological function of α-synuclein is not known; it is
a lipid-binding protein located in neurons at the synapse level, where it is involved
in vesicle traffic and neurotransmission (Goedert 2001a, b; Abeliovich et al.
2000). Other functions such as chaperone protein, inhibitor of phospholipase D2,
and participation in oxidative stress have also been described. Lewy bodies are
intracytoplasmic inclusions containing α-synuclein aggregations and the hall-
mark pathological signature of Parkinson’s disease and diffuse Lewy body dis-
ease. However, they are also present in 7–37 % of normal aging individuals
(Zaccai et al. 2008) and most notably in cases of Alzheimer’s disease (Hansen
et al. 1990).
Aside from being present in normal aging, Lewy bodies have been described in
conditions other than α-synuclein disorders such as AD, PSP, Guam disease, FTD,
and Down syndrome (Ozawa et al. 2004; Uchikado et al. 2006; Dickson et al. 2002;
Jellinger 2008; Yamazaki et al. 2000; Gregory et al. 2009). Less commonly, they
have also been found in Niemann–Pick type C, a sphingolipid storage disorder
which results from autosomal recessive inherited deficiencies of lysosomal and
intracellular lipids, trafficking proteins (Carstea et al. 1997). Acid β-glycosidase-
associated neurodegeneration is an inherited disorder in which an enzyme known
as β-glycosidase loses its capacity to catalyze glucosylceramide breakdown to
ceramide causing Gaucher disease types I, II, and III (Clark et al. 2009; Sidransky
1 Neurodegenerative Disorders 11
Polyglutamine Disorders
Prion Diseases
Neuroferritinopathies
Recent genetic and molecular data have led to considerable changes in the classifi-
cation and nomenclature of tauopathies (Williams 2006; Hasegawa 2006; van
Slegtenhorst et al. 2000). Original simplistic molecular classifications have become
considerably more complex, mainly as a result of studies performed in FTD group
cases. In light of the described complexity, discrimination between different disease
entities is often only possible after describing the location of “hallmark molecular
inclusions.” These may be in the cytoplasm (neuronal cytoplasmatic inclusions
NCI), in the nuclei (neuronal intranuclear inclusions NII), or in glial cells (glial
inclusions GI). The latter may in turn be glial cytoplasmic inclusions (GCI) or astro-
cytic “plaques” (Neumann et al. 2006).
Conclusion
References
Brown RG, Marsden CD. ‘Subcortical dementia’: the neuropsychological evidence. Neuroscience.
1988;25(2):363–87.
Braga-Neto P, Felicio AC, Hoexter MQ, et al. Cognitive and olfactory deficits in Machado-Joseph
disease: a dopamine transporter study. Parkinsonism Relat Disord. 2012;18(7):854–8.
Burn DJ, Jaros E. Multiple system atrophy: cellular and molecular pathology. Mol Pathol.
2001;54:419–26.
Carstea ED, Morris JA, Coleman KG, et al. Niemann-Pick C1 disease gene: homology to media-
tors of cholesterol homeostasis. Science. 1997;277:228–31.
Clark LN, Kartsaklis LA, Wolf Gilbert R, et al. Association of glucocerebrosidase mutations with
dementia with Lewy bodies. Arch Neurol. 2009;66:578–83.
Cummings CJ, Zoghbi HY. Trinucleotide repeats: mechanisms and pathophysiology. Annu Rev
Genomics Hum Genet. 2000;1:281–328.
Cummings JL. Subcortical dementia. Neuropsychology, neuropsychiatry, and pathophysiology. Br
J Psychiatry. 1986;149:682–97.
Darvesh S, Freedman M. Subcortical dementia: a neurobehavioral approach. Brain Cogn.
1996;31(2):230–49.
Dickson DW, Bergeron C, Chin SS, et al. Office of Rare Diseases neuropathologic criteria for
corticobasal degeneration. J Neuropathol Exp Neurol. 2002;61:935–46.
Dubois B, Burn D, Goetz C, et al. Diagnostic procedures for Parkinson’s disease dementia: recom-
mendations from the movement disorder society task force. Mov Disord. 2007;22(16):
2314–24.
Duckett S, Stern J. Origins of the Creutzfeldt and Jakob concept. J Hist Neurosci. 1999;8:21–34.
Durr A. Autosomal dominant cerebellar ataxias: polyglutamine expansions and beyond. Lancet
Neurol. 2010;9(9):885–94.
Espay AJ, Litvan I. Parkinsonism and frontotemporal dementia: the clinical overlap. J Mol
Neurosci. 2011;45(3):343–9.
Fahn S, Jankovic J, Hallett M. Principles and practice of movement disorders. 2nd ed. Philadelphia:
Elsevier; 2011.
Feany MB, Dickson DW. Neurodegenerative disorders with ex-tensive tau pathology: a compara-
tive study and review. Ann Neurol. 1996;40:139–48.
Förstl H. The Lewy body variant of Alzheimer’s disease: clinical, pathophysiological and concep-
tual issues. Eur Arch Psych Clin Neurol. 1999;249:64–7.
Galvin JE, Lee VM, Trojanowski JQ. Synucleinopathies: clinical and pathological implications.
Arch Neurol. 2001;58:186–90.
Galpern WR, Lang AE. Interface between tauopathies and synucleinopathies: a tale of two pro-
teins. Ann Neurol. 2006;59(3):449–58.
Goedert M, Spillantini MG. Tau gene mutations and neurodegeneration. Biochem Soc Symp.
2001;67:59–71.
Goedert M. The significance of tau and α-synuclein inclusions in neurodegenerative disease. Curr
Opin Genet Dev. 2001a;11(3):343–51.
Goedert M. Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci. 2001b;2:492–501.
Goldstein LH, Abrahams S. Changes in cognition and behaviour in amyotrophic lateral sclerosis:
nature of impairment and implications for assessment. Lancet Neurol. 2013;12(4):368–80.
Graeber MB, Kosel S, Egensperger R, Banati RB, Muller U, Bise K, Hoff P, Moller HJ, Fujisawa
K, Mehraein P. Rediscovery of the case described by Alois Alzheimer in 1911: historical, his-
tological and molecular genetic analysis. Neurogenetics. 1997;1:73–80.
Gregory A, Polster BJ, Hayflick SJ. Clinical and genetic delineation of neurodegeneration with
brain iron accumulation. J Med Genet. 2009;46:73–80.
Hainfellner JA, Wanschitz J, Jellinger K, Liberski PP, Gullotta F, Budka H. Coexistence of
Alzheimer-type neuropathology in Creutzfeldt-Jakob disease. Acta Neuropathol.
1998;96:116–22.
Hansen L, Salmon D, Galasko D, Masliah E, Katzman R, DeTeresa R, Thal L, Pay MM, Hofstetter
R, Klauber M, et al. The Lewy body variant of Alzheimer’s disease: a clinical and pathologic
entity. Neurology. 1990;40(1):1–8.
14 M. Merello and M. Rossi
Hardy J. Alzheimer’s disease: the amyloid cascade hypothesis: an update and reappraisal. J
Alzheimers Dis. 2006;9(3 Suppl):151–3.
Hasegawa M. Biochemistry and molecular biology of tauopathies. Neuropathology. 2006;
26(5):484–90.
Holton JL, Ghiso J, Lashley T, et al. Regional distribution of amyloid-Bri deposition and its asso-
ciation with neurofibrillary degeneration in familial British dementia. Am J Pathol.
2001;158:515–26.
Holton JL, Lashley T, Ghiso J, et al. Familial Danish dementia: a novel form of cerebral amyloido-
sis associated with deposition of both amyloid-Dan and amyloid-beta. J Neuropathol Exp
Neurol. 2002;61:254–67.
Hughes T, Ross H, Madeley P, Finlayson G, Mindham RH, Biggins CA. Subcortical dementia.
BMJ. 1993;307(6902):503.
Hutton M. Missense and splice site mutations in tau associated with FTDP-17: multiple pathogenic
mechanisms. Neurology. 2001;56(11 Suppl 4):S21–5.
Jellinger KA. Neuropathological aspects of Alzheimer disease, Parkinson disease and frontotem-
poral dementia. Neurodegener Dis. 2008;5:118–21.
Jellinger KA, Attems J. Prevalence and impact of vascular and Alzheimer pathologies in Lewy
body disease. Acta Neuropathol. 2008;115(4):427–36.
Katsuno M, Banno H, Suzuki K, Adachi H, Tanaka F, Sobue G. Molecular pathophysiology and
disease-modifying therapies or spinal and bulbar muscular atrophy. Arch Neurol.
2012;69(4):436–40.
Kayed R, Head E, Thompson JL, et al. Common structure of soluble amyloid oligomers implies
common mechanism of pathogenesis. Science. 2003;300:486–9.
Khateeb S, Flusser H, Ofir R, Shelef I, Narkis G, Vardi G, Shorer Z, Levy R, Galil A, Elbedour K,
Birk OS. PLA2G6 mutation underlies infantile neuroaxonal dystrophy. Am J Hum Genet.
2006;79(5):942–8.
Koeppen AH, Mazurkiewicz JE. Friedreich ataxia: neuropathology revised. J Neuropathol Exp
Neurol. 2013;72(2):78–90.
Koide R, Ikeuchi T, Onodera O, et al. Unstable expansion of CAG repeat in hereditary dentatorubral-
pallidoluysian atrophy (DRPLA). Nat Genet. 1994;6:9–13.
Kotzbauer PT, Cairns NJ, Campbell MC, Willis AW, Racette BA, Tabbal SD, Perlmutter JS.
Pathologic accumulation of α-synuclein and Aβ in Parkinson disease patients with dementia.
Arch Neurol. 2012;69(10):1326–31.
Kremen SA, Mendez MF, Tsai PH, Teng E. Extrapyramidal signs in the primary progressive apha-
sias. Am J Alzheimers Dis Other Demen. 2011;26(1):72–7.
Lee S, Antony L, Hartmann R, Knaus KJ, Surewicz K, Surewicz WK, Yee VC. Conformational
diversity in prion protein variants influences intermolecular beta-sheet formation. EMBO
J. 2010;29(1):251–62.
Litvan I, Agid Y, Calne D, Campbell G, Dubois B, Davoisen RC, Goetz CG, Golbe LI, Grafman J,
Growden JH, Hallett M, Jankovic J, Quinn NP, Tolisa E, Zee DS, Chase TW, FitzGibbon EJ,
Hall Z, Juncos J, Nelson KB, Oliver E, Pramstaller P, Reich SG, Verny M. Clinical research
criteria for the diagnosis of progressive supranuclear palsy (Steele-Richardson-Olszewski syn-
drome): report of the NINDS-SPSP International Workshop. Neurology. 1996;47:
1–9.
Ludolph AC, Kassubek J, Landwehrmeyer BG, et al. Tauopathies with parkinsonism: clinical
spectrum, neuropathologic basis, biological markers, and treatment options. Eur J Neurol.
2009;16(3):297–309.
Luheshi LM, Dobson CM. Bridging the gap: from protein misfolding to protein misfolding dis-
eases. FEBS Lett. 2009;583:2581–6.
Mackenzie IR, Rademakers R. The molecular genetics and neuropathology of frontotemporal
lobar degeneration: recent developments. Neurogenetics. 2007;8:237–48.
Mackenzie IR, Neumann M, Bigio EH, et al. Nomenclature and nosology for neuropathologic
subtypes of frontotemporal lobar degeneration: an update. Acta Neuropathol. 2010;119(1):
1–4.
1 Neurodegenerative Disorders 15
Williams DR. Tauopathies: classification and clinical update on neurodegenerative diseases associ-
ated with microtubule-associated protein tau. Intern Med J. 2006;36(10):652–60.
Yamazaki M, Arai Y, Baba M, et al. Alpha-synuclein inclusions in amygdala in the brains of
patients with the parkinsonism-dementia complex of Guam. J Neuropathol Exp Neurol.
2000;59:585–91.
Zaccai J, Brayne C, McKeith I, Matthews F, Ince PG, MRC Cognitive Function, Ageing
Neuropathology Study. Patterns and stages of alpha-synucleinopathy: relevance in a population-
based cohort. Neurology. 2008;70(13):1042–8.
Zhou B, Westaway SK, Levinson B, Johnson MA, Gitschier J, Hayflick SJ. A novel pantothenate
kinase gene (PANK2) is defective in Hallervorden-Spatz syndrome. Nat Genet. 2001;28(4):
345–9.
Chapter 2
Gait Disorders in Patients with Cognitive
Impairment or Dementia
Introduction
aspects of functional decline of the nervous and the musculoskeletal systems; how-
ever, the extent to which these can be attributed to normal aging is not clear
(Alexander 1996; Hausdorff 2007).
Many studies have quantitatively examined the kinematics of walking in older
people in order to define normal walking. Most of these studies have found that dur-
ing aging, gait velocity decreases, stride length becomes shorter, the base of support
widens to increase stability, and double-limb support and stride-to-stride variability
increase (Giladi et al. 2005; Maki 1997; Verghese and Xue 2011). The magnitude of
these changes depends, to a large degree, on the inclusion and exclusion criteria
applied. Among older adults with very “successful aging,” these changes in gait
may be small and nonsignificant (Odenheimer et al. 1994), whereas gait dysfunc-
tion can become debilitating in others. Imaging studies have been used to better
understand the origin and impact of clinical changes in gait among older adults. For
example, the volume of white matter hyperintensities (WMH) and/or periventricu-
lar high signals and ventricular volumes have been associated with mobility prob-
lems and falls among older adults (Holtzer et al. 2006; Maki 1997; Odenheimer
et al. 1994; Yogev-Seligmann et al. 2008). The source of these subclinical changes
and the impact of microvascular alterations on gait among older adults remain to be
more fully delineated. Nonetheless, despite the heterogeneity in function and the
existence of a subgroup of individuals among whom gait appears to be no different
from that of younger adults, there is some decline in gait that can generally be
observed in older adults that is partially related to aging.
Gait disorders are common among subjects with cognitive impairments (Chen et al.
2012; Holtzer et al. 2006; Yogev-Seligmann et al. 2008). Gait is a complex task that
requires cognitive input from multiple systems to maintain upright posture, in addi-
tion to the reliance on the motor control system (Hausdorff et al. 2005; Montero-
Odasso et al. 2012c; Woollacott and Shumway-Cook 2002; Yogev-Seligmann et al.
2008). Walking in the real world is not an automatic motor task since it integrates
high-level cognitive functions, such as attention, obstacle negotiation, and set shift-
ing, in order to successfully ambulate in complex environments while carrying out
multiple tasks (Yogev-Seligmann et al. 2008). Given these requirements of every-
day walking, it is not surprising that emerging evidence suggests that early distur-
bances in cognitive abilities are associated with slower gait and gait instability
during both single- and dual-task walking (Montero-Odasso et al. 2009a).
Risk of Falls
Falls are a serious health problem in the elderly population (Ganz et al. 2007). Falls
contribute to functional decline and loss of mobility and independence, and
20 M. Dorfman et al.
recurrent falls reflect frailty, immobility, and acute and chronic health impairments.
The impact of falls is a major concern since about one-third of older adults living in
the community over the age of 65 years experience falls every year (Hausdorff et al.
2001; Tinetti 1994), with the figure reaching as high as 50 % among adults over the
age of 85 years (Blake et al. 1988). Cognitive impairment and dementia are known
risk factors for falls (Harlein et al. 2009), and the incidence of falls in older people
with cognitive impairment is approximately three times higher than that of cogni-
tively intact adults, ranging from 1.1 to 6.4 (Asada et al. 1996; de Carle and Kohn
2001; Kallin et al. 2004; Shaw 2007; Tinetti et al. 1988; van Doorn et al. 2003). The
higher risk of falls among people with dementia increases the risk for serious inju-
ries, morbidity, and institutionalization (Kallin et al. 2004; Tinetti 1994; van Dijk
et al. 1993). Furthermore, the population of fallers with cognitive decline is approxi-
mately five times more likely to be admitted to institutional care compared to people
with cognitive problems who do not fall (Morris et al. 1987). Cognitive decline
apparently exacerbates both the risk of falls and the impact of falls on function and
health-related quality of life.
Until recently, falls and dementia had been studied and assessed as separate
geriatric disorders. These two events are now considered as being intertwined,
and so studying their interconnection paves the way to better understanding of
motor–cognitive interactions (Montero-Odasso et al. 2012c; Sheridan and
Hausdorff 2007) (see Fig. 2.1). In general, most falls among older adults occur
during weight shifting while walking particularly while attention is directed to
another task (Nevitt and Cummings 1993). There is interplay between gait vari-
ability, cognition, a particular executive function (EF), attention, and the risk of
falls (Sheridan and Hausdorff 2007). Falls can be described as a complex, multi-
factorial phenomenon which is caused by several risk factors (Ganz et al. 2007).
Eight categories of risk factors were identified in a review by Harlein et al.
(Harlein et al. 2009). According to those authors, the type and severity of demen-
tia, motor impairments, vision impairments, behavioral disturbances, functional
impairments, fall history, neuroleptics, and low bone mineral density play an
important role as factors that can lead to falls among people with cognitive decline.
The association between type of dementia and falls was examined by Ballard
et al. (1999) who noted that people with dementia with Lewy bodies were found
to be more likely to fall and that the incidence of multiple falls was extremely high
in this population (five or more falls in 3 months) (Ballard et al. 1999). Nakamura
et al. assessed the influence of the severity of dementia on falls and found that the
number of fallers was higher in the intermediate stage of the disease compared
with the milder stage (Nakamura et al. 1996). Their findings, however, were not
confirmed by other studies (Asada et al. 1996; Buchner and Larson 1987). The
discrepancy in the findings can be explained by the use of different methods
assessing severity.
Motor impairments, particularly gait disturbances, are considered one of the
most consistent predictor of falls among older adults with cognitive decline
(Ganz et al. 2007). Impaired gait, reduced muscular strength, and impaired bal-
ance (Harlein et al. 2009) as well as impairments in activity level and mobility
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 21
MCI
Cognitive impairment Dementia
b
Slow gait velocity
Instability Falls – fractures
Fig. 2.1 Parallel decline in gait and cognitive function with aging. (a) Traditional view. Gait per-
formance and cognitive function deteriorate with aging, yielding two geriatric entities: falls and
dementia. (b) Alternative view. Cognition predicts mobility decline and falls, and mobility decline
and slow gait predict cognitive deterioration. These phenomena occur concurrently. MCI mild
cognitive impairment (Reprinted with permission from Journal of the American Geriatrics Society
(Montero-Odasso et al. 2012c))
measures (Suttanon et al. 2013) contribute to fall risk in this population. Other
measures of disability, such as peripheral neuropathy, musculoskeletal prob-
lems, and impaired tandem gait as a marker for ataxia, have also been shown to
increase the risk of falls in patients with Alzheimer’s disease (AD) (Buchner and
Larson 1987). In a prospective study on 135 Japanese older adults (Suzuki et al.
2012), Suzuki et al. proposed a model of 11 fall-related behaviors that may be
effective indicators to predict falls. The indicators consisted of intrinsic motor
and behavioral aspects, such as being agitated and wandering, inability to main-
tain seated balance in a wheelchair, and impairments in judgment, such as imme-
diate desire to urinate or defecate. Interestingly, according to this model,
behaviors such as delirium, awareness of self, symptoms of Parkinson’s disease,
and wandering were not related to falls (Suzuki et al. 2012). In addition to intrin-
sic factors, psychotropic medications were found to also increase the risk of falls
by causing sedation and postural hypotension (Ensrud et al. 2002; Husted et al.
2000; Thapa et al. 1995). These reports suggest that falls and risk of falls in older
adults and specifically those with cognitive deficits or dementia are multifacto-
rial in nature.
22 M. Dorfman et al.
Gait variability is a measure of gait consistency and reflects the stride-to-stride fluc-
tuations in walking. Gait variability offers a complementary way of quantifying
locomotion and its changes with aging and disease, and it is considered a reliable
measure of fall risk as well (Hausdorff 2005). The variability of several spatiotem-
poral gait parameters has been widely studied, among them stride time, stance time,
stride length, and step width (Brach et al. 2005, 2008; Richardson et al. 2004).
Stride time variability reflects one of the final pathways of the outcomes that the
central nervous system regulates (Hausdorff 2005). The general assumption is that
there is an inverse association between stride time variability and gait stability
(Montero-Odasso et al. 2012c). Gait variability tends to be higher in people with
AD compared to age-matched controls, and it has been associated with a higher risk
of falls among these individuals (Nakamura et al. 1996; Visser 1983). Cognitive
deterioration, especially EF, is associated with increased gait variability and thus
independently associated with future falls (Beauchet et al. 2008; Buracchio et al.
2011; Chen et al. 2012; Herman et al. 2010; Yogev-Seligmann et al. 2008).
Dual Task
resources between walking and cognitive tasks. Several studies showed that sub-
jects with dementia exhibited greater gait changes compared to normal age-matched
controls during the execution of dual tasks (Allali et al. 2008; Camicioli et al. 1997a,
b; Woollacott and Shumway-Cook 2002), particularly in stride time and stride-to-
stride variability. Interestingly, the magnitude of the effect of the secondary task on
gait stability is reportedly in direct relationship with the complexity of the given
dual task (Montero-Odasso et al. 2012b) and to the individual’s cognitive capacity,
with the greatest deterioration of gait performance observed in patients with cogni-
tive impairments (Muir et al. 2012). These findings may help to explain the greater
risk of falls and injuries among older adults with cognitive impairment or
dementia.
MCI is defined as a transition state between normal cognition and dementia in older
adults (Petersen 2007). It has been recently established that cognitive decline is
accompanied by early motoric decline, manifesting either as two separate biological
processes or as a cascading intertwined process (Aggarwal et al. 2006; Boyle et al.
2005, 2007; Kluger et al. 1999; Louis et al. 2005; Verghese et al. 2002, 2007).
Table 2.1 summarizes some of the key findings on gait changes in MCI.
Older people are often defined as having MCI based on impairments in memory
(amnestic) or non-memory (non-amnestic) domains (Moretti et al. 2013). Both
MCI subtypes have poorer performance on most gait variables, such as gait veloc-
ity, stride length, gait variability, cadence, swing time, and double support time,
than age-matched healthy controls (Verghese et al. 2008). Conversely, the two
MCI subtypes differ in certain gait domains. Subjects with amnestic MCI have
poorer swing time, stride time variability, and stride length variability than those
with non-amnestic MCI, suggesting that individuals with amnestic MCI tend to
have more variability and a decreased rhythmical pattern of gait and control than
individuals with non-amnestic MCI (Verghese et al. 2008). Serious gait alterations,
such as hemiparetic gait and frontal or parkinsonian gait, are also more frequent in
amnestic MCI than non-amnestic MCI (Verghese et al. 2008). MCI patients with
two or more vascular factors were shown to have greater frontal–subcortical dys-
function (Montero-Odasso et al. 2012a). An interaction was found between a high
vascular burden (two or more vascular factors) and the triad of executive dysfunc-
tion, gait disorders, and depressive symptoms among MCI patients (Montero-
Odasso et al. 2012a). A magnetic resonance spectroscopy and volumetric imaging
study exhibited an association between lower metabolite ratios and volume of the
primary motor cortex and poor gait performance in both single- and dual-task con-
ditions in mild cognitive impairment. These findings point to a possible involve-
ment of decreased neuronal function in the primary motor cortex causing gait
disorders (Annweiler et al. 2013), although other pathways are also likely to be
involved.
Table 2.1 Gait and falls in mild cognitive impairment (MCI)
24
Boyle et al. 598 participants cognitively intact (mean Clinical evaluations, including Parkinsonism signs, especially gait disturbance, bradykinesia,
(2005) age 79.6 ± 6.8 years) and 237 assessments of parkinsonian signs and rigidity, were more common in MCI subjects compared
participants with MCI (mean age and cognitive function to cognitively intact subjects
82.8 ± 6.9 years) Lower levels of cognitive function, particularly in perceptual
speed, were associated with higher levels of parkinsonism
among individuals with MCI
Participants with non-amnestic MCI showed more gait
disturbances compared to participants with amnestic MCI
Verghese et al. 295 participants cognitively intact (mean Clinical and quantitative gait Neurologically impaired gaits were more common in amnestic
(2008) age 79.3 ± 4.7 years), 54 participants performance MCI than in non-amnestic MCI and controls. Quantitative
with amnestic MCI (mean age gait in multiple parameters was worse in both MCI types
82.6 ± 5.7 years), and 62 participants than in controls. Factor analysis revealed three independent
with non-amnestic MCI (mean age factors representing pace, rhythm, and variability. Subjects
81.8 ± 6.2 years) with amnestic MCI had worse rhythm and variability scores
than those with non-amnestic MCI and controls. Subjects
with non-amnestic MCI had worse performance in the pace
domain than the other two groups. Subjects with MCI and
gait abnormalities had higher disability scores than subjects
with MCI without gait abnormalities
M. Dorfman et al.
2
Muir et al. 22 participants cognitively intact (mean Evaluation of gait performance while Gait velocity and stride time variability were not significantly
(2012) age 71.0 ± 5.0 years), 29 participants performing single and dual tasks under the single-task condition
with MCI (mean age 73.6 ± 6.2 years), In contrast, gait velocity decreased and strides time and stride
and 23 participants with AD (mean age time variability increased under the dual-task condition in
77.5 ± 5.0 years) people with MCI and AD
The greatest deterioration of gait performance occurred under
complex motor task tests
Montero- 35 participants with MCI (mean age Assessment to determine whether Participants with two or more vascular factors had greater
Odasso 75.5 ± 1.1 years) community-dwelling older adults frontal–subcortical dysfunction
et al. with MCI and a high vascular There was an interaction between a high vascular burden, two or
(2012a) burden were more likely to exhibit more vascular factors, and the three components of the triad
the frontal–subcortical triad of The number of vascular factors was directly associated with the
executive dysfunction, gait number of frontal–subcortical dysfunctions
disorders, and depressive
The vascular burden was associated with the number of frontal–
symptoms than those with a low
subcortical dysfunctions
vascular burden
Montero- 25 participants cognitively intact (mean Gait assessment under single (usual There was a significant difference within and between groups of
Odasso age 71.5 ± 4.1 years) and 53 partici- walking) and dual tasking increasing gait variability as dual-task complexity increased
et al. pants with MCI (mean age (naming animals and subtracting
(2012b, c) 75.1 ± 6.3 years) serial 7 s) Gait velocity decreased within groups as dual-task complexity
increased
Annweiler 20 participants with MCI (mean age Gait velocity and stride time The neurochemistry and volume of the primary motor cortex
Gait Disorders in Patients with Cognitive Impairment or Dementia
et al. 76 ± 11 years) variability while performing were associated with gait performance while carrying out
(2013) single and dual tasking single and dual tasking. Cortical volume correlated with
Ratios of N-acetylaspartate to creatine faster gait velocity during single (P = 0.029) and dual tasking
and choline to creatine and cortical (P = 0.037) and with decreased stride time variability during
volume were calculated in the single tasking
primary motor cortex
(continued)
25
Table 2.1 (continued)
26
A MEDLINE literature search was conducted using the terms “gait,” “gait disorder,” “gait disorders,” “falls,” “walking,” “Alzheimer’s disease,” “dementia,” or “mild
cognitive impairment” to identify potential papers. After review of the abstracts and cross-references, the relevant papers were studied and categorized into the entries
shown in the tables. A formal meta-analysis process was not employed
M. Dorfman et al.
AD Alzheimer-type dementia
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 27
Table 2.2 summarizes some of the key findings on gait changes in dementia. The
prevalence of vascular dementia and other non-AD is 30–50 % of all dementias in
older adults (Chui et al. 2000; Roman 2004; Verghese et al. 2002). Gait disorders
28 M. Dorfman et al.
of dementia
according to gait status at 0.6
enrollment. Dotted lines
represent 95 % confidence 0.4
intervals (From New England
Journal of Medicine, 0.2
Abnormal gait (n = 85)
Verghese et al. (2002).
Copyright © 2002 0.0
Massachusetts Medical 0 5 10 15 20
Society, Reprinted with Years of follow-up
permission from
Massachusetts Medical b
Society) 1.0 Normal gait (n = 337)
Proportion surviving free
of vascular dementia
0.8
0.6
0.2
0.0
0 5 10 15 20
Years of follow-up
c
1.0
Proportion surviving free
of alzheimer’s disease
0.6
0.2
0.0
0 5 10 15 20
Years of follow-up
occur late in the progression of Alzheimer’s disease, while they present earlier or
even precede vascular dementia (McKhann et al. 1984; Roman et al. 1993). Patients
with AD present with a decrease in gait velocity and stride length as well as an
increase in double support and stride length variability than healthy older adults
(Alexander et al. 1995; Nakamura et al. 1996; Tanaka et al. 1995; Visser 1983).
These changes could be considered as reflecting a more exaggerated impact of
“aging.” These gait features decline further as the disease continues to progress (van
Iersel et al. 2004). Furthermore, individuals with VaD exhibit even slower walking
2
Nakamura et al. Cross-sectional 45 AD patients and 15 control Gait and posture evaluation Mild-stage AD participants increased postural sway
(1997) subjects and examination of rCBF associated with a reduced mean value of rCBF
in different clinical stages in the cortex
Reduced mean rCBF values in the cortex and in the
frontal lobe of moderate-stage AD participants
were associated with increased postural sway
and stride length variability and with decreased
stride length
Reduced rCBF in the basal ganglia and in the
frontal lobe was also associated with increased
postural sway, double support time, and stride
length variability and with decreased walking
speed and stride length in severe-stage
participants
Van Iersel et al. Cross-sectional 63 participants with dementia Participants walked twice at After adjustment for parkinsonism and walking
(2006) (mean age 79.8 ± 7.4 years) preferred speed and twice aids, participants with dementia walked faster
and 62 participants without while counting than participants without dementia
dementia (mean age backwards (dual task)
74.7 ± 6.5 years)
AD Alzheimer-type dementia, VaD vascular dementia, rCBF regional cerebral blood flow
Gait Disorders in Patients with Cognitive Impairment or Dementia
31
32 M. Dorfman et al.
velocities compared to patients with AD (van Iersel et al. 2004). Another common
gait feature in people with dementia is a relatively shorter step length, which cor-
relates to decreased gait velocity and disease progression (Alexander et al. 1995;
Nakamura et al. 1997; Tanaka et al. 1995; Visser 1983). Patients with AD exhibit
significantly slower gait velocity and shorter step length compared to healthy con-
trols, and patients with VaD exhibit an even greater reduction in gait speed and
stride length relative to patients with AD (Tanaka et al. 1995). The short step length
and decreased velocity also contribute to an increase in double support time and
increase in stride length variability which, in turn, can increase unsteadiness and the
risk of falls (Alexander et al. 1995; van Iersel et al. 2004).
The authors of a prospective study of the relationship between falls and stride
length variability in 96 Japanese patients with AD observed a significant increase in
fall incidence among moderate-stage AD patients compared to mild-stage AD
patients, as well as a significant decrease in gait parameters, such as gait speed and
stride length (Nakamura et al. 1996). In addition, stride length variability was sig-
nificantly higher (worse) in patients with more severe AD than in patients with mild
AD. Furthermore, there was a significant difference only in stride length variability
when gait measures of fallers and non-fallers were compared according to the sever-
ity of dementia (Nakamura et al. 1996). The results of that study highlight the
important relationship between specific aspects of gait and falls in patients with
dementia. In contrast to those findings, however, only van Iersel et al.’s study dem-
onstrated that patients with dementia walked faster than patients without dementia,
and those authors considered that frontal lobe disinhibition and lack of insight might
be responsible for this phenomenon (van Iersel et al. 2006).
“Cautious gait” is characterized by a decrease in gait velocity, step length, and
static and dynamic balance, a widened base of support, hesitation and freezing, and
a reduction in postural responses (Scherder et al. 2007). Cautious gait is likely to
appear in the early stages of AD (O’Keeffe and Lavan 1996; Prehogan and Cohen
2004), while a “frontal gait disorder” is more common in more advanced stages of
AD and it is characterized by pseudoparkinsonian symptoms, such as shuffling, and
start and turn hesitation (O’Keeffe and Lavan 1996). The number of patients exhib-
iting parkinsonian symptoms increases during the course of evolving dementia.
These symptoms reflect extrapyramidal signs, such as bradykinesia, cogwheel
rigidity, rest tremor, and parkinsonian gait (Burns et al. 2005). Burns et al. suggest
that extrapyramidal signs are related to basal ganglia pathologies that may account
for the increasing prevalence of extrapyramidal signs as AD progresses (Burns et al.
2005).
Gait disorders in AD could be explained by a high burden of age-related subcor-
tical hyperintensities on the frontal–subcortical circuits together with hippocampal
degeneration (Annweiler et al. 2013). Increasing lines of evidence from clinical
practice, epidemiological studies, and neuroimaging studies show that the vascular
burden plays a key role in the onset and progression of AD (van Norden et al. 2012).
Therefore, it is not surprising that the vascular component of AD appears to be
involved in gait disorders among patients with AD. Gait velocity, stride length, and
step width are the gait parameters most commonly affected in the presence of WMH
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 33
(Annweiler et al. 2012). In their review, Annweiler et al. (2012) suggested that
quantitative parameters of gait, such as slower gait velocity and shorter stride length
in AD-related gait disorders, are associated with WMH specifically in the frontal
area and the basal ganglia, which are both part of the frontal–subcortical circuits.
These findings could also account for the correlation between the lower hippocam-
pal volume and function and qualitative gait disorders, such as higher stride length
variability. Interestingly, the nigrostriatal dopamine system was found to remain
unaffected. Zimmerman et al. reported that higher stride length variability in AD
was associated with lower metabolism in the hippocampal cortex (Zimmerman
et al. 2009) which has been suggested as the first cortical region damaged in AD
(van Norden et al. 2012).
For many years, dementia was not treated due to the notion that further deterioration
was unavoidable once it had been diagnosed. In contrast to normal aging, only a
limited number of studies have examined therapeutic methods to enhance gait and
motor performance in patients with dementia. These few studies generally target
cognition and behavior. It has been reported that improvement in cognition, particu-
larly EF, may influence gait (Tanaka et al. 1995; Yogev-Seligmann et al. 2008), but
only a few studies have directly assessed the effect of interventions on gait, balance,
or falls in dementia. Table 2.3 summarizes interventions aimed at enhancing motor
performance in dementia. These interventions can be divided into non-pharmacologic
and pharmacologic types. The former includes cognitive training (Schwenk et al.
2010) which is based on the notion that dual-task deficits have been linked to func-
tional decline and falls (Camicioli et al. 1997a; Yogev-Seligmann et al. 2008).
Studies among patients with dementia have shown that music therapy may improve
behavioral and psychological symptoms as well as cognitive functions and that it
may also have a potential positive cardiovascular effect (Raglio et al. 2012).
Unfortunately, only one study focused on the benefit of music therapy on motor
performances (Clair and O’Konski 2006). There is some evidence that physical
activity delays the onset of dementia in healthy older adults and slows down cogni-
tive decline. A physical exercise program appears to be a promising non-
pharmacologic strategy for slowing down cognitive decline (Balsamo et al. 2013).
In addition to having a positive influence on cognition, several studies demonstrated
that intensive dementia-specific motor training also increased the level of physical
activity in this population (Hageman and Thomas 2002; Hauer et al. 2012; Rolland
et al. 2007; Schwenk et al. 2010; Toulotte et al. 2003; Venturelli et al. 2011;
Zieschang et al. 2013), with evidence of retention of gains even at 9 months after the
end of training (Zieschang et al. 2013).
Pharmacologic interventions to improve cognition, particularly EF, have also
been shown to influence motor abilities and gait performance (Auriel et al. 2006;
Ben-Itzhak et al. 2008). Initial findings in older adults suggested that
Table 2.3 Interventions for improving gait in dementia
34
Duration of
Reference Study design Participants Type of intervention intervention Outcome measures Summary of findings
Non-pharmacologic treatment of gait disturbances in patients with dementia or cognitive decline
Hageman and Pre–posttest 26 elderly demented Moderate physical 2–3 times/week Comfortable and Improvement was observed on all
Thomas design patients (mean age activity, progressive for 6 weeks fast gait gait measures, but the only
(2002) 79.9 ± 5.5 years) resistance, and lower velocity over a significant change was in fast
extremity exercise 6-m distance gait time
using Thera-Band and the TUG
Toulotte et al. RCT 20 elderly demented The intervention group Once/week for Get up and go test, Gait speed, flexibility, and static
(2003) patients with a history received physical 45 min, sit on chair and balance improved. The
of falling (mean age activity training: 16 weeks reach test, 10-m intervention group did not fall
81.4 ± 4.7 years) strength exercise, walk test, and while the control subjects fell 6
proprioception, static posturography times during the 16 weeks. The
and dynamic balance, intervention group started to
and flexibility. No fall again after the end of
exercise for controls training
Clair et al. Pre–posttest 28 patients with advanced Comparison of gait 16 weeks Speed, cadence, No significant differences in gait
(2006) design dementia parameters in three and stride were observed, but caregiver
conditions of music length burden apparently benefited
therapy from the use of acoustic
stimuli
Rolland et al. RCT Intervention group of 67 The intervention group 1 h twice weekly Katz ADL score, Improvement in the intervention
(2007) patients with AD had a program of during ADL score group was observed on the
(mean age combined walking, 12 months measures of 6WTt, but there was no effect
82.8 ± 7.8 years) and exercise, muscle physical on cognitive responses
control group of 67 strength, balance, and performance,
patients with AD flexibility tasks. The nutritional
(mean age control group received status,
83.1 ± 7.0 years) routine medical care behavioral
disturbance, and
depression
M. Dorfman et al.
2
Schwenk et al. RCT Intervention group of 20 Intervention subjects 2 h twice weekly, Improvement in Specific training improved
(2010) patients with mild to received cognitive 12 weeks dual-task performance on a challenging
moderate dementia training of progres- performance dual-task condition (e.g.,
(mean age sively dual-task compared to serial three subtractions while
80.4 ± 7.1 years) and training. Controls baseline single walking), but not under less
control group of 29 received low-intensity tasks challenging dual-task
patients with mild to exercise conditions
moderate dementia
(mean age
82.3 ± 7.9 years)
Venturelli et al. RCT Intervention group of 11 The intervention group Minimum 30 A 6WT, the Barthel Improvement in the intervention
(2011) patients (mean age performed simple min of index of ADL, group was observed on the
83 ± 6 years) and aerobic activities. The moderate and MMSE 6WT and ADLs. Both groups
control group of 10 control group carried exercise, 4 had a decline in MMSE, but
patients (mean age out daily organized times a week the intervention group’s was
85 ± 5 years) with activities (e.g., bingo, during slower
advanced AD patchwork sewing, and 24 weeks
music therapy)
Hauer et al. RCT Intervention group of 62 Intervention subjects 3-month Strength, physical Intensive, dementia-adjusted
(2012) patients with mild to received progressive intervention function, and training was feasible and
moderate dementia resistance and and 3-month physical activity substantially improved motor
(mean age functional training. follow-up performance
82.3 ± 6.6 years) and Controls received a
Gait Disorders in Patients with Cognitive Impairment or Dementia
Assal et al. Pre–posttest Intervention group of Intervention group 24 weeks Stride time before Stride time was shorter under dual
(2008) design nine subjects with received galantamine and after task after treatment. There
mild to moderate AD treatment at a mean 6 months of was no change in controls
(mean age dose of 17.8 ± 3.5 mg/ galantamine
77.9 ± 2.1 years) and day according to treatment
18 controls without standard criteria during single
dementia (mean age and dual tasking
78.1 ± 1.0 years) while walking
Montero-Odasso Open-label 6 mild AD subjects 6 AD patients received 4 months Gait velocity and Gait speed and gait variability
et al. (2009b) design (mean age donepezil during gait variability improved under both single-
79.9 ± 4.0 years) and 1 month with 5 mg/ and dual-task walking in the
8 no-treatment MCI day of donepezil and intervention group compared
control subjects 3 months with 10 mg/ to control group. The
(mean age day of donepezil. 8 increases in gait speed were
75.6 ± 6.2 years) MCI patients received sustained and continued to
no treatment during improve after 4 months, while
4 months MCI patients had decreased
mean gait velocity and
increased gait variability
Beauchet et al. Pre–posttest Intervention group of Intervention group 211 ± 78.2 days Mean and CV of Stride CV improved in the
(2011) design 17 AD patients (mean received memantine stride time were intervention group. There was
age 83.8 ± 5.8 years) 20 mg once daily in determined no other significant difference
and 32 AD controls the morning (titrated before and after between groups
Gait Disorders in Patients with Cognitive Impairment or Dementia
methylphenidate (MPH) can enhance cognitive and motor function and that it may
have a role as a therapeutic option for reducing fall risk (Auriel et al. 2006; Ben-
Itzhak et al. 2008; Shorer et al. 2013). However, there is no study specific to people
with dementia. The use of MPH (i.e., Ritalin) is well known in treating children
with attention deficit hyperactivity disorder, but much less is known about its poten-
tial to improve motor function and gait among elderly people.
Acetylcholinesterase (AChE) inhibitors (e.g., donepezil, galantamine, and riv-
astigmine) are the most useful symptomatic treatment for AD and VaD (Seltzer
et al. 2004). It has been suggested that AChE inhibitors may improve gait perfor-
mance (Assal and van der Meulen 2009) by enhancing attentional resource alloca-
tion. The effects of donepezil and galantamine were evaluated in a few small studies
and noted that patients treated with donepezil and galantamine had better motor
performances (Assal et al. 2008; Drever et al. 2011; Litvinenko et al. 2008; Montero-
Odasso et al. 2009b). However, one should keep in mind that AChE inhibitors may
provoke parkinsonian symptoms in AD (Trabace et al. 2000), although Gurevich
et al. found that rivastigmine caused only slight worsening of tremor without delete-
riously affecting other PD symptoms while improving cognition among demented
PD patients (Gurevich et al. 2006). Memantine is another symptomatic treatment
for AD, and it reportedly has a beneficial effect on global cognitive function
(McShane et al. 2006). Memantine-related decrease in stride time variability among
people with AD has also been reported (Beauchet et al. 2011). It has been suggested
that the combination of memantine plus vitamin D may be more protective than
memantine alone against the neuronal loss and the subsequent declines in cognitive
and gait performance in AD (Annweiler et al. 2012; Annweiler and Beauchet 2011).
Overall, the findings of both the pharmacologic and non-pharmacologic studies for
improving gait and reducing falls in the presence of dementia need to be replicated
and investigated in large-scale, prospective, randomized, and controlled trials.
Cognitive alterations are the dominant symptom in dementia, but falls and gait distur-
bances are ubiquitous among affected individuals. A growing body of literature dem-
onstrates that gait abnormalities can also be present in the early stage of the disease,
even before dementia has been diagnosed. These gait abnormalities may serve as a
biomarker and assist in the prediction of dementia. The results of the studies men-
tioned in this review emphasize the importance of routinely including gait assess-
ment—specifically, gait speed and gait variability—in the examination of patients
with cognitive decline (Hausdorff and Buchman 2013). Questions about the underly-
ing mechanisms linking motor and cognitive function still remain unanswered. It is
hoped that improvement in our ability to quantify and understand microvascular
changes will lead to better understanding of the interactions between aging, gait alter-
ations, and cognitive impairment and provide better targeted treatment to combat the
functional deterioration in older adults with cognitive impairments and dementia.
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 39
References
Aggarwal NT, Wilson RS, Beck TL, et al. Motor dysfunction in mild cognitive impairment and the
risk of incident Alzheimer disease. Arch Neurol. 2006;63(12):1763–9.
Alexander NB. Gait disorders in older adults. J Am Geriatr Soc. 1996;44(4):434–51.
Alexander NB, Mollo JM, Giordani B, et al. Maintenance of balance, gait patterns, and obstacle
clearance in Alzheimer’s disease. Neurology. 1995;45(5):908–14.
Allali G, Assal F, Kressig RW, et al. Impact of impaired executive function on gait stability.
Dement Geriatr Cogn Disord. 2008;26(4):364–9.
Annweiler C, Beauchet O. Vitamin D-mentia: randomized clinical trials should be the next step.
Neuroepidemiology. 2011;37(3–4):249–58.
Annweiler C, Beauchet O, Celle S, et al. Contribution of brain imaging to the understanding of gait
disorders in Alzheimer’s disease: a systematic review. Am J Alzheimers Dis Other Demen.
2012;27(6):371–80.
Annweiler C, Beauchet O, Bartha R, et al. Motor cortex and gait in mild cognitive impairment: a
magnetic resonance spectroscopy and volumetric imaging study. Brain. 2013;136(Pt
3):859–71.
Asada T, Kariya T, Kinoshita T, et al. Predictors of fall-related injuries among community-dwelling
elderly people with dementia. Age Ageing. 1996;25(1):22–8.
Assal F, van der Meulen M. Pharmacological interventions in primary care: hopes and illusions.
Front Neurol Neurosci. 2009;24:54–65.
Assal F, Allali G, Kressig RW, et al. Galantamine improves gait performance in patients with
Alzheimer’s disease. J Am Geriatr Soc. 2008;56(5):946–7.
Atkinson HH, Rapp SR, Williamson JD, et al. The relationship between cognitive function and
physical performance in older women: results from the women’s health initiative memory
study. J Gerontol A Biol Sci Med Sci. 2010;65(3):300–6.
Auriel E, Hausdorff JM, Herman T, et al. Effects of methylphenidate on cognitive function and gait
in patients with Parkinson’s disease: a pilot study. Clin Neuropharmacol. 2006;29(1):15–7.
Ballard CG, Shaw F, Lowery K, et al. The prevalence, assessment and associations of falls in
dementia with Lewy bodies and Alzheimer’s disease. Dement Geriatr Cogn Disord.
1999;10(2):97–103.
Balsamo S, Willardson JM, Frederico SS, et al. Effectiveness of exercise on cognitive impairment
and Alzheimer’s disease. Int J Gen Med. 2013;6:387–91.
Beauchet O, Annweiler C, Allali G, et al. Recurrent falls and dual task-related decrease in walking
speed: is there a relationship? J Am Geriatr Soc. 2008;56(7):1265–9.
Beauchet O, Annweiler C, Dubost V, et al. Stops walking when talking: a predictor of falls in older
adults? Eur J Neurol. 2009;16(7):786–95.
Beauchet O, Allali G, Thiery S, et al. Association between high variability of gait speed and mild
cognitive impairment: a cross-sectional pilot study. J Am Geriatr Soc. 2011;59(10):1973–4.
Ben-Itzhak R, Giladi N, Gruendlinger L, et al. Can methylphenidate reduce fall risk in community-
living older adults? A double-blind, single-dose cross-over study. J Am Geriatr Soc.
2008;4(56):695–700.
Blake AJ, Morgan K, Bendall MJ, et al. Falls by elderly people at home: prevalence and associated
factors. Age Ageing. 1988;17(6):365–72.
Boyle PA, Wilson RS, Aggarwal NT, et al. Parkinsonian signs in subjects with mild cognitive
impairment. Neurology. 2005;65(12):1901–6.
Boyle PA, Wilson RS, Buchman AS, et al. Lower extremity motor function and disability in mild
cognitive impairment. Exp Aging Res. 2007;33(3):355–71.
Brach JS, Berlin JE, VanSwearingen JM, et al. Too much or too little step width variability is asso-
ciated with a fall history in older persons who walk at or near normal gait speed. J Neuroeng
Rehabil. 2005;2:21.
Brach JS, Studenski S, Perera S, et al. Stance time and step width variability have unique contribut-
ing impairments in older persons. Gait Posture. 2008;27(3):431–9.
40 M. Dorfman et al.
Buchner DM, Larson EB. Falls and fractures in patients with Alzheimer-type dementia. JAMA.
1987;257(11):1492–5.
Buracchio T, Dodge HH, Howieson D, et al. The trajectory of gait speed preceding mild cognitive
impairment. Arch Neurol. 2010;67(8):980–6.
Buracchio TJ, Mattek NC, Dodge HH, et al. Executive function predicts risk of falls in older adults
without balance impairment. BMC Geriatr. 2011;11:74.
Burns JM, Galvin JE, Roe CM, et al. The pathology of the substantia nigra in Alzheimer disease
with extrapyramidal signs. Neurology. 2005;64(8):1397–403.
Camicioli R, Howieson D, Lehman S, et al. Talking while walking: the effect of a dual task in
aging and Alzheimer’s disease. Neurology. 1997a;48(4):955–8.
Camicioli R, Panzer VP, Kaye J. Balance in the healthy elderly: posturography and clinical assess-
ment. Arch Neurol. 1997b;54(8):976–81.
Camicioli R, Oken BS, Sexton G, et al. Verbal fluency task affects gait in Parkinson’s disease with
motor freezing. J Geriatr Psychiatry Neurol. 1998;11(4):181–5.
Chen TY, Peronto CL, Edwards JD. Cognitive function as a prospective predictor of falls. J
Gerontol B Psychol Sci Soc Sci. 2012;67(6):720–8.
Chui HC, Mack W, Jackson JE, et al. Clinical criteria for the diagnosis of vascular dementia: a
multicenter study of comparability and interrater reliability. Arch Neurol. 2000;57(2):191–6.
Clair AA, O’Konski M. The effect of rhythmic auditory stimulation (RAS) on gait characteristics
of cadence, velocity, and stride length in persons with late stage dementia. J Music Ther.
2006;43(2):154–63.
de Carle AJ, Kohn R. Risk factors for falling in a psychogeriatric unit. Int J Geriatr Psychiatry.
2001;16(8):762–7.
Della SS, Spinnler H, Venneri A. Walking difficulties in patients with Alzheimer’s disease might
originate from gait apraxia. J Neurol Neurosurg Psychiatry. 2004;75(2):196–201.
Drever BD, Riedel G, Platt B. The cholinergic system and hippocampal plasticity. Behav Brain
Res. 2011;221(2):505–14.
Elble RJ, Moody C, Leffler K, et al. The initiation of normal walking. Mov Disord.
1994;9(2):139–46.
Ensrud KE, Blackwell TL, Mangione CM, et al. Central nervous system-active medications and
risk for falls in older women. J Am Geriatr Soc. 2002;50(10):1629–37.
Ganz DA, Bao Y, Shekelle PG, et al. Will my patient fall? JAMA. 2007;297(1):77–86.
Giladi N, Herman T, Reider-Groswasser II, et al. Clinical characteristics of elderly patients with a
cautious gait of unknown origin. J Neurol. 2005;252(3):300–6.
Gurevich TY, Shabtai H, Korczyn AD, et al. Effect of rivastigmine on tremor in patients with
Parkinson’s disease and dementia. Mov Disord. 2006;21(10):1663–6.
Hageman PA, Thomas VS. Gait performance in dementia: the effects of a 6-week resistance train-
ing program in an adult day-care setting. Int J Geriatr Psychiatry. 2002;17(4):329–34.
Harlein J, Dassen T, Halfens RJ, et al. Fall risk factors in older people with dementia or cognitive
impairment: a systematic review. J Adv Nurs. 2009;65(5):922–33.
Hauer K, Schwenk M, Zieschang T, et al. Physical training improves motor performance in people
with dementia: a randomized controlled trial. J Am Geriatr Soc. 2012;60(1):8–15.
Hausdorff JM. Gait variability: methods, modeling and meaning. J Neuroeng Rehabil. 2005;2:19.
Hausdorff JM. Gait dynamics, fractals and falls: finding meaning in the stride-to-stride fluctuations
of human walking. Hum Mov Sci. 2007;26(4):555–89.
Hausdorff JM, Buchman AS. What links gait speed and MCI with dementia? A fresh look at the
association between motor and cognitive function. J Gerontol A Biol Sci Med Sci.
2013;68(4):409–11.
Hausdorff JM, Rios DA, Edelberg HK. Gait variability and fall risk in community-living older
adults: a 1-year prospective study. Arch Phys Med Rehabil. 2001;82(8):1050–6.
Hausdorff JM, Yogev G, Springer S, et al. Walking is more like catching than tapping: gait in the
elderly as a complex cognitive task. Exp Rain Res. 2005;164(4):541–8.
Hausdorff JM, Schweiger A, Herman T, et al. Dual-task decrements in gait: contributing factors
among healthy older adults. J Gerontol A Biol Sci Med Sci. 2008;63(12):1335–43.
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 41
Herman T, Mirelman A, Giladi N, et al. Executive control deficits as a prodrome to falls in healthy
older adults: a prospective study linking thinking, walking, and falling. J Gerontol A Biol Sci
Med Sci. 2010;65(10):1086–92.
Heuninckx S, Wenderoth N, Debaere F, et al. Neural basis of aging: the penetration of cognition
into action control. J Neurosci. 2005;25(29):6787–96.
Holtzer R, Verghese J, Xue X, et al. Cognitive processes related to gait velocity: results from the
Einstein Aging Study. Neuropsychology. 2006;20(2):215–23.
Husted JA, Cook RJ, Farewell VT, et al. Methods for assessing responsiveness: a critical review
and recommendations. J Clin Epidemiol. 2000;53(5):459–68.
Inzitari M, Baldereschi M, Di CA, et al. Impaired attention predicts motor performance decline in
older community-dwellers with normal baseline mobility: results from the Italian Longitudinal
Study on Aging (ILSA). J Gerontol A Biol Sci Med Sci. 2007;62(8):837–43.
Kallin K, Gustafson Y, Sandman PO, et al. Drugs and falls in older people in geriatric care settings.
Aging Clin Exp Res. 2004;16(4):270–6.
Kluger A, Ferris SH, Golomb J, et al. Neuropsychological prediction of decline to dementia in
nondemented elderly. J Geriatr Psychiatry Neurol. 1999;12(4):168–79.
Litvinenko IV, Odinak MM, Mogil’naia VI, et al. Memantine (akatinol) therapy of cognitive
impairment in Parkinson’s disease complicated by dementia. Zh Nevrol Psikhiatr Im S S
Korsakova. 2008;108(10):37–42.
Louis ED, Schupf N, Manly J, et al. Association between mild parkinsonian signs and mild cogni-
tive impairment in a community. Neurology. 2005;64(7):1157–61.
Maki BE. Gait changes in older adults: predictors of falls or indicators of fear. J Am Geriatr Soc.
1997;45(3):313–20.
McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer’s disease: report of
the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human
Services Task Force on Alzheimer’s Disease. Neurology. 1984;34(7):939–44.
McShane, R, Areosa, SA, Minakaran, N. Memantine for dementia. Cochrane Database Syst Rev.
2006;2:CD003154.
Montero-Odasso M, Bergman H, Phillips NA, et al. Dual-tasking and gait in people with mild
cognitive impairment. The effect of working memory. BMC Geriatr. 2009a;9:41.
Montero-Odasso M, Wells J, Borrie M. Can cognitive enhancers reduce the risk of falls in people
with dementia? An open-label study with controls. J Am Geriatr Soc. 2009b;57(2):359–60.
Montero-Odasso M, Annweiler C, Hachinski V, et al. Vascular burden predicts gait, mood, and
executive function disturbances in older adults with mild cognitive impairment: results from
the gait and brain study. J Am Geriatr Soc. 2012a;60(10):1988–90.
Montero-Odasso M, Muir SW, Speechley M. Dual-task complexity affects gait in people with mild
cognitive impairment: the interplay between gait variability, dual tasking, and risk of falls.
Arch Phys Med Rehabil. 2012b;93(2):293–9.
Montero-Odasso M, Verghese J, Beauchet O, et al. Gait and cognition: a complementary approach
to understanding brain function and the risk of falling. J Am Geriatr Soc. 2012c;60(11):
2127–36.
Moretti F, De RD, Palmer K, et al. Prevalence and characteristics of mild cognitive impairment in
the general population. Data from an Italian population-based study: The Faenza Project.
Aging Ment Health. 2013;17(3):267–75.
Morris JC, Rubin EH, Morris EJ, et al. Senile dementia of the Alzheimer’s type: an important risk
factor for serious falls. J Gerontol. 1987;42(4):412–7.
Muir SW, Speechley M, Wells J, et al. Gait assessment in mild cognitive impairment and
Alzheimer’s disease: the effect of dual-task challenges across the cognitive spectrum. Gait
Posture. 2012;35(1):96–100.
Nakamura T, Meguro K, Sasaki H. Relationship between falls and stride length variability in senile
dementia of the Alzheimer type. Gerontology. 1996;42(2):108–13.
Nakamura T, Meguro K, Yamazaki H, et al. Postural and gait disturbance correlated with decreased
frontal cerebral blood flow in Alzheimer disease. Alzheimer Dis Assoc Disord. 1997;
11(3):132–9.
42 M. Dorfman et al.
Nevitt MC, Cummings SR. Type of fall and risk of hip and wrist fractures: the study of osteopo-
rotic fractures. The Study of Osteoporotic Fractures Research Group. J Am Geriatr Soc.
1993;41(11):1226–34.
Njegovan V, Hing MM, Mitchell SL, et al. The hierarchy of functional loss associated with cogni-
tive decline in older persons. J Gerontol A Biol Sci Med Sci. 2001;56(10):M638–43.
O’Keeffe ST, Lavan JN. Predicting delirium in elderly patients: development and validation of a
risk-stratification model. Age Ageing. 1996;25(4):317–21.
Odenheimer G, Funkenstein HH, Beckett L, et al. Comparison of neurologic changes in ‘success-
fully aging’ persons vs the total aging population. Arch Neurol. 1994;51(6):573–80.
Ostchega Y, Harris TB, Hirsch R, et al. The prevalence of functional limitations and disability in
older persons in the US: data from the National Health and Nutrition Examination Survey III.
J Am Geriatr Soc. 2000;48(9):1132–5.
Perry RJ, Hodges JR. Attention and executive deficits in Alzheimer’s disease. A critical review.
Brain. 1999;122(Pt 3):383–404.
Petersen RC. Mild cognitive impairment: current research and clinical implications. Semin Neurol.
2007;27(1):22–31.
Prehogan A, Cohen CI. Motor dysfunction in dementias. Geriatrics. 2004;59(11):53–60.
Raglio A, Bellelli G, Mazzola P, et al. Music, music therapy and dementia: a review of literature
and the recommendations of the Italian Psychogeriatric Association. Maturitas.
2012;72(4):305–10.
Richardson JK, Thies SB, DeMott TK, et al. Interventions improve gait regularity in patients with
peripheral neuropathy while walking on an irregular surface under low light. J Am Geriatr Soc.
2004;52(4):510–5.
Rolland Y, Pillard F, Klapouszczak A, et al. Exercise program for nursing home residents with
Alzheimer’s disease: a 1-year randomized, controlled trial. J Am Geriatr Soc.
2007;55(2):158–65.
Roman GC. Vascular dementia. Advances in nosology, diagnosis, treatment and prevention.
Panminerva Med. 2004;46(4):207–15.
Roman GC, Tatemichi TK, Erkinjuntti T, et al. Vascular dementia: diagnostic criteria for research
studies. Report of the NINDS-AIREN International Workshop. Neurology. 1993;43(2):250–60.
Scherder E, Eggermont L, Swaab D, et al. Gait in ageing and associated dementias, its relationship
with cognition. Neurosci Biobehav Rev. 2007;31(4):485–97.
Schwenk M, Zieschang T, Oster P, et al. Dual-task performances can be improved in patients with
dementia: a randomized controlled trial. Neurology. 2010;74(24):1961–8.
Segev-Jacubovski O, Herman T, Yogev-Seligmann G, et al. The interplay between gait, falls and
cognition: can cognitive therapy reduce fall risk? Expert Rev Neurother. 2011;11(7):1057–75.
Seltzer B, Zolnouni P, Nunez M, et al. Efficacy of donepezil in early-stage Alzheimer disease: a
randomized placebo-controlled trial. Arch Neurol. 2004;61(12):1852–6.
Shaw FE. Prevention of falls in older people with dementia. J Neural Transm.
2007;114(10):1259–64.
Sheridan PL, Hausdorff JM. The role of higher-level cognitive function in gait: executive dysfunc-
tion contributes to fall risk in Alzheimer’s disease. Dement Geriatr Cogn Disord.
2007;2(24):125–37.
Sheridan PL, Solomont J, Kowall N, et al. Influence of executive function on locomotor function:
divided attention increases gait variability in Alzheimer’s disease. J Am Geriatr Soc.
2003;51(11):1633–7.
Shorer Z, Yaakov B, Guy T, et al. Effect of single dose methylphenidate on walking and postural
stability under single- and dual-task conditions in older adults–a double-blind randomized con-
trol trial. J Gerontol A Biol Sci Med Sci. 2013;68(10):1271–80.
Soumare A, Tavernier B, Alperovitch A, et al. A cross-sectional and longitudinal study of the
relationship between walking speed and cognitive function in community-dwelling elderly
people. J Gerontol A Biol Sci Med Sci. 2009;64(10):1058–65.
Springer S, Giladi N, Peretz C, et al. Dual-tasking effects on gait variability: the role of aging, falls,
and executive function. Mov Disord. 2006;21(7):950–7.
2 Gait Disorders in Patients with Cognitive Impairment or Dementia 43
Suttanon P, Hill KD, Said CM, et al. A longitudinal study of change in falls risk and balance and
mobility in healthy older people and people with Alzheimer disease. Am J Phys Med Rehabil.
2013;92(8):676–85.
Suzuki M, Kurata S, Yamamoto E, et al. Impact of fall-related behaviors as risk factors for falls
among the elderly patients with dementia in a geriatric facility in Japan. Am J Alzheimers Dis
Other Demen. 2012;27(6):439–46.
Tabbarah M, Crimmins EM, Seeman TE. The relationship between cognitive and physical perfor-
mance: MacArthur Studies of Successful Aging. J Gerontol A Biol Sci Med Sci.
2002;57(4):M228–35.
Tanaka A, Okuzumi H, Kobayashi I, et al. Gait disturbance of patients with vascular and Alzheimer-
type dementias. Percept Mot Skills. 1995;80(3 Pt 1):735–8.
Taniguchi Y, Yoshida H, Fujiwara Y, et al. A prospective study of gait performance and subsequent
cognitive decline in a general population of older Japanese. J Gerontol A Biol Sci Med Sci.
2012;67(7):796–803.
Thapa PB, Gideon P, Fought RL, et al. Psychotropic drugs and risk of recurrent falls in ambulatory
nursing home residents. Am J Epidemiol. 1995;142(2):202–11.
Tinetti ME. Prevention of falls and fall injuries in elderly persons: a research agenda. Prev Med.
1994;23(5):756–62.
Tinetti ME, Speechley M, Ginter SF. Risk factors for falls among elderly persons living in the com-
munity. N Engl J Med. 1988;26(319):1701–7.
Toulotte C, Fabre C, Dangremont B, et al. Effects of physical training on the physical capacity of
frail, demented patients with a history of falling: a randomised controlled trial. Age Ageing.
2003;32(1):67–73.
Trabace L, Cassano T, Steardo L, et al. Biochemical and neurobehavioral profile of CHF2819, a
novel, orally active acetylcholinesterase inhibitor for Alzheimer’s disease. J Pharmacol Exp
Ther. 2000;294(1):187–94.
van Dijk PT, Meulenberg OG, van de Sande HJ, et al. Falls in dementia patients. Gerontologist.
1993;33(2):200–4.
van Doorn C, Gruber-Baldini AL, Zimmerman S, et al. Dementia as a risk factor for falls and fall
injuries among nursing home residents. J Am Geriatr Soc. 2003;51(9):1213–8.
van Iersel MB, Hoefsloot W, Munneke M, et al. Systematic review of quantitative clinical gait
analysis in patients with dementia. Z Gerontol Geriatr. 2004;37(1):27–32.
van Iersel MB, Verbeek AL, Bloem BR, et al. Frail elderly patients with dementia go too fast. J
Neurol Neurosurg Psychiatry. 2006;77(7):874–6.
van Norden AG, van Dijk EJ, de Laat KF, et al. Dementia: Alzheimer pathology and vascular fac-
tors: from mutually exclusive to interaction. Biochim Biophys Acta. 2012;1822(3):
340–9.
Venturelli M, Scarsini R, Schena F. Six-month walking program changes cognitive and ADL per-
formance in patients with Alzheimer. Am J Alzheimers Dis Other Demen. 2011;26(5):381–8.
Verghese J, Xue X. Predisability gait patterns in older adults. Gait Posture. 2011;33(1):98–101.
Verghese J, Lipton RB, Hall CB, et al. Abnormality of gait as a predictor of non-Alzheimer’s
dementia. N Engl J Med. 2002;347(22):1761–8.
Verghese J, Kuslansky G, Holtzer R, et al. Walking while talking: effect of task prioritization in the
elderly. Arch Phys Med Rehabil. 2007;88(1):50–3.
Verghese J, Robbins M, Holtzer R, et al. Gait dysfunction in mild cognitive impairment syn-
dromes. J Am Geriatr Soc. 2008;56(7):1244–51.
Verghese J, Wang C, Lipton RB, et al. Motoric cognitive risk syndrome and the risk of dementia.
J Gerontol A Biol Sci Med Sci. 2013;68(4):412–8.
Visser H. Gait balance in senile dementia of Alzheimer’s type. Age Ageing.
1983;12(4):296–301.
Waite LM, Grayson DA, Piguet O, et al. Gait slowing as a predictor of incident dementia: 6-year
longitudinal data from the Sydney Older Persons Study. J Neurol Sci. 2005;229–230:89–93.
Watson GS, Leverenz JB. Profile of cognitive impairment in Parkinson’s disease. Brain Pathol.
2010;20(3):640–5.
44 M. Dorfman et al.
Woollacott M, Shumway-Cook A. Attention the control of posture and gait: a review of an emerg-
ing area of research. Gait Posture. 2002;16(1):1–14.
Yogev-Seligmann G, Hausdorff JM, Giladi N. The role of executive function and attention in gait.
Mov Disord. 2008;23(3):329–42.
Zieschang T, Schwenk M, Oster P, et al. Sustainability of motor training effects in older people
with dementia. J Alzheimers Dis. 2013;34(1):191–202.
Zimmerman ME, Pan JW, Hetherington HP, et al. Hippocampal correlates of pain in healthy
elderly adults: a pilot study. Neurology. 2009;73(19):1567–70.
Chapter 3
Falls in Patients with Dementia
Abstract Falls are a major problem in dementia with an annual reported incidence
of about 70–80 %, double that of older adults without cognitive impairment.
Remaining mobile and free from falls is a priority, and understanding falls in
dementia is of critical importance to mitigate the burden caused not only to the
patient but also the carer. Despite this, there is currently no convincing evidence to
mitigate falls risk in dementia. Gait and balance impairments are important falls risk
factors as are cognitive impairment, and more recently the interdependency between
these features has been recognized and is influencing assessment and the develop-
ment of novel therapeutic strategies. This chapter covers four relevant and important
key areas: (1) prevalence, incidence, and risk of falls with respect to different types
of dementia including a focus on disorders in gait and balance which are important
contributors and medication; (2) emerging evidence for contributory mechanisms;
(3) evolving concepts relating to the interrelationship between gait, balance, cogni-
tion, and falls; and finally (4) approaches to mitigate falls risk and implications for
clinical practice.
Introduction
Falls are the fifth leading cause of death among older adults (World Health
Organisation 2013) and as such constitute a major health concern. Approximately
one third of people over the age of 65 fall each year, with an increase in prevalence
3 Falls in Patients with Dementia 47
Table 3.1 Annual prevalence and incidence of falls (falls/1,000 persons) and prevalence of gait
and balance disorders in different dementia subtypes
Controls AD VAD DLB PDD PD
Falls prevalence (%) 36 47 47 77 90 61
Falls incidence 1,023 2,486 3,135 9,087 19,000 4,617
(falls/1,000
people)
Gait disorder N (%) 3/42 (7) 10/40 (25) 31/39 (79) 24/32 (75) 43/46 (93) 20/46 (43)
Taken from Allan et al. (2005, 2009)
in dementia. Broadly speaking, people with dementia are two to three times more
likely to fall than age-matched healthy counterparts, with institutionalized adults
also at a higher risk compared with community living (Kröpelin et al. 2013).
Although estimates vary widely according to the method of data collection, type of
dementia, and domestic environment, figures suggest a prevalence of around 40 %
for community-dwelling adults (Allan et al. 2009; Salva et al. 2012) and 60 % for
people living in residential care (Eriksson et al. 2008). Several reports indicate a
higher prevalence in people with dementia associated with Parkinson’s disease (PD)
compared with dementia related to vascular disease – differences that are also
reflected in incidence figures (Table 3.1). Annual incidence figures are consistent for
prospective studies, with estimates as noted above between 70 and 85 %, twice that
of healthy older adults (Shaw 2002). Incidence figures for retrospective studies are
less consistent and produce much lower estimates (Shaw 2002). The prevalence and
incidence of falls is set to increase given the aging population, which is estimated to
triple from 600 million to 2 billion by 2050 (World Health Organisation 2009),
a third of who will end their lives with dementia (Department of Health 2009).
Compared with demented older adults who do not fall, fallers have a higher risk
of functional decline and are five times more likely to be institutionalized (Salva
et al. 2012). Falls in dementia are also a common cause of hospitalization. In a lon-
gitudinal study of 682 people with AD, falls resulted in admission in just over a fifth
of the 26.2 % hospitalized (Voisin et al. 2010). People with dementia constitute a
significant proportion of the total population of elderly hip fracture patients in hos-
pitals (about 30–50 %) (Stenvall et al. 2012). Other specific issues include increase
risk of delirium, poor recovery, increased institutionalization, and increased mortal-
ity (Scandol et al. 2013).
An extensive body of research has identified risk factors for falls in people with
cognitive impairment and dementia. The evidence is difficult to summarize because
of inconsistencies in the classification and severity of cognitive impairment and the
environment in which people are tested. Overall, the key factors include type of
dementia, gait and postural impairment, medication, neurocardiovascular instabil-
ity, and environment (Kröpelin et al. 2013; Shaw 2007; Harlein et al. 2009). Below
we focus on three of the major risk factors for falls in patients with dementia –
dementia subtype, gait and balance impairments, and medication.
(a) Type of Dementia. Dementia itself is a recognized risk factor for falls with
recent studies reporting the annual prevalence of falls in people with dementia
living in the community range from 35 to 90 % (Allan et al. 2009; Shaw 2002;
Salva et al. 2012) and those in residential care between 40 and 62 % (Eriksson
48 L. Rochester et al.
Key
1.0
control
AD
VAD
0.8 DLB
PDD
Cumulative survival
0.6
0.4
0.2
0.0
Fig. 3.1 Survival curve showing time to first fall by diagnosis (Reprinted from Allan et al. (2009))
et al. 2008). Cohorts are typically heterogeneous in nature and include demen-
tias of mixed etiology and a broad range of severity. Understanding falls risk
with respect to dementia subtype is highly relevant to a better understanding of
the underlying mechanisms and to identify and target key at-risk groups for
intervention. Allan et al. (2005) reported retrospective falls in dementia sub-
types (AD, VAD, DLB, and PDD) and found that although people with demen-
tia were more likely to report single or multiple falls in the previous year, the
risk of falls was highly dependent upon subtype (see Table 3.1). A follow-on
prospective study confirmed these findings and described the annual incidence
of falls according to dementia and dementia subtype (Allan et al. 2009). Fall
prevalence and rates with respect to dementia subtype are shown in Table 3.1,
while Fig. 3.1 shows the time to first fall. It is clear that Lewy body dementias
(DLB and PDD) have a far higher risk for falls than AD and VAD, a feature that
also informs the diagnostic criteria for DLB (McKeith et al. 2000). It is also
pertinent that fall-related injuries were greater in Lewy body dementias, while
Lewy body disease and parkinsonism were identified as risk factors for falls in
patients with dementia (Allan et al. 2009; Lowery et al. 2000b).
(b) Gait and Balance Impairment. Gait and balance disturbances are more common
in older adults with a 35 % prevalence of gait disorders in older adults over the
age of 70 (Verghese et al. 2006) and are important risk factors for falls (Ambrose
et al. 2013). Classification schemes identify specific syndromes of gait such as
hypokinetic-rigid gait, cautious gait, and higher-level gait disorders (Snijders
et al. 2007) that include gait deficits such as slow, cautious gait; freezing of gait;
start hesitation; balance impairment; and associated cognitive impairments.
3 Falls in Patients with Dementia 49
These features are also associated with increased falls risk, for example, parkin-
sonism (slow gait with freezing) (Lim et al. 2008; Kerr et al. 2010; Latt et al.
2009) as well as mild cognitive impairment (Taylor et al. 2012; Camicioli and
Majumdar 2010). The prevalence of gait and balance impairments is even
greater in dementia (Ambrose et al. 2013; Taylor et al. 2012; van Iersel et al.
2004) accompanying the increased falls risk. The prevalence and severity of
gait disorders are also dependent upon the dementia subtype (Allan et al. 2005)
(see Table 3.1). The highest level of gait and balance impairments was also
similar in pattern to falls rates with respect to dementia subtype being greatest
in LBD, especially PDD, and the lowest in AD, although all groups had a higher
prevalence of gait and balance impairment than controls. It is also worth noting,
however, that the high incidence of falls in DLB could not be attributed purely
to movement disorders such as parkinsonism as falls risk was found to be
increased even in those without extrapyramidal signs (Imamura et al. 2000).
What is evident is the added burden apparently placed by gait and balance dis-
orders on falls risk in dementia, highlighting the complex relationship between
gait, balance, cognition, and falls and will be discussed in section “The Complex
Relationship Between Gait, Balance, Cognition, and Falls: Emerging Concepts.”
(c) Medication. As discussed earlier, advanced age is the greatest risk factor for
increased dementia risk. As age increases, so too does the number of medica-
tions prescribed, with polypharmacy itself a risk factor for falls in older adults
(Neutel et al. 2002). In addition, patients with cognitive impairment and associ-
ated behavioral symptoms may be treated with psychotropic medications, which
have long been associated with increased falls risk (Ensrud et al. 2002; Fick
et al. 2007; Leipzig et al. 1999; Sheahan et al. 1995). Mechanisms through
which falls occur in association with psychotropic medications may include
fatigue/somnolence, decreased awareness of surroundings, confusion, ortho-
static hypotension, or syncope. Benzodiazepines in particular, used to treat sleep
disturbance in older adults and REM sleep behavior disorder in DLB and PDD,
increase the risk of fall and subsequent hip fracture by at least 50 % (Cumming
and Le Couteur 2003). More recently it has been recognized that antidepres-
sants are associated with falls, with selective serotonin reuptake inhibitors asso-
ciated with the highest adjusted hazard ratios for falls (1.66) (Coupland et al.
2011). Specific medications that may increase falls risk in patients with PDD
include dopaminergic agents, which may precipitate falls through dyskinesias
(Robinson et al. 2005). Therefore, it is prudent to consider rationalization of
medications that may not be required or which may increase falls risk.
Cholinergic Dysfunction
In PD and PDD, falls are frequent and are closely related to cognition especially
attention (Latt et al. 2009; Allcock et al. 2009; Burn et al. 2003; Burn et al. 2006),
leading to an assumption that they may have a similar neurochemical mechanism,
and therefore cognitive disturbance and falling may not be independent. Two major
cholinergic projection systems in the brain could contribute and are shown in
Fig. 3.2a. The pedunculopontine nucleus (PPN) in the brainstem contains choliner-
gic neurons that have a powerful influence on gait and postural control (Jenkinson
et al. 2009; Karachi et al. 2010). Thalamic acetylcholinesterase (AChE) activity
reflects cholinergic activity from neurons in the PPN (Bohnen and Albin 2011),
which is reduced in early PD (Bohnen and Albin 2011; Gilman et al. 2010; Shimada
et al. 2009) and more so in PD fallers (Bohnen et al. 2009a), suggesting a relation-
ship with gait and postural dyscontrol. Cholinergic function (estimated using short-
latency afferent inhibition) was also found to be highly associated to gait in PD (see
Fig. 3.2b) even after adjusting for age and motor dysfunction (Rochester et al.
2012). Cholinergic neurons in the nucleus basalis of Meynert (nbM) play a role in
attentional control and executive function (Yarnall et al. 2011) which also contribute
to gait disturbance in PD (Lord et al. 2010, 2011a, b; Rochester et al. 2008; Yogev-
Seligmann et al. 2005).
Leukoaraiosis or lesions of the white matter (WML) are seen as areas of increased
signal intensity on brain scans and are common findings in older adults who are
otherwise fit and well (LADIS study group et al. 2011). WML are associated with
small vessel cerebrovascular disease (CVD) as a result of hypertension (an addi-
tional risk factor for falls in dementia) causing structural lesions in white matter
tracts which can lead to changes in motor and cognitive function (LADIS study
group et al. 2011). Structural changes can also occur in normal-appearing white
matter and lead to motor and cognitive impairments (LADIS study group et al.
2011). In older adults without cognitive impairment, WML are associated with gait
and balance disorders (Zheng et al. 2011; de Laat et al. 2011; Baezner et al. 2008;
Rosano et al. 2006), in particular reduced speed and increased variability (Srikanth
et al. 2009). They are also associated prospectively with increased falls risk (Srikanth
et al. 2009; Blahak et al. 2008). Lesions in frontal-subcortical motor circuits were
significantly associated with balance disturbances and increased falls risk (Blahak
et al. 2008). In adults with high-level gait disorders (HLGD), a gait syndrome
reflecting a more cautious hesitant gait, associated changes in white matter were
found with diffusor tension imaging in pathways related to both motor and cognitive
function (Kafri et al. 2013). The roles of WML and CVD are also increasingly rec-
ognized as contributory features to the pathophysiology of different dementia
3 Falls in Patients with Dementia 51
a Striatum
Thalamus
nbM
PPN
1.8 1.8
b Control PD
r = –.040, p = .860 r = –.606, p = .003
1.6 1.6
Walking speed
1.4 1.4
(m.s–1)
1.2 1.2
1.0 1.0
.8 .8
Fig. 3.2 (a) Schematic representation of cholinergic output in the cortex (Reprinted from Yarnall
et al. (2011) with permission from John Wiley and Sons). Cholinergic interneurons in the striatum
are shown in blue. The pedunculopontine nucleus (PPN; shown in red) provides the majority of
cholinergic input to the thalamus, with other projections to the nucleus basalis of Meynert (nbM),
striatum, substantia nigra, subthalamic nucleus, globus pallidus interna, cerebellum, and spinal
cord. The nbM (shown in green) sends cholinergic projections to the cerebral cortex and also to
thalamic nuclei. (b) Correlation of walking speed and short-latency afferent inhibition (SAI) in PD
(n = 22) and control participants (n = 22) showing a significant association for PD only (Reprinted
from Rochester et al. (2012), by permission of Oxford University Press)
subtypes leading to speculation that CVD and WML burden might be contributory
pathological features in gait, balance, and cognitive impairment and therefore a
common mechanism of falls in dementia. For example, AD is linked to CVD with
evidence to suggest that CVD may accelerate the pathology and symptoms
52 L. Rochester et al.
Until relatively recently, falls and dementia were treated as two relatively unrelated
entities (Fig. 3.2a), with distinct pathways and causal features. This view is chang-
ing. A 2010 addition to American and British Geriatric Societies (American
Geriatric Society/British Geriatric Society 2011) clinical practice guidelines for
prevention of falls suggests that further to questioning patients about a fall, clini-
cians should also ask about the presence of any gait problems. This move reflects
the wider, recent recognition of the complex interrelationship between gait, cogni-
tion, and falls, as depicted in Fig. 3.2b.
For the past decade, the link between cognition, gait, and falls has intensified.
The notion that safe and effective gait (and by corollary, absence of falls) is due
solely to an intact motor system has given way to a more complex model that
reflects the cognitive control of balance and gait (Montero-Odasso et al. 2012).
This “top-down” control is evident well before dementia states emerge. The cogni-
tive “cost” of balance and walking is revealed when the motor system is stressed
during assessment. The most common approach to producing “stress” is to use a
dual-task testing paradigm, when the subject performs a simultaneous (usually cog-
nitive) task during a balance task or during gait. For any adult, dual tasking is atten-
tion demanding but the effect is marked in people with cognitive impairment and
dementia. In a seminal study in 1997, Lundin-Olsson (Lundin-Olsson et al. 1997)
reported increased falls in institutionalized adults with dementia who were unable
to continue walking when a conversation was initiated. This gave rise to the “stops
walking when talking” test as a predictive measure of falls. Attention is a powerful
modifier of gait and falls and may “drive” other cognitive features also involved.
Allcock and colleagues (2009) demonstrated that power of attention and reaction
time variability scores were significantly associated with falls, even when adjusted
for motor severity. However, the relationship between dual task and falls is not
3 Falls in Patients with Dementia 53
clear cut, given the wide range of response to dual task and protocols used in test-
ing. The link between cognition, balance, and gait has focused attention on gait
measurement and what it can reveal about future cognitive (and health) states. In
longitudinal cohorts of community-dwelling older adults, gait impairment has been
shown to predict mild cognitive decline and dementia (Verghese et al. 2007), along
with falls (Ambrose et al. 2013) and adverse health outcomes (Abellan van Kan
2009).
The recent emphasis on cognitive control of gait and falls does not preclude the
important contribution of sensorimotor function to balance, gait, and falls which is
well established in the literature for non-demented fallers. For example, muscle
strength and vision have been shown to be key determinants of balance, gait, and
falls, and a comprehensive falls assessment includes these in a wider battery along
with proprioceptive testing (Lord et al. 2003). The relative contribution of cognitive
function to motor function is likely to be different in dementia, with cognitive func-
tion overriding other systems. This has not been studied in detail. However, knowl-
edge of the role of cognition in pre-dementia states provides a basis for novel
interventions, which are discussed below.
As stated at the beginning of the chapter, the most recent clinical practice guideline
for prevention of falls in older persons (American Geriatric Society/British
Geriatric Society 2011) concluded that “There is insufficient evidence to recom-
mend for or against multifactorial or single interventions to prevent falls in older
persons with known dementia living in the community or in long-term care facili-
ties,” due in large part to lack of studies and poor quality of studies. Widely accepted
falls reduction programs for older adults (e.g., FAME, Otago Falls Programme, Tai
Chi) emerge from research that has excluded people with dementia or cognitive
impairment (Gillespie et al. 2009). Exercise is feasible for people with dementia,
but has focused on cardiovascular fitness rather than exercise to improve balance
(Hill et al. 2009). Multicomponent interventions that target endurance, strength,
and balance have been shown to improve physical functioning (Blankevoort et al.
2010), but the translation to falls is less convincing. Individualized, tailor-made
programs may be more effective than generic interventions. A recent RCT reported
beneficial outcomes for people with dementia who were exposed to an individual-
ized, multidisciplinary medical and rehabilitation regime after hip fracture (Stenvall
et al. 2012), and a recent pilot study reported a 32.6 % reduction in falls and
decreased agitation in people with dementia living in a residential care home
through the use of individualized therapies based on behavioral and cognitive pro-
files (Bharwani et al. 2012). Current evidence points to the need for targeted selec-
tion of participants, with consideration given to severity and type of dementia,
presence of gait abnormalities and functional limitations, previous history of falls,
54 L. Rochester et al.
living environment, and age. The most critical of these is likely to be severity of
dementia: above a certain threshold interventions will be ineffective (Shaw 2002).
Physical activity is overall protective of cognitive decline and protective for first
falls in dementia (Allan et al. 2009), but this may depend on the setting. Sherrington
(Sherrington et al. 2008) reported that exercise alone had no benefit in reducing
falls in people with dementia living in care homes, possibly because the exercises
were mostly in sitting which does not sufficiently challenge postural mechanisms.
Over time, interest in physical activity diminishes and mobility becomes more
fragmented and related to less purposeful actions such as wandering during delir-
ium. This trajectory of change needs to be considered when designing falls inter-
vention program.
These studies do not tackle the complex interaction between cognitive impair-
ment and gait and balance impairment which are common and related risk factors
for falls in dementia as depicted in Fig. 3.3. Taking this contemporary view, new
approaches are emerging that aim to address the role of cognition in control of gait
and postural control using either pharmacological or non-pharmacological thera-
pies (see Table 3.2 for examples) (see Montero-Odasso et al., for review (Montero-
Odasso et al. 2012)) and are briefly considered below. This type of approach is
typically aimed far earlier in the disease process targeting those with mild dementia
or even earlier with mild cognitive impairment.
a
Slow gait velocity
Instability Falls – fractures
MCI
Cognitive Impairment Dementia
b
Slow gait velocity
Instability Falls – fractures
Fig. 3.3 Traditional and contemporary views of the decline in cognition and mobility and their
relationship with falls. (a) Traditional view. (b) Alternative, emerging view (Reprinted from
Montero-Odasso et al. (2012) with permission from John Wiley and Sons)
3 Falls in Patients with Dementia 55
Table 3.2 Falls interventions in dementia: tradition and contemporary approaches with selected
examples
Traditional Contemporary
Single domain intervention approach Pharmacological approaches targeting cognitive
(e.g., exercise) (Sherrington et al. impairment – cholinergic dysfunction (Montero-
2008) Odasso et al. 2012; Chung et al. 2010)
Multifactorial intervention based on Exercise to reduce CVD as a potential risk factor
generic risk factors (Stenvall et al.
2012)
Multifactorial intervention based on Tailor-made intervention, based on individual profile of,
risk factors specific to participants e.g., falls history, behavior, and cognitive impair-
(e.g., residential setting, severity of ment (Bharwani et al. 2012)
dementia) (Oliver et al. 2007) Cognitive remediation using complex cognitive and
motor skill training typically involving dual-task
scenarios and decision making (Montero-Odasso
et al. 2012; Schwenk et al. 2010)
Pharmacological Approaches
Non-pharmacological Approaches
Cognitive remediation therapies are also receiving interest and have been shown to
improve executive function and attention in older adults with some emerging evi-
dence in people with MCI and dementia (Montero-Odasso et al. 2012). These types
of approaches involve complex cognitive and motor skill training typically involv-
ing dual-task scenarios and decision making. A recent study in people with demen-
tia combining gait and cognitive exercise training (dual-task training) saw improved
gait performance compared to simple exercises (Schwenk et al. 2010). Activity has
been recognized as being protective against falls in dementia (Allan et al. 2009) and
may be effective to target the risk factors for CVD and WML, providing a further
potential therapeutic target for falls interventions (Srikanth et al. 2009).
56 L. Rochester et al.
Additional Considerations
Any fall, especially in older, frailer adults, is associated with risk of fracture.
Fragility fractures are a major cause of morbidity and mortality, with 1-year mortal-
ity following fractured neck of femur up to 30 % (Wiles et al. 2011). Dementia is an
independent risk factor for fracture in PD (Melton et al. 2006), where the risk of
osteoporosis is increased due to reduced bone mineral density, vitamin D insuffi-
ciency, immobility, and reduced body mass index (Dobson et al. 2013; Ishizaki et al.
1993; Sato et al. 1997). In extrapyramidal disorders, hip fractures are more common
than upper limb fractures, probably due to the mechanism of falling, with some
evidence that this may also be true in AD (Williams et al. 2006). Therefore, bone
health should be assessed in these patients, using tools such as the WHO FRAX tool
(Kanis et al. 2008) or QFracture algorithm (Hippisley-Cox and Coupland 2012),
with subsequent referral for dual energy X-ray absorptiometry (DXA) where appro-
priate. Investigation for secondary causes of osteoporosis (such as vitamin D levels
and thyroid function tests) and attention to lifestyle factors (such as diet and smok-
ing) should be initiated where necessary. The treatment of osteoporosis should fol-
low locally agreed guidelines, with consideration to reduced mean life expectancy
in older people with more advanced dementia: here quality of life and reduction in
morbidity are important considerations. Lastly, occupational therapy home assess-
ment and visual assessment and treatment may also reduce the rate of falling in
community-dwelling older adults (Gillespie et al. 2009), although again the evi-
dence in those with additional cognitive impairment is lacking.
Ultimately, earlier intervention may however be more appropriate when potential
for compensation is greater. Targeting falls risk in mild cognitive impairment and
treating this as a prodromal stage for dementia may ultimately yield the greatest
benefits. Taking a combined approach to target multiple underlying mechanisms and
behavioral remediation of gait and cognitive impairment may represent potentially
exciting future developments to ameliorate falls risk in people with dementia.
Clinical Implications
• Gait and balance are useful biomarkers for falls risk in dementia and should be
included in assessment.
• Currently, there is no clear evidence for effective interventions.
• Emerging approaches to mitigate risk should be considered.
• Early intervention may be more beneficial using MCI as a prodromal state for
dementia when there is greater potential for compensation.
• Rationalization of medications, in particular with respect to centrally acting
medications, should be considered at an early stage to limit potential iatrogenic
harm.
3 Falls in Patients with Dementia 57
References
Abellan van Kan G. Gait speed at usual pace as a predictor of adverse outcomes in community-
dwelling older people. J Nutr Health Aging. 2009;13(10):881–9.
Allan LM, et al. Prevalence and severity of gait disorders in Alzheimer’s and non-Alzheimer’s
dementias. J Am Geriatr Soc. 2005;53(10):1681–7.
Allan LM, et al. Incidence and prediction of falls in dementia: a prospective study in older people.
PLoS One. 2009;4(5):e5521.
Allcock LM, et al. Impaired attention predicts falling in Parkinson’s disease. Parkinsonism Relat
Disord. 2009;15(2):110–5.
Ambrose AF, Paul G, Hausdorff JM. Risk factors for falls among older adults: a review of the lit-
erature. Maturitas. 2013;75(1):51–61.
American Geriatric Society/British Geriatric Society. Summary of the updated American Geriatrics
Society/British Geriatrics Society clinical practice guideline for prevention of falls in older
persons. J Am Geriatr Soc. 2011;591(1):148–57.
Annweiler C, et al. Contribution of brain imaging to the understanding of gait disorders in
Alzheimer’s disease: a systematic review. Am J Alzheimers Dis Other Demen.
2012;27(6):371–80.
Assal F, et al. Galantamine improves gait performance in patients with Alzheimer’s disease. J Am
Geriatr Soc. 2008;56(5):946–7.
Baezner H, et al. Association of gait and balance disorders with age-related white matter changes:
the LADIS study. Neurology. 2008;70(12):935–42.
Bharwani G, et al. Individualized behavior management program for Alzheimer’s/dementia resi-
dents using behavior-based ergonomic therapies. Am J Alzheimers Dis Other Demen.
2012;27(3):188–95.
Blahak C, et al. Association of gait and balance disorders with age-related white mater changes:
the LADIS study. Neurology. 2008;70:935–42.
Blankevoort CG, et al. Review of effects of physical activity on strength, balance, mobility and
ADL performance in elderly subjects with dementia. Dement Geriatr Cogn Disord.
2010;30(5):392–402.
Bohnen NI, Albin RL. The cholinergic system and Parkinson disease. Behav Brain Res.
2011;221(2):564–73.
Bohnen NI, et al. History of falls in Parkinson disease is associated with reduced cholinergic activ-
ity. Neurology. 2009a;73(20):1670–6.
Bohnen NI, et al. Age-associated leukoaraiosis and cortical cholinergic deafferentation. Neurology.
2009b;72(16):1411–6.
Bohnen NI, et al. Leucoaraiosis, nigrostriatal denervation and motor symptoms in Parkinson’s
disease. Brain. 2011;134(Pt 8):2358–65.
Burn DJ, et al. Extrapyramidal features in Parkinson’s disease with and without dementia and
dementia with Lewy bodies: a cross-sectional comparative study. Mov Disord.
2003;18(8):884–9.
Burn DJ, et al. Motor subtype and cognitive decline in Parkinson’s disease, Parkinson’s disease
with dementia, and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry.
2006;77(5):585–9.
Camicioli R, Majumdar SR. Relationship between mild cognitive impairment and falls in older
people with and without Parkinson’s disease: 1-year Prospective Cohort Study. Gait Posture.
2010;32(1):87–91.
Chung KA, et al. Effects of a central cholinesterase inhibitor on reducing falls in Parkinson dis-
ease. Neurology. 2010;75(14):1263–9.
Coupland C, et al. Antidepressant use and risk of adverse outcomes in older people: population
based cohort study. BMJ. 2011;343:d4551–66.
58 L. Rochester et al.
Cumming RG, Le Couteur DG. Benzodiazepines and risk of hip fractures in older people –
a review of the evidence. CNS Drugs. 2003;17(11):825–37.
de Laat KF, et al. Loss of white matter integrity is associated with gait disorders in cerebral small
vessel disease. Brain. 2011;134(Pt 1):73–83.
Department of Health. Improving care for people with dementia. 2013. https://www.gov.uk/gov-
ernment/policies/improving-care-for-people-with-dementia.
Dobson R, et al. Bone health in chronic neurological diseases: a focus on multiple sclerosis and
Parkinsonian syndromes. Pract Neurol. 2013;13(2):70–9.
Ensrud KE, et al. Central nervous system-active medications and risk for falls in older women.
J Am Geriatr Soc. 2002;50(10):1629–37.
Eriksson S, Gustafson Y, Lundin-Olsson L. Risk factors for falls in people with and without a
diagnosis of dementia living in residential care facilities: a prospective study. Arch Gerontol
Geriatr. 2008;46(3):293–306.
Ferri C, et al. Global prevalence of dementia: a Delphi consensus study. Lancet. 2005;366:2112–7.
Fick D, Kolanowski A, Waller J. High prevalence of central nervous system medications in
community-dwelling older adults with dementia over a three-year period. Aging Ment Health.
2007;11(5):588–95.
Gillespie LD, et al. Interventions for preventing falls in older people living in the community.
Cochrane Database Syst Rev. 2009;2, CD007146.
Gilman S, et al. Cerebral cortical and subcortical cholinergic deficits in Parkinsonian syndromes.
Neurology. 2010;74(18):1416–23.
Harlein J, et al. Fall risk factors in older people with dementia or cognitive impairment: a system-
atic review. J Adv Nurs. 2009;65(5):922–33.
Hill KD, et al. Effectiveness of balance training exercise in people with mild to moderate severity
Alzheimer’s disease: protocol for a randomised trial. BMC Geriatr. 2009;9:29.
Hippisley-Cox J, Coupland C. Derivation and validation of updated QFracture algorithm to predict
risk of osteoporotic fracture in primary care in the United Kingdom: prospective open cohort
study. BMJ. 2012;344:e3427–43.
Honjo K, Black SE, Verhoeff NP. Alzheimer’s disease, cerebrovascular disease, and the beta-
amyloid cascade. Can J Neurol Sci. 2012;39(6):712–28.
Imamura T, et al. Fall-related injuries in dementia with Lewy bodies (DLB) and Alzheimer’s dis-
ease. Eur J Neurol. 2000;7(1):77–9.
Inzitari M, et al. Cerebrovascular disease and gait and balance impairment in mild to moderate
Alzheimer’s disease. J Nutr Health Aging. 2013;17(1):45–8.
Ishizaki F, et al. Relationship between osteopenia and clinical characteristics of Parkinson’s dis-
ease. Mov Disord. 1993;8(4):507–11.
Jenkinson N, et al. Anatomy, physiology, and pathophysiology of the pedunculopontine nucleus.
Mov Disord. 2009;24(3):319–28.
Kafri M, et al. High-level gait disorder: associations with specific white matter changes observed
on advanced diffusion imaging. J Neuroimaging. 2013;23(1):39–46.
Kallin K, Gustafson Y, Lundin-Olsson L. Factors associated with falls among older, cognitively
impaired people in geriatric care settings. Am J Geriatr Psychiatry. 2005;13:501–9.
Kanis JA, et al. FRAX and the assessment of fracture probability in men and women from the UK.
Osteoporos Int. 2008;19(4):385–97.
Karachi C, et al. Cholinergic mesencephalic neurons are involved in gait and postural disorders in
Parkinson disease. J Clin Invest. 2010;120(8):2745–54.
Kerr G, et al. Predictors of future falls in Parkinson disease. Neurology. 2010;75(2):116–24.
Kröpelin TF, et al. Fall determinants in older long-term care residents with dementia: a systematic
review. Int Psychogeriatr. 2013;25(04):549–63.
LADIS study group et al. 2001-2011: A decade of the LADIS (Leukoaraiosis and DISability)
study: what have we learned about white matter changes and small-vessel disease? Cerebrovasc
Dis. 2011;32(6):577–88.
Latt M, et al. Clinical and physiological assessments for elucidating falls risk in Parkinson’s dis-
ease. Mov Disord. 2009;24(9):1280–9.
3 Falls in Patients with Dementia 59
Leipzig RM, Cumming RG, Tinetti ME. Drugs and falls in older people: a systematic review and
meta-analysis: II. Cardiac and analgesic drugs. J Am Geriatr Soc. 1999;47(1):40–50.
Lim I, et al. Identifying fallers with Parkinson’s disease using home-based tests: who is at risk?
Mov Disord. 2008;23(16):2411–5.
Lord S, Menz H, Tiedemann A. A physiological profile approach to falls risk assessment and pre-
vention. Phys Ther. 2003;83:237–52.
Lord S, et al. Executive dysfunction and attention contribute to gait interference in ‘off’ state
Parkinson’s disease. Gait Posture. 2010;31(2):169–74.
Lord S, et al. Gait variability in Parkinson’s disease: an indicator of non-dopaminergic contributors
to gait dysfunction? J Neurol. 2011a;258(4):566–72.
Lord S, et al. Attention rather than spatial memory predicts distinct characteristics of Parkinson’s
disease. Mov Disord. 2011b;26(Suppl2):S392–424.
Lowery K, et al. Depression in the carers of dementia sufferers: a comparison of the carers of
patients suffering from dementia with Lewy bodies and the carers of patients with Alzheimer’s
disease. J Affect Disord. 2000a;59(1):61–5.
Lowery K, Buri H, Ballard C. What is the prevalence of environmental hazards in the homes of
dementia sufferers and are they associated with falls. Int J Geriatr Psychiatry.
2000b;15(10):883–6.
Lundin-Olsson L, Nyberg L, Gustafson Y. “Stops walking when talking” as a predictor of falls in
elderly people. Lancet. 1997;349(9052):617.
McKeith I, et al. Prospective validation of consensus criteria for the diagnosis of dementia with
Lewy bodies. Neurology. 2000;54(5):1050–8.
Melton 3rd LJ, et al. Fracture risk after the diagnosis of Parkinson’s disease: influence of concomi-
tant dementia. Mov Disord. 2006;21(9):1361–7.
Montero-Odasso M, Wells J, Borrie M. Can cognitive enhancers reduce the risk of falls in peo-
ple with dementia? An open-label study with controls. J Am Geriatr Soc. 2009;57(2):
359–60.
Montero-Odasso M, et al. Gait and cognition: a complementary approach to understanding brain
function and the risk of falling. J Am Geriatr Soc. 2012;60(11):2127–36.
Morris J, et al. Senile dementia of the Alzheimer’s type: an important risk factor for serious falls.
J Gerontol. 1987;42:412–7.
Morrison R, Siu A. Mortality from pneumonia and hip fractures in patients with advanced demen-
tia. JAMA. 2000;284(19):2447–8.
Neutel CI, Perry S, Maxwell C. Medication use and risk of falls. Pharmacoepidemiol Drug Saf.
2002;11(2):97–104.
Oliver D, et al. Strategies to prevent falls and fractures in hospitals and care homes and effect of
cognitive impairment: systematic review and meta-analyses. BMJ. 2007;334(7584):82.
Robinson K, et al. Falling risk factors in Parkinson’s disease. NeuroRehabilitation. 2005;20(3):
169–82.
Rochester L, et al. Walking speed during single and dual tasks in Parkinson’s disease: which char-
acteristics are important? Mov Disord. 2008;23(16):2312–8.
Rochester L, et al. Cholinergic dysfunction contributes to gait disturbance in early Parkinson’s
disease. Brain. 2012;135(Pt 9):2779–88.
Rosano C, et al. Quantitative measures of gait characteristics indicate prevalence of underlying
subclinical structural brain abnormalities in high-functioning older adults. Neuroepidemiology.
2006;26(1):52–60.
Salva A, et al. Falls and risk factors for falls in community-dwelling adults with dementia (NutriAlz
trial). Alzheimer Dis Assoc Disord. 2012;26(1):74–80.
Sato Y, Kikuyama M, Oizumi K. High prevalence of vitamin D deficiency and reduced bone mass
in Parkinson’s disease. Neurology. 1997;49(5):1273–8.
Scandol J, Toson B, Close J. Fall-related hip fracture hospitalisations and the prevalence of demen-
tia within older people in New South Wales, Australia: an analysis of linked data. Injury.
2013;44(6):776–83.
60 L. Rochester et al.
Schwenk M, et al. Dual-task performances can be improved in patients with dementia: a random-
ized controlled trial. Neurology. 2010;74(24):1961–8.
Shaw FE. Falls in cognitive impairment and dementia. Clin Geriatr Med. 2002;18(2):159–73.
Shaw FE. Prevention of falls in older people with dementia. J Neural Transm. 2007;
114(10):1259–64.
Sheahan SL, et al. Psychoactive medication, alcohol-use, and falls among older adults. J Behav
Med. 1995;18(2):127–40.
Sherrington C, et al. Effective exercise for the prevention of falls: a systematic review and meta-
analysis. J Am Geriatr Soc. 2008;56(12):2234–43.
Shimada H, et al. Mapping of brain acetylcholinesterase alterations in Lewy body disease by PET.
Neurology. 2009;73(4):273–8.
Snijders AH, et al. Neurological gait disorders in elderly people: clinical approach and classifica-
tion. Lancet Neurol. 2007;6(1):63–74.
Srikanth V, et al. Cerebral white matter lesions, gait, and the risk of incident falls: a prospective
population-based study. Stroke. 2009;40(1):175–80.
Stenvall M, et al. A multidisciplinary intervention program improved the outcome after hip frac-
ture for people with dementia–subgroup analyses of a randomized controlled trial. Arch
Gerontol Geriatr. 2012;54(3):e284–9.
Taylor ME, et al. Gait impairment and falls in cognitively impaired older adults: an explanatory
model of sensorimotor and neuropsychological mediators. Age Ageing. 2012;41(5):665–9.
Tinetti M, Speechley M, Ginter S. Risk factors for falls among elderly people living in the com-
munity. N Engl J Med. 1988;319:1701–7.
van Iersel MB, et al. Systematic review of quantitative clinical gait analysis in patients with
dementia. Z Gerontol Geriatr. 2004;37(1):27–32.
Verghese J, et al. Epidemiology of gait disorders in community-residing older adults. J Am Geriatr
Soc. 2006;54(2):255–61.
Verghese J, et al. Quantitative gait dysfunction and risk of cognitive decline and dementia. J Neurol
Neurosurg Psychiatry. 2007;78(9):929–35.
Voisin T, et al. Predictive factors of hospitalizations in Alzheimer’s disease: a two-year prospective
study in 686 patients of the REAL.FR study. J Nutr Health Aging. 2010;14(4):288–91.
Wiles MD, et al. Nottingham Hip Fracture Score as a predictor of one year mortality in patients
undergoing surgical repair of fractured neck of femur. Br J Anaesth. 2011;106(4):501–4.
Williams DR, Watt HC, Lees AJ. Predictors of falls and fractures in bradykinetic rigid syndromes:
a retrospective study. J Neurol Neurosurg Psychiatry. 2006;77(4):468–73.
World Health Organisation. Violence and injury prevention and disability-falls. 2013. http://www.
who.int/violence_injury_prevention_other_injury/falls/en/.
World Health Organisation. Global Health and Ageing. 2011. http://who.int/ageing/publications/
global_health/en/.
Yarnall A, Rochester L, Burn DJ. The interplay of cholinergic function, attention, and falls in
Parkinson’s disease. Mov Disord. 2011;26(14):2496–503.
Yogev-Seligmann G, et al. Dual tasking, gait rhythmicity, and Parkinson’s disease: which aspects
are attention demanding. Eur J Neurosci. 2005;22:124–1256.
Zheng JJ, et al. Impact of white matter lesions on physical functioning and fall risk in older people:
a systematic review. Stroke. 2011;42(7):2086–90.
Chapter 4
Treatment of Parkinsonism in Patients
with Non-Parkinson Dementia
Introduction
R. Mehanna, MD
Department of Neurology, University of Texas Health Science Center in Houston,
Houston, TX, USA
H.H. Fernandez, MD (*)
Department of Neurology, Cleveland Clinic Lerner College of Medicine,
Cleveland, OH, USA
e-mail: [email protected]
The treatment of parkinsonian in patients with DLB follows the same treatment
principles of idiopathic PD, where dopamine replacement remains the cornerstone
of treatment. There are, however, some differences (Poewe 2005). The main con-
cern is that pro-dopaminergic agents used to treat parkinsonism more readily exac-
erbate or cause hallucinations in DLB (McKeith et al. 2003; Frank 2003; Zupancic
et al. 2011) as compared to idiopathic PD.
Because of their greater hallucination-inducing potential, dopamine agonists are
generally not considered first-line agents in DLB and in fact should be avoided
(Rascol et al. 2000; Parkinson Study Group 2000; Drach 2011).
4 Treatment of Parkinsonism in Patients with Non-Parkinson Dementia 63
The efficacy and safety of levodopa in DLB has been assessed only in small
series of up to 20 patients (Molloy et al. 2005; Bonelli et al. 2004; Goldman et al.
2008). Clinically significant improvement of parkinsonism after an oral dose of
levodopa has been reported in up to 50 % of patients with DLB (Bonelli et al.
2004), but this series did not report adverse events. However, in a series of 19
patients with DLB, Goldman et al. (2008) reported that only 22 % had improve-
ment in their motor symptoms with levodopa without worsening of their halluci-
nations. In a more recent series comparing the effect of levodopa on 24 DLB and
21 PD patients, Lucetti et al. (2010) reported a positive response to an acute oral
load of 250 mg of levodopa in 55 % of the DLB patients and 90 % of the PD
patients. A positive response was defined as an improvement of at least 15 % in the
tapping test and at least 25 % in the walking test and rigidity score or tremor score.
Patients were then treated with up to 600 mg per day of levodopa and reassessed
at 6 and 12 months. At both follow-up visits, DLB patients who responded to the
initial acute levodopa dose showed a greater motor benefit compared to DLB who
did not satisfactorily respond to the acute levodopa dose challenge. However,
when compared to PD acute levodopa responders, the benefit observed in DLB
acute levodopa responders was comparable at 6 months but was less at 12 months,
implying a more rapid reduction of levodopa efficacy in DLB. In this series, 2
DLB (8.3 %) patients were excluded because of exacerbation of hallucination on
levodopa and not included in the analysis. Moreover, 30 % of the DLB patients
were on clozapine and 50 % were on acetylcholinesterase inhibitors, which could
have prevented or masked potential worsening of hallucination from levodopa.
Overall, parkinsonism tends to respond less well to levodopa in DLB than in PD
64 R. Mehanna and H.H. Fernandez
or PD with dementia (PDD) (McKeith et al. 1996, 2005; Molloy et al. 2005;
Bonelli et al. 2004; Goldman et al. 2008; Drach 2011). Levodopa-induced dyski-
nesia were reported less frequently in DLB than in PD patients in one retrospec-
tive study of 25 DLB and 64 PD patients (Papapetropoulos et al. 2006), but this
could not be confirmed in prospective series (Lucetti et al. 2010). Should this be
true, it would be another indicator of decreased sensibility of DLB patient to dopa-
mine replacement therapy. In a cross-sectional study comparing the motor pheno-
type in 43 PDD and 26 DLB cases to that of 38 patients with PD and no dementia
(Burn et al. 2003), the postural instability/gait difficulty-phenotype was signifi-
cantly overrepresented in DLB compared to the uncomplicated PD group (69 %
versus 38 % of cases).
In the less frequent cases where tremor is the dominant symptom of parkinson-
ism, anticholinergic drugs should still be avoided as they can worsen the cognitive
deficit and/or induce delirium (Drach 2011). Amantadine 150 mg/day was reported
to partially improve parkinsonism in one patient with DLB (Sato et al. 2010), but it
can also worsen psychosis (Drach 2011) and is usually not recommended (Possin
and Kaufer 2010).
Although not routinely used in clinical practice, a daily dose of 25–100 mg of
zonisamide in addition to levodopa was shown to decrease off time without worsen-
ing dyskinesia in a randomized, double-blinded, controlled trial on 347 PD patients
(Murata et al. 2007). While no such trial exists for DLB patients, some cases have
been reported. Sato et al. (2010) reported motor improvement in one DLB patient
after 4 weeks of a daily dose of 25 mg of zonisamide, and with marked gait improve-
ment. The dose was then increased progressively to 75 mg daily with additional
improvement of aggression, apathy, and irritability. No side effects were reported.
The patient was also on a stable dose of amantadine 150 mg per day and donepezil
5 mg per day. Odawara et al. (2010) reported three additional DLB patients in whom
zonisamide was added to levodopa for motor symptom control. Two patients had
mild to moderate motor improvement at 25 mg/day, with dizziness at 50 mg/day.
The third patient had no motor improvement but experienced drowsiness at 100 mg
per day. All side effects improved after decreasing the dose. Interestingly, the first
two patients had previously responded to levodopa while the third one did not. The
effects of zonisamide on parkinsonism can be mediated by an increase in dopamine
synthesis at doses between 25 and 100 mg per day (Murata et al. 2007) as well as an
increase in extracellular levels of dopamine and serotonin (Farooq et al. 2008;
Murata et al. 2007).
In summary, and in an attempt to optimize motor control without worsening
cognitive or psychotic symptoms, low doses of levodopa should be used, possibly in
combination with zonisamide. Dopamine agonists, anticholinergics, and amanta-
dine should be avoided, as well as selegiline (Possin and Kaufer 2010), and perhaps
other monoamine oxidase B (MAO-B) inhibitors. Data on catechol-O-
methyltransferase (COMT) inhibitors in the DLB population are wanting, although
presumably they are less likely, but far from exempt, in precipitating hallucinations
among demented parkinsonian patients.
4 Treatment of Parkinsonism in Patients with Non-Parkinson Dementia 65
Although the response to levodopa may be poor or transient (van Balken and Litvan
2006), and parkinsonism in PSP seem to be less responsive to levodopa than other
Parkinson-plus syndromes (Birdi et al. 2002), patients with PSP more often still
receive a trial of levodopa/carbidopa. At least two studies involving up to 170 PSP
patients have demonstrated an improvement of motor symptoms with levodopa in
38–54 % of patients (Nieforth and Golbe 1993; Golbe et al. 1990). The amplitude
and duration of this response were however not specified (Burn and Warren 2005).
Another retrospective study of 12 PSP patients showed a more modest response but
reported significant adverse effects such as worsening of parkinsonism and postural
hypotension in more than half the patients (Kompoliti et al. 1998a). It should be
noted that the minimum clinically beneficial levodopa dose may be higher in PSP
than that in PD (Lubarsky and Juncos 2008) and may require up to 1 g of levodopa
per day. However, the response remains modest and is often short lived (3–5 years).
Over time, dopaminergic therapy can reversibly worsen most motor and behavioral
symptoms of PSP, and the difficult decision to stop these medications may need to
be taken. However, since this drug-induced worsening can be dose dependent, a
gradual de-escalation of dopaminergic drugs will give the opportunity to look for a
lower dose with a more favorable risk–benefit ratio (Lubarsky and Juncos 2008).
Moreover, abrupt discontinuation of dopaminergic drugs should be avoided to pre-
vent a withdrawal-induced neuroleptic malignant-like syndrome (Serrano-Duenas
2003; Yoshikawa et al. 1997). Finally, PSP patients are less likely to develop severe
motor fluctuations, dyskinesias, or dopaminergic-induced visual hallucinations than
PD patients (Aarsland et al. 2001), suggestive of a more dopamine-resistant
pathology.
In patients who do not respond to levodopa, dopamine receptor agonists such
as pramipexole or ropinirole may be considered. Indeed, unlike levodopa, these
medications act directly at the postsynaptic terminal in the striatum, bypassing the
substantia nigra that is commonly affected with severe neuronal loss in PSP
(Rabinovici and Miller 2010). If used as a first-line treatment, the response to
these agents is often comparable or inferior to that of levodopa (Burn and Warren
2005).
Amantadine has been reported to transiently improve parkinsonism in 15 % of
PSP patients (Irene and Yves 1992).
Finally, based on decreased frontal cortical GABA receptors on brain imaging of
PSP patients, a crossover trial of the GABA agonist zolpidem on 10 PSP patients
was conducted (Daniele et al. 1999). A single 5 mg zolpidem dose was found to
improve the motor subscale of the United Parkinson’s Disease Rating Scale by more
than 20 %, while a dose of 10 mg of zolpidem or 250 mg of levodopa failed to exert
such a response. The main side effect of this drug was mild drowsiness in 50 % of
the patients and moderate drowsiness in an additional 10 %. These results have not
been duplicated.
66 R. Mehanna and H.H. Fernandez
CBS is notoriously resistant to levodopa, even more so than PSP. However, although
the response to levodopa may be poor or transient, patients often receive a trial of
levodopa. In a retrospective review of 147 patients with CBS, Kompoliti et al.
(1998b) reported carbidopa/levodopa use in 87 % of the patients with a median
daily dose of 300 mg (range 100–2,000 mg) but with improvement in only 26 % of
the patients exposed to the drug. Twenty-five percent were treated with pergolide or
bromocriptine, of whom only 6 % improved. Selegiline was used in 20 % of the
patients and produced motor benefits in only 10 % of patients. Amantadine was
prescribed in 16 % and improved bradykinesia, rigidity, and gait in 13 % of patients.
Anticholinergic agents were used in 27 % of the patients and improved parkinson-
ism in 10 %. Finally, anticonvulsants were given to 9 % of the patients and improved
parkinsonism, especially tremor, in 23 % of them, a success rate second only to
levodopa. However, the study did not report the magnitude and duration of observed
improvements. Dyskinesias did not occur even at high doses of dopaminergic
therapy.
Overall, the response of parkinsonism to therapy is limited, with levodopa being
the most efficacious agent. In patients who do not respond to levodopa, dopamine
receptor agonists such as pramipexole or ropinirole may be considered. Indeed,
unlike levodopa, these medications act directly at the postsynaptic terminal in the
striatum, bypassing the substantia nigra which, similar to PSP, is also affected with
severe neuronal loss in CBS (Rabinovici and Miller 2010). Finally, anticonvulsants
might be useful in tremulous CBS patients.
No empiric data exists for the treatment of parkinsonism in FTLD, but some expert
opinions have been published on the subject. A trial of levodopa should be attempted in
FTLD patients with parkinsonism, but the response to levodopa may be poor or transient
68 R. Mehanna and H.H. Fernandez
(Rabinovici and Miller 2010). Dopamine receptor agonists, such as pramipexole or rop-
inirole, should be considered in cases of levodopa failure (Rabinovici and Miller 2010).
Contrary to patients with DLB, FTLD patients are less prone to hallucinations;
thus, dopaminergic medications can be used more liberally. While not specifically
assessed for the treatment of parkinsonism in FTLD, the potential benefit of dopa-
mine agonists on apathy, perseveration, and executive functioning (Imamura et al.
1998; Rahman et al. 2001; Allain et al. 2003) and of selegiline on neuropsychologi-
cal symptoms (Moretti et al. 2002) should prompt the consideration of these anti-
parkinsonian treatments in FTLD patients with parkinsonism.
Parkinsonism associated with FTLD is usually of the non-tremulous type. If
tremor is a major source of complaints, anticholinergic medications can be consid-
ered but should be used very sparingly as they may worsen the cognitive and neuro-
psychiatric symptoms.
Frontotemporal dementia with parkinsonism is an autosomal dominant syn-
drome linked to chromosome 17 than be caused by tenths of different mutations
(Graff-Radford and Woodruff 2007). It can be subdivided in two clinical pheno-
types (Reed et al. 2001), one with a dementia-predominant symptomatology and the
other with a parkinsonism-predominant phenotype. Most patients respond poorly, if
at all, to levodopa (Wszolek et al. 2006). FTLD can also overlap clinically and
pathologically with PSP and/or CBD (Graff-Radford and Woodruff 2007). In these
cases, the symptomatic treatment of parkinsonism would be as detailed in the sec-
tion on PSP and CBD, respectively.
Conclusion
References
Goldman JG, Goetz CG, Brandabur M, Sanfilippo M, Stebbins GT. Effects of dopaminergic medi-
cations on psychosis and motor function in dementia with Lewy bodies. Mov Disord.
2008;23(15):2248–50.
Graff-Radford NR, Woodruff BK. Frontotemporal dementia. Semin Neurol. 2007;27(1):48–57.
Imamura T, Takanashi M, Hattori N, et al. Bromocriptine treatment for perseveration in demented
patients. Alzheimer Dis Assoc Disord. 1998;12:109–13.
Irene L, Yves A, editors. Progressive supranuclear palsy: clinical and research approaches. New
York: Oxford University Press; 1992.
Jankovic J, Newmark M, Peter P. Parkinsonism and acquired hydrocephalus. Mov Disord.
1986;1:59–64.
Kompoliti K, Goetz CG, Litvan I, Jellinger K, Verny M. Pharmacological therapy in progressive
supranuclear palsy. Arch Neurol. 1998a;55(8):1099–102.
Kompoliti K, Goetz CG, Boeve BF, Maraganore DM, Ahlskog JE, Marsden CD, et al. Clinical
presentation and pharmacologic therapy in corticobasal degeneration. Arch Neurol.
1998b;55:957–61.
Kurlan R, Richard IH, Papka M, Marshall F. Movement disorders in Alzheimer's disease: more
rigidity of definitions needed. Mov Disord. 2000;15:24–9.
Liepelt-Scarfone I, Jamour M, Maetzler W. Co-occurrence of parkinsonism and dementia in clini-
cal practice: relevant differential diagnoses. Z Gerontol Geriatr. 2012;45(1):23–33.
Lim T, Ahmed A, Itin I, Gostkowski M, Rudolph J, Cooper S, Fernandez HH. Is 6 months of neu-
roleptic withdrawal sufficient to distinguish drug-induced parkinsonism from Parkinson’s dis-
ease? Int J Neurosci. 2013;123(3):170–4.
Lubarsky M, Juncos JL. Progressive supranuclear palsy: a current review. Neurologist.
2008;14(2):79–88.
Lucetti C, Logi C, Del Dotto P, Berti C, Ceravolo R, Baldacci F, Dolciotti C, Gambaccini G, Rossi
G, Bonuccelli U. Levodopa response in dementia with Lewy bodies: a 1-year follow-up study.
Parkinsonism Relat Disord. 2010;16(8):522–6.
Mehanna R, Jankovic J. Respiratory problems in neurologic movement disorders. Parkinsonism
Relat Disord. 2010;16(10):628–38.
Mehanna R, Jankovic J. Movement disorders in multiple sclerosis and other demyelinating dis-
eases. J Neurol Sci. 2013a;328(1–2):1–8.
Mehanna R, Jankovic J. Movement disorders in cerebrovascular disease. Lancet Neurol.
2013b;12(6):597–608.
Morishita T, Foote KD, Okun MS. INPH and Parkinson disease: differentiation by levodopa
response. Nat Rev Neurol. 2010;6(1):52–6.
McKeith IG, Galasko D, Kosaka K, Perry EK, Dickson DW, Hansen LA, et al. Consensus
guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB):
report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113–24.
McKeith IG, Burn DJ, Ballard CG, Collerton D, Jaros E, Morris CM, et al. Dementia with Lewy
bodies. Semin Clin Neuropsychiatry. 2003;8(1):46–57.
McKeith IG, Dickson DW, Lowe J, Emre M, O’Brien JT, Feldman H, et al., Consortium on DLB.
Diagnosis and management of dementia with Lewy bodies: third report of the DLB Consortium.
Neurology. 2005;65(12):1863–72.
Molloy S, McKeith IG, O'Brien JT, Burn DJ. The role of levodopa in the management of dementia
with Lewy bodies. J Neurol Neurosurg Psychiatry. 2005;76(9):1200–3.
Moretti R, Torre P, Antonello RM, Cazzato G, Bava A. Effects of selegiline on frontotemporal
dementia: a neuropsychological evaluation. Int J Geriatr Psychiatry. 2002;17:391–2.
Murata M, Hasegawa K, Kanazawa I, The Japanese Zonisamide on PD Study Group. Zonisamide
improves motor function in Parkinson disease: a randomized, doubleblind study. Neurology.
2007;68:45–50.
Nieforth KA, Golbe LI. Retrospective study of drug response in 87 patients with progressive
supranuclear palsy. Clin Neuropharmacol. 1993;16:338–46.
Odawara T, Shiozaki K, Togo T, Hirayasu Y. Administration of zonisamide in three cases of
dementia with Lewy bodies. Psychiatry Clin Neurosci. 2010;64(3):327–9.
4 Treatment of Parkinsonism in Patients with Non-Parkinson Dementia 71
Papapetropoulos S, Lieberman A, Gonzalez J, Singer C, Laufer DZ, Mash DC. Family history of
dementia: dementia with Lewy bodies and dementia in Parkinson’s disease. J Neuropsychiatry
Clin Neurosci. 2006;18(1):113–6.
Parkinson Study Group. Pramipexole vs. levodopa as initial treatment for Parkinson disease. A
randomized controlled trial. JAMA. 2000;84:1931–8.
Possin KL, Kaufer DI. Parkinsonian dementias. Continuum. 2010;16(2 Dementia):57–79.
Poewe W. Treatment of dementia with Lewy bodies and Parkinson's disease dementia. Mov
Disord. 2005;20 Suppl 12:S77–82.
Pujari S, Kharkar S, Metellus P, Shuck J, Williams MA, Rigamonti D. Normal pressure hydro-
cephalus: long-term outcome after shunt surgery. J Neurol Neurosurg Psychiatry.
2008;79(11):1282–6.
Racette BA, Esper GJ, Antenor J, Black KJ, Burkey A, Moerlein SM, et al. Pathophysiology of
parkinsonism due to hydrocephalus. J Neurol Neurosurg Psychiatry. 2004;75:1617–9.
Rahman S, Robbins TW, Sahakian BJ. Comparative cognitive neuropsychological studies of fron-
tal lobe function: implications for therapeutic strategies in frontal variant frontotemporal
dementia. Dement Geriatr Cogn Disord. 2001;10 Suppl 1:15–28.
Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the
incidence of dyskinesia in patients with Parkinson’s disease who were treated with ropinirole
or levodopa. 056 Study Group. N Engl J Med. 2000;18(342):1481–91.
Rabinovici GD, Miller BL. Frontotemporal lobar degeneration: epidemiology, pathophysiology,
diagnosis and management. CNS Drugs. 2010;24(5):375–98.
Reed LA, Wszolek ZK, Hutton M. Phenotypic correlations in FTDP-17. Neurobiol Aging.
2001;22:89–107.
Sato S, Mizukami K, Asada T. Successful treatment of extrapyramidal and psychotic symptoms
with zonisamide in a patient with dementia with Lewy bodies. Prog Neuropsychopharmacol
Biol Psychiatry. 2010;34(6):1130–1.
Serrano-Duenas M. Neuroleptic malignant syndrome-like, or—dopaminergic malignant syn-
drome—due to levodopa therapy withdrawal. Clinical features in 11 patients. Parkinsonism
Relat Disord. 2003;9:175–8.
van Balken I, Litvan I. Current and future treatments in progressive supranuclear palsy. Curr Treat
Options Neurol. 2006;8:211–23.
Vanneste JA. Diagnosis and management of normal-pressure hydrocephalus. J Neurol.
2000;247:5–14.
Wilson RS, Schneider JA, Bienias JL, Evans DA, Bennett DA. Parkinsonian like signs and risk of
incident Alzheimer disease in older persons. Arch Neurol. 2003;60:539–44.
Wurtman RJ. Personalized medicine strategies for managing patients with parkinsonism and cog-
nitive deficits. Metabolism. 2013;62 Suppl 1:S27–9.
Wszolek ZK, Tsuboi Y, Ghetti B, Pickering-Brown S, Baba Y, Cheshire WP. Frontotemporal
dementia and parkinsonism linked to chromosome 17 (FTDP-17). Orphanet J Rare Dis.
2006;1:30.
Yoshikawa H, Oda Y, Sakajiri K, Takamori M, Nakanishi I, Makifuchi T, et al. Pure akinesia mani-
fested neuroleptic malignant syndrome: a clinical variant of progressive Supranuclear palsy.
Acta Neuropathol. 1997;93:306–9.
Zeidler M, Dorman PJ, Ferguson IT, Bateman DE. Parkinsonism associated with obstructive
hydrocephalus due to idiopathic aqueductal stenosis. J Neurol Neurosurg Psychiatry.
1998;64(5):657–9.
Zupancic M, Mahajan A, Handa K. Dementia with Lewy bodies: diagnosis and management for
primary care providers. Prim Care Companion CNS Disord. 2011;13(5). pii: PCC.11r01190.
doi: 10.4088/PCC.11r01190. PMID:22295275.
Chapter 5
Psychiatric Complications of Alzheimer’s
Disease Overlapping with Parkinsonism:
Depression, Apathy, Catatonia, and Psychosis
Introduction
For instance, depression and apathy may mimic the bradykinesia and bradyphrenia
of AD patients with parkinsonism; catatonia may mimic rigidity and blunted expres-
sion, and side effects of psychotropic drugs may produce not only parkinsonism but
also “positive” motor disorders such as akathisia and dyskinesia.
This chapter will review the phenomenology, frequency and treatment of depres-
sion, and apathy and catatonia in AD, with special emphasis on their motor mani-
festations in AD. We will conclude by examining the motor side effects on
antipsychotic medication frequently used in dementia.
Frequency of Depression in AD
Management of Depression in AD
and active treatment. Parkinsonism was not reported a significant side effect of the
active compounds (Nelson and Devanand 2011).
Enache et al. identified 11 RCTs for depression in AD or other types of dementia,
with a total of 1,514 patients included. Of these, five studies reported that the antide-
pressant (sertraline, clomipramine, maprotiline, moclobemide, or citalopram) was more
effective than placebo, whereas six studies using sertraline, mirtazapine, venlafaxine,
imipramine, fluoxetine, or estrogen replacement therapy were negative. No parkinson-
ism was significantly associated with any of these medications (Enache et al. 2011).
The impact of discontinuing antidepressants among individuals with dementia
was examined in a RCT that included 128 patients with dementia who had been
prescribed escitalopram, citalopram, sertraline, or paroxetine for 3 months or more.
The main finding was that patients who discontinued antidepressant treatment had
significantly higher depression scores after 25 weeks as compared to the continua-
tion group. Parkinsonism or other motor problems were not reported as relevant side
effects of any of the psychotropic compounds (Bergh et al. 2012).
In conclusion, the efficacy of antidepressants in AD remains unclear. Two large
RCTs demonstrated lack of efficacy for sertraline as compared to placebo, and one
of them also showed lack of efficacy for mirtazapine. On the other hand, discontinu-
ation of antidepressants was found to lead to a significant relapse of depression.
Parkinsonism or other motor problems were not reported as significant side effects
in any of the major RCTs or meta-analyses. One limitation of these studies is that
the method used to asses for parkinsonism was not clearly specified.
The question arises as to whether AD patients with parkinsonism may have a higher
frequency of depression as compared to AD patients without parkinsonism. Choi
et al. (2013) assessed parkinsonism in a series of 2,614 neuroleptic-free AD patients
using a structured neurologic evaluation. After controlling for demographic, clini-
cal, and cognitive variables, they found that parkinsonism in AD was significantly
associated with higher depression scores, as measured with the Geriatric Depression
Scale-15 (GDS-15) (Choi et al. 2013). Starkstein et al. (1996a) compared 33 patients
with AD and 33 patients with PD matched for age, gender, and MMSE score. Major
depression was significantly more frequent among PD patients (30 %) as compared
to AD patients (6 %) (P < 0.05).
Diagnostic Issues
Frequency of Apathy in AD
Starkstein et al. (2001) examined the prevalence of apathy in a study that included a
consecutive series of 319 patients with AD, 117 patients with depression but no
dementia, and 36 age-comparable healthy individuals (Starkstein et al. 2001). Based
on apathy scale scores, apathy was diagnosed in 37 % of the AD patients, as
5 Psychiatric Complications of Alzheimer’s Disease Overlapping with Parkinsonism 79
Treatment of Apathy in AD
There are no RCTs specifically designed to assess the efficacy of psychotropic com-
pound to treat apathy in AD, and most of the literature is based on small case series
or RCTs with apathy as a secondary outcome measure.
In a study of a series of 40 AD patients treated with the anticholinesterase inhibi-
tor tacrine, Kaufer and colleagues (1998) reported a significant reduction of apathy
in the group with moderate dementia (Kaufer et al. 1998), without significant motor
side effects. Mega and coworkers (2005) examined changes in the severity of apathy
in 19 patients with mild to moderate AD examined before and after treatment with
the cholinesterase inhibitor galantamine. There was no significant change in clinical
outcomes (Mega et al. 2005), but no parkinsonism was reported. A recent 6-week
RCT that included 67 AD patients with apathy treated with methylphenidate
(Rosenberg et al. 2013) failed to show significant benefits of the active compound
as compared to placebo.
Psychostimulants have been used to treat apathy in AD in small series of patients,
but no formal RCTs have been carried out. Galynker et al. (1997) reported a reduc-
tion in negative symptoms, as measured with the Scale for the Assessment of
Negative Symptoms (SANS) in 12 patients with AD treated with methylphenidate.
Parkinsonism was not reported to be a side effect of treatment (Galynker et al. 1997).
Siddique et al. (2009) assessed the efficacy of the SSRI citalopram (mean dose =
30 mg/day) on apathy NPI scores in a sample of 44 patients with AD previously
treated with placebo in the context of a RCT (Siddique et al. 2009). While patients on
citalopram showed a reduction of 60 % on the NPI apathy scale as compared to pla-
cebo treatment, this difference was not statistically significant (Siddique et al. 2009).
Parkinsonian signs are frequent in AD (see chapter by Starkstein and Merello) and
are associated with a faster cognitive decline, worse quality of life, and early
80 S.E. Starkstein and J. Pahissa
Catatonia
Diagnosis of Catatonia
Frequency of Catatonia in AD
A recent study examined for the presence of catatonia all those patients referred to
a consultation liaison service in a general hospital. Using the BFCS, the authors
diagnosed catatonia in 9 % of the consults (Jaimes-Albornoz and Serra Mestres
82 S.E. Starkstein and J. Pahissa
2013). Rates of catatonia may be higher among hospitalized medical patients given
that withdrawn patients (such those with a hypokinetic delirious) may not be recog-
nized as catatonic (Zarr and Nowak 1990; Carroll et al. 2000; Cottencin et al. 2007).
Treatment of Catatonia
Starkstein et al. (1996b) reported that 20 % of 79 older adults with major depression
referred to a psychiatric service met DSM-IV criteria for catatonia. A regression
analysis demonstrated that UPDRS scores, HAM-D scores, and older age contrib-
uted significantly to catatonia scores. When patients with catatonia were matched
with non-catatonic patients with PD based on UPDRS scores, catatonic patients had
significantly higher scores on the DSM-IV clusters of stupor, excessive motor activ-
ity, extreme negativism, and posturing. Furthermore, apomorphine did not improve
catatonic symptoms (Starkstein et al. 1996b).
Alzheimer’s disease (AD), the most common cause of dementia in the elderly, is
often associated with psychotic symptoms. The prevalence of psychosis in AD was
recently estimated to be 7 %, with a 2-year cumulative incidence of 15 % (Vilalta-
Franch et al. 2012). Psychotic symptoms persisted for 1 year or more in 69 % of
patients with psychosis at baseline (Vilalta-Franch et al. 2012).
De Deyn et al. (2013) examined the efficacy of aripiprazole in a RCT that
included 208 AD patients with psychosis. Aripiprazole was started at 2 mg/day and
titrated upward up to 15 mg/day) depending on efficacy (mean dose = 10 mg/day).
5 Psychiatric Complications of Alzheimer’s Disease Overlapping with Parkinsonism 83
Efficacy was similar for both active drug and placebo groups, and there were no
between-group differences on the frequency of parkinsonism (De Deyn et al. 2013).
Schneider et al. (2006) evaluated the effectiveness of atypical antipsychotic
drugs for the treatment of psychosis, aggression, or agitation in AD. This RCT
included 421 outpatients with AD and psychosis, aggression, or agitation who were
randomly assigned to receive olanzapine, quetiapine, risperidone, or placebo for up
to 36 weeks. The main outcomes were the time from initial treatment to the discon-
tinuation of treatment for any reason and the number of patients with at least mini-
mal improvement on the Clinical Global Impression of Change (CGIC) scale at
12 weeks. Overall, 24 % of patients who received olanzapine, 18 % of patients who
received risperidone, 16 % of patients who received quetiapine, and 5 % of patients
who received placebo discontinued their assigned treatment owing to intolerability
(P < 0.01). No significant differences were noted on the CGIC scale by the end of
the follow-up. Behavioral improvement was observed in 32 % of patients assigned
to olanzapine, 29 % of patients assigned to risperidone, 26 % of patients assigned
to quetiapine, and 21 % of patients assigned to placebo (P = 0.22) (Schneider
et al. 2006). Parkinsonism was significantly more frequent in patients on olanzap-
ine (12 %) or risperidone (12 %) as compared to quetiapine (2 %) or the placebo
groups (1 %).
Devanand and coworkers (2012) examined the risk of a recurrence of psychotic
symptoms after discontinuation of risperidone (mean dose = 0.97 mg daily) in
patients with AD for 4–8 months. The severity of psychosis and agitation were
reduced with risperidone, although there was a mild increase in parkinsonism, and
discontinuation of risperidone was associated with an increased risk of relapse.
During the first 16-week period, there were no significant differences in adverse
events as defined by increases above prespecified thresholds on scales measuring
parkinsonism between patients receiving risperidone and those receiving placebo.
Moreover, there were no significant differences between patients who received ris-
peridone continuously for 32 weeks and those who received placebo with respect to
parkinsonism and other movement disorders (Devanand et al. 2012).
Rocca et al. (2007) reported findings on a retrospective, naturalistic study on the
effects of 6 months’ treatment with risperidone, olanzapine, or quetiapine on behav-
ioral disturbances in outpatients with mild to moderate AD. All three drugs produced
significant improvements in behavioral disturbances, and medications were well tol-
erated with no significant differences emerging among treatments (Rocca et al. 2007).
Chiabrando et al. (2010) studied the prescriptive profile of antipsychotic drugs in
392 patients with dementia (49 % with AD) in terms of the choice of active sub-
stance and the clinical characteristics of patients. Hallucinations were present in
50 % of the cases and aggression in 53 %. There was an increased consumption of
quetiapine and a parallel decrease in the use of risperidone and olanzapine during
the study period. Neuroleptic doses were on average much lower than those used for
treating non-AD psychoses. The most frequently observed adverse events were
tremors (Chiabrando et al. 2010).
84 S.E. Starkstein and J. Pahissa
Conclusion
References
American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 4th ed.
Washington, DC: American Psychiatric Press; 1994.
American Psychiatric Association. Diagnostic and statistical manual of mental disorders. 5th ed.
Arlington: American Psychiatric Publishing; 2013.
Bains J, Birks JS, Dening TR. The efficacy of antidepressants in the treatment of depression in
dementia. Cochrane Database Syst Rev. 2002;(4):CD003944.
Banerjee S, Hellier J, Dewey M, et al. Sertraline or mirtazapine for depression in dementia (HTA-
SADD): a randomised, multicentre, double-blind, placebo-controlled trial. Lancet. 2011;378:
403–11.
Bergh S, Selbæk G, Engedal K. Discontinuation of antidepressants in people with dementia and
neuropsychiatric symptoms (DESEP study): double blind, randomised, parallel group, placebo
controlled trial. BMJ. 2012;344:e1566.
Bush G, Fink M, Petrides G, Dowling F, Francis A. Catatonia. I. Rating scale and standardized
examination. Acta Psychiatr Scand. 1996;93(2):129–36.
Carroll BT, Kennedy JC, Goforth HW. Catatonic signs in medical and psychiatric catatonias. CNS
Spectr. 2000;5:66–9.
Chiabrando G, Bianchi S, Poluzzi E, Montanaro N, Scanavacca P. Profile of atypical-antipsychotics
use in patients affected by dementia in the University Hospital of Ferrara. Eur J Clin Pharmacol.
2010;66(7):661–9.
Choi J, Myung W, Chung JW, et al. Association between functional impairment, depression, and
extrapyramidal signs in neuroleptic-free patients with Alzheimer disease. J Geriatr Psychiatry
Neurol. 2013;26(3):144–50.
Cohen-Cole SA, Stoudemire A. Major depression and physical illness. Psychiatr Clin North Am.
1987;10:1–17.
Cottencin O, Warembourg F, de Chouly de Lenclave M, et al. Catatonia and consultation-liaison
psychiatry study of 12 cases. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31:1170–6.
De Deyn PP, Drenth AF, Kremer BP, Oude Voshaar RC, Van Dam D. Aripiprazole in the treatment
of Alzheimer’s disease. Expert Opin Pharmacother. 2013;14(4):459–74.
Devanand DP, Mintzer J, Schultz SK, et al. Relapse risk after discontinuation of risperidone in
Alzheimer’s disease. N Engl J Med. 2012;367(16):1497–507.
Enache D, Winblad B, Aarsland D. Depression in dementia: epidemiology, mechanisms, and treat-
ment. Curr Opin Psychiatry. 2011;24(6):461–72.
5 Psychiatric Complications of Alzheimer’s Disease Overlapping with Parkinsonism 85
Fink M, Shorter E, Taylor MA. Catatonia is not Schizophrenia: Kraepelin’s error and the need to
recognize catatonia as an independent syndrome in medical nomenclature. Schizophr Bull.
2010;36:314–20.
Francis A. Catatonia: diagnosis, classification, and treatment. Curr Psychiatry Rep. 2010;12:
180–5.
Gallo JJ, Rabins PV, Anthony JC. Sadness in older persons: 13-year follow-up of a community
sample in Baltimore, Maryland. Psychol Med. 1999;29:341–50.
Galynker I, Ieronimo C, Miner C, Rosenblum J, Vilkas N, Rosenthal R. Methylphenidate treatment
of negative symptoms in patients with dementia. J Neuropsychiatry Clin Neurosci.
1997;9(2):231–9.
Jaimes-Albornoz W, Serra-Mestres J. Catatonia in the emergency department. Emerg Med
J. 2012;29:863–7.
Jaimes-Albornoz W, Serra-Mestres J. Prevalence and clinical correlations of catatonia in older
adults referred to a liaison psychiatry service in a general hospital. Gen Hosp Psychiatry.
2013;35:512–6.
Kahlbaum KL. Die Katatonie oder das Sannungsirreiseins. Berlin: Hirschwald; 1874.
Kaufer D, Cummings JL, Christine D. Differential neuropsychiatric symptom responses to tacrine
in Alzheimer's disease: relationship to dementia severity. J Neuropsychiatry Clin Neurosci.
1998;10(1):55–63.
Lyketsos CG, Breitner JC, Rabins PV. An evidence-based proposal for the classification of neuro-
psychiatric disturbance in Alzheimer's disease. Int J Geriatr Psychiatry. 2001;11:1037–42.
Marin RS. Apathy: a neuropsychiatric syndrome. J Neuropsychiatry Clin Neurosci.
1991;3:243–54.
Mega MS, Dinov ID, Porter V, et al. Metabolic patterns associated with the clinical response to
galantamine therapy: a fludeoxyglucose f 18 positron emission tomographic study. Arch
Neurol. 2005;62(5):721–8.
Migliorelli R, Teson A, Sabe L, Petrachi M, Leiguarda R, Starkstein SE. Prevalence and correlates
of dysthymia and major depression among patients with Alzheimer’s disease. Am J Psychiatry.
1995;152:37–44.
Mulin E, Leone E, Dujardin K, et al. Diagnostic criteria for apathy in clinical practice. Int J Geriatr
Psychiatry. 2011;26(2):158–65. doi:10.1002/gps.2508.
Nelson JC, Devanand DP. A systematic review and meta-analysis of placebo-controlled antide-
pressant studies in people with depression and dementia. J Am Geriatr Soc. 2011;59:577–85.
Northoff G, Koch A, Wenke J, et al. Catatonia as a psychomotor syndrome: a rating scale and
extrapyramidal motor symptoms. Mov Disord. 1999;14:404–16.
Olin JT, Katz IR, Meyers BS, Schneider LS, Lebowitz BD. Provisional diagnostic criteria for
depression of Alzheimer disease: rationale and background. Am J Geriatr Psychiatry.
2002a;10(2):129–41.
Olin JT, Schneider LS, Katz IR, et al. Provisional diagnostic criteria for depression of Alzheimer
disease. Am J Geriatr Psychiatry. 2002b;10(2):125–8.
Richard E, Reitz C, Honig LH, et al. Late-life depression, mild cognitive impairment, and demen-
tia. JAMA Neurol. 2013;70(3):374–82.
Rocca P, Marino F, Montemagni C, et al. Risperidone, olanzapine and quetiapine in the treatment
of behavioral and psychological symptoms in patients with Alzheimer's disease: preliminary
findings from a naturalistic, retrospective study. Psychiatry Clin Neurosci. 2007;61(6):622–9.
Rosenberg PB, Drye LT, Martin BK, et al. Sertraline for the treatment of depression in Alzheimer
disease. Am J Geriatr Psychiatry. 2010;18(2):136–45.
Rosenberg PB, Lanctôt KL, Drye LT, Herrmann N, Scherer RW, Bachman DL, Mintzer JE. Safety
and efficacy of methylphenidate for apathy in Alzheimer's disease: a randomized, placebo-
controlled trial. J Clin Psychiatry. 2013;74:810–6.
Schneider LS, Tariot PN, Dagerman KS, et al. Effectiveness of atypical antipsychotic drugs in
patients with Alzheimer's disease. N Engl J Med. 2006;355(15):1525–38.
86 S.E. Starkstein and J. Pahissa
Siddique H, Hynan LS, Weiner MF. Effect of a serotonin reuptake inhibitor on irritability, apathy,
and psychotic symptoms in patients with Alzheimer’s disease. J Clin Psychiatry. 2009;70:6.
Starkstein SE, Leentjens AF. The nosological position of apathy in clinical practice. J Neurol
Neurosurg Psychiatry. 2008;79:1088–92.
Starkstein SE, Petracca G, Teson A, et al. Catatonia in depression: prevalence, clinical correlates,
and validation of a scale. J Neurol Neurosurg Psychiatry. 1996a;60:326–32.
Starkstein SE, Sabe L, Petracca G, et al. Neuropsychological and psychiatric differences between
Alzheimer's disease and Parkinson's disease with dementia. J Neurol Neurosurg Psychiatry.
1996b;61:381–7.
Starkstein SE, Petracca G, Chemerinski E, Kremer J. Syndromic validity of apathy in Alzheimer's
disease. Am J Psychiatry. 2001;158(6):872–7.
Starkstein SE, Ingram L, Garau ML, Mizrahi R. On the overlap between apathy and depression in
dementia. J Neurol Neurosurg Psychiatry. 2005a;76(8):1070–4.
Starkstein SE, Jorge R, Mizrahi R, Robinson RG. The construct of minor and major depression in
Alzheimer's disease. Am J Psychiatry. 2005b;162(11):2086–93.
Starkstein SE, Jorge R, Mizrahi R, Robinson RG. A prospective longitudinal study of apathy in
Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2006;77:8–11.
Starkstein SE, Brockman S, Bruce D, Petracca G. Anosognosia is a significant predictor for apathy
in Alzheimer’s disease. J Neuropsychiat Clin Neurosci 2010;22(2):378–83.
Starkstein SE, Dragovic M, Jorge R, Brockman S, Robinson RG. Diagnostic criteria for depression
in Alzheimer disease: a study of symptom patterns using latent class analysis. Am J Geriatr
Psychiatry. 2011;19(6):551–8.
Stauder K. Die totliche Katatonie. Archives fur Neurologie und Psychiatrie. 1934;23:614–32.
Thompson S, Herrmann N, Rapoport MJ, Lanctôt KL. Efficacy and safety of antidepressants for treat-
ment of depression in Alzheimer's disease: a metaanalysis. Can J Psychiatry. 2007;
52(4):248–55.
Vilalta-Franch J, López-Pousa S, Calvó-Perxas L, Garre-Olmo J. Psychosis of Alzheimer disease:
prevalence, incidence, persistence, risk factors, and mortality. Am J Geriatr Psychiatry.
2012;21(11):1135–43.
Vilalta-Franch J, Calvo-Perxas L, Garre-Olmo J, Turro Garriga O, Lopez-Pousa S. Apathy syn-
drome in Alzheimer’s disease epidemiology: prevalence, incidence, persistence, and risk and
mortality factors. J Alzheimers Dis. 2013;33:535–43.
Weintraub D, Rosenberg PB, Drye LT, et al. Sertraline for the treatment of depression in Alzheimer
disease: week-24 outcomes. Am J Geriatr Psychiatry. 2010;18(4):332–40.
Zarr ML, Nowak T. Catatonia and burns. Burns. 1990;16:133–4.
Chapter 6
Drug-Induced Movement Disorders
in Elderly Patients
Symptoms and time to onset of DIMDs vary significantly and include parkinsonism
as well as motor restlessness (akathisia), dystonia, and the entire spectrum of hyper-
kinesias (namely, chorea, stereotypies, myoclonus, dystonia, and tics) (Caligiuri
et al. 2000). They can be categorized as acute (immediate), continuous (insidious),
or persistent (tardive) (Table 6.1) (Rodnitzky 2002). Dystonia represents the most
frequent acute drug reaction. Its development is fast, and in some cases it may be as
severe as to require hospitalization. Continuous DIMDs persist only while the
offending drug is being administered and remit after its discontinuation, either
immediately or a variable period of time after discontinuation. They include akathi-
sia, tremor, parkinsonism, chorea, and myoclonus. In case of parkinsonism, lack of
remission after discontinuation could indicate that drug exposition unmasked a pre-
existent PD (Lopez-Sendon et al. 2013). Finally, tardive syndromes consist in a
variety of DIMDs appearing long after beginning drug use. Typically, offending
drug discontinuation will not relieve this kind of disorders (Rodnitzky 2002).
Acute Dystonia
Acute Akathisia
Akathisia (Greek “not to sit”) consists in difficulty remaining still and a subjective
sense of restlessness (Bakheit 1997; Akagi and Kumar 2002). It is a well-known
adverse drug reaction of antipsychotics, antiemetics, and antidepressants, among
others (Akagi and Kumar 2002). Difficult to detect reliably, it may present unex-
pectedly in a variety of clinical settings and be accompanied by unpleasant oral or
genital paresthesia, burning, or lancinating pain not responding to conventional
treatments. Principal risk factors are advanced age, presence of an affective disor-
der, cognitive impairment, female gender, and mental retardation.
Principal differential diagnoses are restless legs syndrome (RLS), mania (in
bipolar patients), and dyskinesias (Bakheit 1997). RLS is characterized by muscle
discomfort, pain and restlessness, or crawling sensations relieved by walking.
Unlike akathisia, which ceases during sleep, RLS occurs mostly at night.
Hyperactivity associated with anxiety states is often indistinguishable from akathi-
sia, especially in psychotic patients on neuroleptics. However, sympathetic overac-
tivity, for example, excessive sweating, palpitations, hyperventilation, tremulousness,
and dilated pupils, characteristic of anxiety and panic attacks, is not seen in patients
6 Drug-Induced Movement Disorders in Elderly Patients 91
with akathisia. In bipolar affective disorders, akathisia can be confused with spon-
taneous mania, although usually milder and short-lived. In addition, spontaneous
mania is usually accompanied by delusions, hallucinations, and bizarre behavior.
Dyskinesia, in contrast to akathisia, is often unilateral or if bilateral tends to be
more pronounced in the more severely affected arm and leg. As a rule, it increases
in severity approximately 1–2 h after each levodopa and/or dopamine agonist dose,
although not always.
Parkinsonism
DIP is the second most common cause of parkinsonian syndrome (Mena and de
Yebenes 2006). The diversity of drugs involved in the production of DIP and the
wide range of clinical disorders in which they are used poses a tough diagnostic
challenge (Esper and Factor 2008). Several population-based studies suggest a prev-
alence between 1.7 and 2.7 % (Barbosa et al. 2006; Benito-Leon et al. 2004; Seijo-
Martinez et al. 2011).
DIP is characterized by its symmetrical presentation with bradykinesia dominat-
ing the overall clinical picture (Mena and de Yebenes 2006; Gershanik 1994).
Typical resting tremor is not frequently observed but when present is postural and of
higher frequency than in idiopathic disease. It develops insidiously after the offend-
ing drug is introduced, taking weeks or months to manifest fully. Cognitive impair-
ment is also frequently present (Kim et al. 2011). Main risk factors are older age,
female gender, and cognitive impairment (Barbosa et al. 2006; Kim and Byun 2009).
The main differential diagnoses are idiopathic PD and the parkinsonian form of
multiple system atrophy (Mena and de Yebenes 2006). DIP should be suspected in
older patients, more prone to take different medications for underlying chronic con-
ditions, including whenever symmetrical symptoms are present, disease onset is not
compatible with idiopathic Parkinson’s disease, or akinesia and postural tremor pre-
dominate over rigidity and rest tremor (Esper and Factor 2008; Gershanik 1994).
Other drug-induced symptoms like akathisia or tardive dyskinesia can provide clues
to potential parkinsonian syndrome origin.
Tardive syndromes often run a persistent course despite cessation of triggering drug
therapy. In some instances, they may become permanent and irreversible. They
should be considered in patients presenting abnormal involuntary movements after
at least 3 months of total cumulative neuroleptic exposure, although they are more
common after longer periods of exposure (1–2 years) (Casey 1990; Caligiuri et al.
2000). They can develop even after antipsychotics dose reduction (unmasked TD)
or even after the causative drug has been withdrawn (covert or withdrawal TD).
TD consists of involuntary movements usually involving muscles of the tongue,
lips, mouth, or face (i.e., the so-called buccolinguomasticatory syndrome) (Haddad
and Dursun 2008; Casey 1990; Caligiuri et al. 2000; Paulson 2005). Upper facial
muscles are less frequently affected by involuntary movements; however, it is pos-
sible to see increased blinking, blepharospasm, arching of the eyebrows, ocular tor-
sion, and deviation.
Other parts of the body can be affected, though less frequently, and a wide range
of movements can be observed including myoclonic jerks, tics, chorea, and dystonia.
Gait can be abnormal, with a broad base, leg jerking, and repetitive irregular flexion
and extension of the knees (Haddad and Dursun 2008; Tarsy 1983; Casey 1990).
While standing in place, affected individuals tend to shift their weight from one leg
to the other or exhibit pacing or marching in place. The diaphragm and accessory
respiratory muscles are often involved causing a fast and irregular breathing pattern
(respiratory dyskinesia). The movements are more pronounced when the patient is
alert or excited and disappear during sleep. Patients can sometimes suppress the
movements through intense voluntary effort. Main risk factors are older age and
female gender, presence of affective disorders, alcoholism, diabetes mellitus, electro-
convulsive treatment, iron deficiency, mental retardation, or organic brain disorder.
6 Drug-Induced Movement Disorders in Elderly Patients 93
Diagnostic Work-Up
During the course of any drug treatment, movement disorders not necessarily related
to intake may occur. Nonetheless, prescription drugs should always be considered a
differential diagnosis for any movement disorder, especially if the patient is on an
agent known to induce them. It should be kept in mind that subjects may not readily
recall all the medications they receive.
Causality assessment is indispensable but many times difficult. The following
aspects of the event should be considered (Rehan et al. 2009; Edwards and Aronson
2000; Montastruc et al. 2006):
• Timing in relation to drug intake. When symptoms begin soon after drug expo-
sure starts, diagnosis may be easy; however, connecting symptoms to long-term
drug use may be difficult.
• Plausibility of the event. If the event result from a known pharmacodynamic
property of the drug (i.e., D2-blockage properties of neuroleptics), it may be
easier to connect to the drug. Nonetheless, in some cases DIMD pathophysiol-
ogy may not be known.
• Exclusion of other causes. DIMD may be diagnosed only after exclusion of
every other possible cause for the event observed.
• Dechallenge may be of aid when feasible. Disappearance of DIMD after drug
discontinuation is indicative of a link to the drug. Nonetheless, some DIMDs
such as TD do not disappear after drug discontinuation.
• Rechallenge, when possible, may lead to the reappearance of the movement dis-
order, thus reassuring its drug-induced nature.
Clinical observation is crucial for DIMD differential diagnosis. In the case of DIP,
while motor symptoms may not allow proper differentiation with PD, the absence of
non-motor symptoms favors the former (Kim et al. 2013a). DIP patients have normal
olfactory function except when dopaminergic loss was present in patients (Bovi et al.
2010). Clinical laboratory is in general not helpful for the diagnosis of DIMDs.
Nonetheless, increased serum hyperprolactinemia can be used as a marker of dopa-
mine receptor blockage when antipsychotics are used (Kinon et al. 2003).
94 S. Perez-Lloret et al.
For DIP and TD however, some diagnostic tools are available. Firstly, acute
dopaminergic challenge with either levodopa or apomorphine may represent a use-
ful tool for differentiating PD from DIP (Merello et al. 2002). Schizophrenic patients
with DIP show a noteworthy absence of response to levodopa or apomorphine dur-
ing a levodopa acute challenge (Merello et al. 1996).
Assessment of dopaminergic nigrostriatal pathway integrity can also be useful
distinguishing DIMD, in which they are intact, from Parkinson’s disease, in which
they are not. This can be accomplished by imaging with positron emission tomog-
raphy (PET) or single photon emission computed tomography (SPECT) using
ligands binding to dopaminergic nigrostriatal system markers (Tolosa et al. 2003).
The presence of the dopamine transporter in the presynaptical buttons of the dopa-
minergic neurons located in the striate nuclei can be assessed by [123I]FP-CIT
SPECT (Tolosa et al. 2003). Death of dopaminergic neurons, for example, in PD,
leads to a reduced number of dopamine transporter, which cause a reduction in the
intensity of the signal that the striatal level in the SPECT. In the case of [18F]-dopa-
PET, what is evaluated is the integrity of presynaptical dopaminergic structures in
charge of uptaking and processing DOPA to form dopamine. Results are interpreted
in an analogous way to those of SPECT.
Nonetheless, these techniques may not be enough in schizophrenic patients, in
whom D2-receptor blockade may coexist with a dopamine nigrostriatal terminal
defect (Tinazzi et al. 2012). In doubtful cases, combination of techniques might be
useful for differential diagnosis (Kim et al. 2013b; Lee et al. 2007). Assessment of
cardiac sympathetic denervation by using 123I-metaiodobenzylguanidine (MIBG)
myocardial scintigraphy may also provide further clues about the origin of the par-
kinsonian syndrome (Kim et al. 2013b; Lee et al. 2007).
For TD diagnosis, neuroimaging studies may helpful, as anomalies in basal gan-
glia and other brain regions in schizophrenic patients with TD have been identified
(Khiat et al. 2008). These anomalies include caudate nuclei, left lentiform nuclei,
and temporal sulci volume differences as well as reduction in T2 relaxation time in
the left caudate nuclei.
Offending Drugs
Antipsychotics
et al. 1997)
(continued)
Table 6.2 (continued)
96
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas Tic
2004)
Bupropion x (Montastruc et al. x (Montastruc
1994) and Durrieu
2004)
Venlafaxine x (Montastruc
and Durrieu
2004)
Nefazodone x (Montastruc
and Durrieu
2004)
Mirtazapine x (Montastruc
and Durrieu
2004)
Antiemetics
Metoclopramide xx (van Harten xx (Jimenez-Jimenez xx (Mena and de xx (Orti-Pareja xx (Morgan and xx (Montastruc
et al. 1999) et al. 1996) Yebenes 2006; et al. 1999; Sethi 2005) and Durrieu
Jimenez-Jimenez Blayac et al. 2004)
et al. 1996) 2004;
Jimenez-
Jimenez
6 Drug-Induced Movement Disorders in Elderly Patients
et al. 1996)
Domperidone x (Bonuccelli
et al. 1991)
Clebopride x (Montagna et al.
1992)
(continued)
97
Table 6.2 (continued)
98
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas Tic
Antipsychotics
Typical xx (van Harten xx (Bakheit 1997; xx (Bakheit 1997; xx (Bakheit xx (Blayac et al. xx (Montastruc x (Montastruc
et al. 1999) Jimenez-Jimenez Jimenez-Jimenez 1997; 2004) and Durrieu and
et al. 1996) et al. 1996) Jimenez- 2004) Durrieu
Jimenez 2004;
et al. 1996) Blayac
et al. 2004)
Atypical x (Bakheit x (Bakheit 1997; x (Bakheit 1997; x (Bakheit 1997; x (Bakheit 1997; x (Bakheit
1997; Gareri et al. Gareri et al. 2006) Gareri et al. Gareri et al. 1997;
Gareri 2006) 2006) 2006) Montastruc
et al. 2006) and Durrieu
2004;
Gareri et al.
2006)
Anxiolytics
Buspirone x (LeWitt et al. x (LeWitt et al. 1993) x (LeWitt et al. 1993) x (LeWitt et al. x (Jimenez- x (Montastruc
1993) 1993) Jimenez and Durrieu
et al. 1997) 2004;
LeWitt et al.
1993)
Diazepam x (Lang 1992) x (Jimenez-Jimenez x (Montastruc
et al. 1997) and Durrieu
2004)
Clonazepam x (Montastruc
and Durrieu
2004)
Lorazepam x (Jimenez-Jimenez x (Orti-Pareja
et al. 1996) et al. 1999)
S. Perez-Lloret et al.
CNS stimulants
Methamphetamine/ x (Morgan and Sethi x (Morgan and x (Rodnitzky x (Montastruc
amphetamines 2005) Sethi 2005) 2002) and
Durrieu
2004;
Blayac
et al. 2004)
Cocaine x (van Harten x (Chung and Chiu x (Morgan and Sethi x (Weiner et al. x (Morgan and xx (Jimenez- x (Montastruc
et al. 1999) 1996) 2005) 2001) Sethi 2005) Jimenez and
et al. 1997) Durrieu
2004;
Blayac
et al. 2004)
Methylphenidate x (Chung and Chiu xx (Rodnitzky x (Montastruc
1996) 2002) and
Durrieu
2004;
Blayac
et al. 2004)
6 Drug-Induced Movement Disorders in Elderly Patients
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas Tic
Pyridostigmine x (Nguyen et al.
2004)
Bethanechol x (Montastruc et al.
1994)
Opiates
Meperidine x (Jimenez-Jimenez x (Nguyen et al. x (van Harten
et al. 1997) 2004) et al. 1999;
Gareri et al.
2006)
Morphine xx (Montastruc
and Durrieu
2004)
Oxycodone x (Lang 1992)
Methadone x (Clark and x (Lang 1992) x (Jimenez-
Elliott 2001) Jimenez
et al. 1997)
Fentanyl x (Blayac et al. x (Petzinger
2004) et al. 1995)
Tramadol x (Montastruc
and Durrieu
2004)
S. Perez-Lloret et al.
Other drugs
Lithium xx (Bakheit 1997) xx (Mena and de x (Orti-Pareja xx (Morgan and x (Montastruc x (Jimenez-
Yebenes 2006; et al. 1999; Sethi 2005) and Durrieu Jimenez
Nguyen et al. Blayac et al. 2004) et al. 1997)
2004; Jimenez- 2004;
Jimenez et al. Jimenez-
1996) Jimenez
et al. 1996)
Tetrabenazine x (Sachdev 1995) xx (Mena and de x (Jimenez-
Yebenes 2006) Jimenez
et al. 1997;
Blayac et al.
2004)
Trihexyphenidyl x (Blayac et al. x (Rodnitzky
2004) 2002)
x uncommon/single report, xx common (frequency >10 %)
6 Drug-Induced Movement Disorders in Elderly Patients
101
Table 6.3 Movement disorders induced by nonpsychotropic drugs
102
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas
Antibiotics
Amphotericin B (Mott et al. 1995) x (Morgan and
Sethi 2005)
Penicillin xx (Montastruc
and Durrieu
2004)
Cephalosporins xx (Montastruc
and Durrieu
2004)
Chloroquine x (van Harten
et al. 1999)
Acyclovir x (Morgan and x (Jimenez-
Sethi 2005) Jimenez et al.
1997)
Vidarabine xx (Morgan and
Sethi 2005)
Co-trimoxazole x (Morgan and
Sethi 2005)
Foscarnet x (Dubow et al.
2008)
Asthma/allergies
Chlorpheniramine x (Montastruc xx (Nguyen et al. x (Lang 1992) x (Jimenez-
et al. 1994) 2004) Jimenez
et al. 1997)
Theophylline x (Jimenez-
Jimenez
et al. 1997)
S. Perez-Lloret et al.
B2-adrenergic xx (Rodnitzky xx (Morgan and
agonists 2002) Sethi 2005)
Antineoplastics
Cytosine arabinoside x (Nguyen et al. 2004) x (Morgan and
Sethi 2005)
Vincristine x (Nguyen et al. 2004) x (Morgan and
Sethi 2005)
Methotrexate x (Nguyen et al. 2004)
5-Fluorouracil x (Nguyen et al. 2004)
Doxorubicin x (Bower and
Muenter 1995)
Thalidomide x (Morgan and
Sethi 2005)
Ifosfamide x (Morgan and
Sethi 2005)
Interferon alfa xx (Morgan and x (Jimenez-
Sethi 2005) Jimenez et al.
1997)
6 Drug-Induced Movement Disorders in Elderly Patients
Cardiovascular disorders
Amiodarone xx (Mena and de x (Werner and xx (Morgan and x (Werner and
Yebenes 2006; Olanow Sethi 2005; Olanow 1989)
Nguyen et al. 1989) Montastruc
2004) and Durrieu
2004)
Procainamide xx (Morgan and
Sethi 2005)
(continued)
103
Table 6.3 (continued)
104
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas
Pindolol x (Morgan and
Sethi 2005)
Diltiazem/verapamil x (Dick and x (Mena and de
Barold 1989) Yebenes 2006;
Nguyen et al.
2004; Jimenez-
Jimenez et al.
1996; Dick and
Barold 1989)
Drugs for vertigo
Prochlorperazine/ xx (Mena and de
thiethylperazine Yebenes 2006)
Cinnarizine/ x (van Harten x (Orti-Pareja xx (Nguyen et al. x (Orti-Pareja
flunarizine et al. 1999) et al. 1999; 2004) et al.
Jimenez- 1999;
Jimenez et al. Jimenez-
1996) Jimenez
et al.
1996)
Endocrine disorders
Levothyroxine xx (Rodnitzky xx (Morgan and
2002) Sethi 2005;
Jimenez-
Jimenez
et al. 1997)
Calcitonin x (Morgan and
Sethi 2005)
S. Perez-Lloret et al.
Contraceptives x (Jimenez-Jimenez x (Shale and x (Rodnitzky
et al. 1997) Tanner 2002)
1996)
Corticosteroids xx (Rodnitzky
2002)
Veralipride x (Nguyen x (De Leo et al. 2006) x (De Leo x (De Leo et al. x (De Leo et al.
(menopause) et al. 2004) et al. 2006) 2006)
2006)
Hypoglycemics x (Ross 1990) x (Jimenez-
Jimenez
et al. 1997)
Anabolic steroids x (Shale and x (Jimenez-
Tanner Jimenez et al.
1996) 1997)
Reflux/gastric ulcers
Cimetidine x (Lang 1992) x (Morgan and x (Rodnitzky
Sethi 2005) 2002)
Ranitidine x (Lang 1992) x (Rodnitzky
6 Drug-Induced Movement Disorders in Elderly Patients
2002)
Misoprostol x (Morgan and
Sethi 2005)
Lansoprazole x (Angles et al. 2002)
Bismuth xx (Rodnitzky
2002)
(continued)
105
Table 6.3 (continued)
106
Drug-induced Tardive
Drug Acute dystonia Akathisia parkinsonism dyskinesias Tremors Myoclonus Other choreas
Other drugs
Cyclosporine/ x (Nguyen et al. 2004) xx (Morgan and
tacrolimus Sethi 2005;
Montastruc
and Durrieu
2004)
Naproxen sodium x (Montastruc et al.
1994)
x uncommon/single report, xx common (frequency >10 %)
S. Perez-Lloret et al.
6 Drug-Induced Movement Disorders in Elderly Patients 107
reactions is dopamine receptor blockage at the level of the striatum. DIMDs are less
frequently associated with the atypical antipsychotics, but dose-related movement
disorders occur with olanzapine, risperidone, and quetiapine at higher doses as well
(Gareri et al. 2006). Nonetheless, a recent intention-to-treat, secondary analysis of
data from an earlier randomized controlled trial failed to find the expected reduc-
tions of DIMD frequency for participants randomized to second-generation drugs
(Peluso et al. 2012).
Mechanisms underlying DIMDs are complex and not fully understood. They are
probably related to alterations within subcortical brain regions (e.g., basal ganglia
and thalamus) and do not involve the corticospinal pyramidal motor system.
Incidence of acute dystonia in treated patients varies from 2 to 64 % according
to different series, and pathogenesis remains unclear. All antipsychotics bind to D2
receptors; it has therefore been suggested that blockage of these receptors in the
caudate, putamen, and globus pallidus is partly responsible for causing acute dysto-
nia (Rupniak et al. 1986).
DIP is observed on average in about 4 % of patients taking neuroleptics (Noyes
et al. 2006; Rochon et al. 2005). DIP is caused by several mechanisms interfering
with the normal nigrostriatal dopamine neuron function. Generally, compounds
responsible for DIP block striatal dopamine D2 receptors, requiring blockade to
exceed 75 % in order to trigger DIP (Remington et al. 2006). Several recent reports
suggest certain neuroleptics produce DIP not only related to excessive blockade of
dopamine receptors but also mediated through direct and persistent toxic effects on
nigrostriatal dopamine neurons (Ulrich et al. 2005).
Akathisia affects about 28 % of patients on antipsychotics (Peluso et al. 2012).
The underlying pathophysiologic mechanism of akathisia is thought to be an imbal-
ance between cortical and nigrostriatal dopaminergic innervation, favoring increased
functional activity of the mesolimbic and nigrostriatal systems, in particular the
nucleus accumbens (Bakheit 1997).
The annual incidence of TD is about 5 % overall, including transient (3 %) and
persistent (2 %) TD (Tarsy and Baldessarini 2006). Risk of TD has proven to be
lower with atypical antipsychotics compared to classic ones. A recent review of
clinical trials on several modern antipsychotics vs. haloperidol in schizophrenia
showed that risperidone, olanzapine, quetiapine, amisulpride, or ziprasidone
induced TD in 2.1 % of cases, which is lower than the aforementioned annual TD
rate for classic antipsychotics. Indeed, among patients under 50, risk of new-onset
TD with haloperidol (5.4 %) was 6.8 times greater than atypical antipsychotics
(Correll et al. 2004). Conversely, TD incidence with modern drugs among patients
over 50 was similar to rates in younger individuals exposed to haloperidol, suggest-
ing an important agent-age interaction (Correll et al. 2004). The incidence of TD
varied remarkably little among modern agents except for increased risk with higher
doses of risperidone.
The pathophysiologic basis of TD remains speculative, but various neurochemi-
cal hypotheses have been proposed, including striatal dopaminergic hypersensitiv-
ity, basal ganglia cholinergic deficiency, dysfunctions of striatonigral γ-aminobutyric
acid (GABA)-mediated neurons, glutamate-induced excitotoxicity, and oxidative
108 S. Perez-Lloret et al.
stress (Casey 2004; Galili et al. 2000; Silvestri et al. 2000). It has been proposed that
antipsychotics result in increased dopamine turnover, followed by excess free radi-
cal production and subsequent damage to striatal GABAergic fibers, with reduced
inhibitory activity on motor circuits (Casey 2004; Galili et al. 2000; Silvestri et al.
2000). Concurrently, chronic blockade of dopamine receptors results in excessive
glutamate activity and resultant excitotoxicity. Likewise, chronic dopamine recep-
tor blockade results in receptor supersensitivity and persistent changes within basal
ganglia motor circuit.
Antidepressants
Other Drugs
Management
The first therapeutic measure is to withdraw the offending drug when possible. This
sole measure will probably suffice for most patients. Nonetheless, in some instances,
extra measures will need to be taken. A summary of such measures is offered in
Table 6.4.
In most instances, acute dystonia presents spontaneous resolution shortly after
drug withdrawal. Nonetheless, if treatment is needed, such as in the case of stridor,
antihistamines and/or benzodiazepines can be of help (van Harten et al. 1999;
Povlsen and Pakkenberg 1990). Intramuscular or intravenous administration of
muscarinic receptor blockers (biperiden 5 mg or procyclidine 5 mg) or antihista-
mines (promethazine 50 mg) is usually effective within 20 min. Occasionally, sec-
ond or third injections are necessary. After resolution, treatment with anticholinergics
should be continued for at least 24–48 h to prevent recurrence.
Anticholinergics have been commonly prescribed as a preventive treatment of
DIP in patients treated with antipsychotics without strong evidence to support this
use. Preliminary unconfirmed evidence supporting the use of vitamin E as a neuro-
protective has been published (Mena and de Yebenes 2006). Once DIP is diagnosed,
the best possible treatment is discontinuation of the causative drug (Mena and de
Yebenes 2006). In the majority of cases, however, it subsides gradually over a period
of weeks or months, but in some cases it may persist as long as a year or more
(Mena and de Yebenes 2006). Patients show global improvement soon after drug
withdrawal, and cognitive and mood disturbances subside more slowly, while
tremor may persist 18 months after drug discontinuation in a significant number of
patients. In those patients in whom parkinsonism becomes persistent and irrevers-
ible after drug withdrawal, diagnosis of latent idiopathic parkinsonism should be
considered. Hyposmia and dopaminergic loss as observed by neuroimaging tech-
niques are indicative of such subclinical disease (Kim et al. 2013b).
Propranolol, alprazolam, or antiserotonergic drugs have been reported to be
effective in the treatment of akathisia (Bakheit 1997; Amsterdam et al. 1994).
Lithium-induced akathisia was claimed to be particularly responsive to mianserin, a
noradrenaline and serotonin reuptake inhibitor. Although low serum iron is com-
monly associated with neuroleptic-induced tardive akathisia, the value of iron sup-
plements is doubtful and may even be harmful.
The best treatment for TD is prevention. To accomplish this, the lowest effec-
tive dose of antipsychotics should be identified for each patient and regularly
rechecked. After TD develops, drug withdrawal is followed by improvement from
0 to 92 % of patients, depending on the study (Casey 1990). This wide range is due
to multiple factors including patient variables (e.g., age), treatment variables (drug
dose and cumulative exposure), and temporal aspects (early diagnosis, duration of
treatment, and duration of TD follow-up). Age is consistently correlated with TD
improvement, with younger patients more likely to improve. It may take up to 5
years for complete remission to occur. Shorter neuroleptic exposure and age under
60 after onset of TD are correlated with greater likelihood of remission.
Dyskinesias, however, may reappear when antipsychotics are reinstituted. In a sig-
nificant number of patients, they may become irreversible despite antipsychotics
cessation.
Treating TD is a clinical challenge. Unfortunately, no drugs are uniformly safe
and effective over extended treatment periods. Reducing dopaminergic function is
the most effective way of suppressing (masking) TD (Jeste and Wyatt 1982). This
strategy is justified only in those rare cases when TD is severe, debilitating, or life-
threatening. Functional reduction of dopamine can be achieved with presynaptical
depletion (reserpine) or by false transmission (methyldopa). Tetrabenazine has been
successfully used for tardive dyskinesia, tardive tremor, and tardive tourettism treat-
ment (Jankovic 2009). Botulinum toxin can be used to treat tardive dystonia
(Jankovic 2009). Muscarinic receptor blockers should not be used as they may lower
the threshold for the appearance of tardive syndrome (Klawans and Rubovits 1974).
Conclusion
Movement disorders are common adverse drug reactions. They should be envi-
sioned when treatment with possible offending drugs is initiated. Off-label use of
movement disorder-inducing drugs or use beyond recommended doses should be
6 Drug-Induced Movement Disorders in Elderly Patients 111
References
Akagi H, Kumar TM. Lesson of the week: akathisia: overlooked at a cost. BMJ. 2002;324:
1506–7.
Amsterdam JD, Hornig-Rohan M, Maislin G. Efficacy of alprazolam in reducing fluoxetine-
induced jitteriness in patients with major depression. J Clin Psychiatry. 1994;55:394–400.
Andlin-Sobocki P, Jonsson B, Wittchen HU, Olesen J. Cost of disorders of the brain in Europe. Eur
J Neurol. 2005;12 Suppl 1:1–27.
Angles A, Bagheri H, Saivin S, Montastruc JL. Interaction between lansoprazole and bromocrip-
tine in a patient with Parkinson’s disease. Therapie. 2002;57:408–10.
Asconape J, Diedrich A, DellaBadia J. Myoclonus associated with the use of gabapentin. Epilepsia.
2000;41:479–81.
Bakheit A. The syndrome of motor restlessness–a treatable but under-recognised disorder. Postgrad
Med J. 1997;73:529–30.
Barbosa MT, Caramelli P, Maia DP, et al. Parkinsonism and Parkinson’s disease in the elderly: a
community-based survey in Brazil (the Bambui study). Mov Disord. 2006;21:800–8.
Barbosa MT, Caramelli P, Cunningham MC, et al. Prevalence and clinical classification of tremor
in elderly-A community-based survey in Brazil. Mov Disord. 2013;28(5):640–6.
Benito-Leon J, Bermejo-Pareja F, Morales-Gonzalez JM, et al. Incidence of Parkinson disease and
parkinsonism in three elderly populations of central Spain. Neurology. 2004;62:734–41.
Blayac JP, Pinzani V, Peyriere H, Hillaire-Buys D. Drug-induced movement disorders: tardive
syndromes. Therapie. 2004;59:113–9.
Bondon-Guitton E, Perez-Lloret S, Bagheri H, et al. Drug-induced parkinsonism: a review of 17
years’ experience in a regional pharmacovigilance center in France. Mov Disord. 2011;26:
2226–31.
Bonuccelli U, Nocchiero A, Napolitano A, et al. Domperidone-induced acute dystonia and poly-
cystic ovary syndrome. Mov Disord. 1991;6:79–81.
Bovi T, Antonini A, Ottaviani S, et al. The status of olfactory function and the striatal dopaminer-
gic system in drug-induced parkinsonism. J Neurol. 2010;257:1882–9.
Bower JH, Muenter MD. Temporary worsening of parkinsonism in a patient with Parkinson’s
disease after treatment with paclitaxel for a metastatic grade IV adenocarcinoma. Mov Disord.
1995;10:681–2.
Brooks DE. Propofol-induced movement disorders. Ann Emerg Med. 2008;51:111–2.
Burke RE, Fahn S, Jankovic J, et al. Tardive dystonia: late-onset and persistent dystonia caused by
antipsychotic drugs. Neurology. 1982;32:1335–46.
Caligiuri MR, Jeste DV, Lacro JP. Antipsychotic-induced movement disorders in the elderly: epi-
demiology and treatment recommendations. Drugs Aging. 2000;17:363–84.
Casey DE. Spontaneous and tardive dyskinesias: clinical and laboratory studies. J Clin Psychiatry.
1985;46:42–7.
Casey DE. Tardive dyskinesia. West J Med. 1990;153:535–41.
112 S. Perez-Lloret et al.
Casey DE. Neuroleptic-induced acute dystonia. In: Lang AE, Weiner WJ, editors. Drug-induced
movement disorders. 1st ed. Mount Kisco: Futura; 1992. p. 21–40.
Casey DE. Pathophysiology of antipsychotic drug-induced movement disorders. J Clin Psychiatry.
2004;65 Suppl 9:25–8.
Chouza C, Scaramelli A, Caamano JL, et al. Parkinsonism, tardive dyskinesia, akathisia, and
depression induced by flunarizine. Lancet. 1986;1:1303–4.
Chung WS, Chiu HP. Drug-induced akathisia revisited. Br J Clin Pract. 1996;50:270–8.
Clark JD, Elliott J. A case of a methadone-induced movement disorder. Clin J Pain.
2001;17:375–7.
Correll CU, Leucht S, Kane JM. Lower risk for tardive dyskinesia associated with second-
generation antipsychotics: a systematic review of 1-year studies. Am J Psychiatry. 2004;
161:414–25.
Das SK, Banerjee TK, Biswas A, et al. Community survey of primary dystonia in the city of
Kolkata, India. Mov Disord. 2007;22:2031–6.
Davies J, Tongroach P. Neuropharmacological studies on the nigro-striatal and raphe-striatal sys-
tem in the rat. Eur J Pharmacol. 1978;51:91–100.
De Leo V, Morgante G, Musacchio MC, et al. The safety of veralipride. Expert Opin Drug Saf.
2006;5:695–701.
de Rijk MC, Tzourio C, Breteler MM, et al. Prevalence of parkinsonism and Parkinson’s disease in
Europe: the EUROPARKINSON Collaborative Study. European Community Concerted Action
on the Epidemiology of Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1997;62:10–5.
Di Mascio M, Di Giovanni G, Di Matteo V, et al. Selective serotonin reuptake inhibitors reduce the
spontaneous activity of dopaminergic neurons in the ventral tegmental area. Brain Res Bull.
1998;46:547–54.
Dick RS, Barold SS. Diltiazem-induced parkinsonism. Am J Med. 1989;87:95–6.
Dray A, Davies J, Oakley NR, et al. The dorsal and medial raphe projections to the substantia nigra
in the rat: electrophysiological, biochemical and behavioural observations. Brain Res.
1978;151:431–42.
Dubow JS, Panush SR, Rezak M, Leikin J. Acute dystonic reaction associated with foscarnet
administration. Am J Ther. 2008;15:184–6.
Edwards IR, Aronson JK. Adverse drug reactions: definitions, diagnosis, and management. Lancet.
2000;356:1255–9.
Esper CD, Factor SA. Failure of recognition of drug-induced parkinsonism in the elderly. Mov
Disord. 2008;23:401–4.
Galili R, Mosberg, Gil-Ad I, et al. Haloperidol-induced neurotoxicity – possible implications for
tardive dyskinesia. J Neural Transm. 2000;107:479–90.
Garcia-Ruiz PJ, de Garcia YJ, Jimenez-Jimenez FJ, et al. Parkinsonism associated with calcium
channel blockers: a prospective follow-up study. Clin Neuropharmacol. 1992;15:19–26.
Gareri P, De Fazio P, De Fazio S, et al. Adverse effects of atypical antipsychotics in the elderly: a
review. Drugs Aging. 2006;23:937–56.
Gershanik OS. Drug-induced parkinsonism in the aged. Recognition and prevention. Drugs Aging.
1994;5:127–32.
Haddad PM, Dursun SM. Neurological complications of psychiatric drugs: clinical features and
management. Hum Psychopharmacol. 2008;23 Suppl 1:15–26.
Jankovic J. Tardive syndromes and other drug-induced movement disorders. Clin Neuropharmacol.
1995;18:197–214.
Jankovic J. Treatment of hyperkinetic movement disorders. Lancet Neurol. 2009;8:844–56.
Jeste DV, Wyatt RJ. Therapeutic strategies against tardive dyskinesia. Two decades of experience.
Arch Gen Psychiatry. 1982;39:803–16.
Jimenez-Jimenez FJ, Molina JA. Extrapyramidal symptoms associated with Selective Serotonin
Reuptake Inhibitors. CNS Drugs. 2000;14:367–79.
Jimenez-Jimenez FJ, Orti-Pareja M, Yuso-Peralta L, et al. Drug-induced parkinsonism in a move-
ment disorders unit: a four-year survey. Parkinsonism Relat Disord. 1996;2:145–9.
6 Drug-Induced Movement Disorders in Elderly Patients 113
Jimenez-Jimenez FJ, Garcia-Ruiz PJ, Molina JA. Drug-induced movement disorders. Drug Saf.
1997;16:180–204.
Kenney C, Hunter C, Davidson A, Jankovic J. Metoclopramide, an increasingly recognized cause
of tardive dyskinesia. J Clin Pharmacol. 2008;48:379–84.
Khiat A, Kuznetsov Y, Blanchet PJ, Boulanger Y. Diffusion-weighted imaging and magnetization
transfer imaging of tardive and edentulous orodyskinesia. Mov Disord. 2008;23:1281–5.
Kim JH, Byun HJ. Non-motor cognitive-perceptual dysfunction associated with drug-induced par-
kinsonism. Hum Psychopharmacol. 2009;24:129–33.
Kim YD, Kim JS, Chung SW, et al. Cognitive dysfunction in drug induced parkinsonism (DIP).
Arch Gerontol Geriatr. 2011;53:e222–6.
Kim JS, Youn J, Shin H, Cho JW. Nonmotor symptoms in drug-induced parkinsonism and drug-
naive Parkinson disease. Can J Neurol Sci. 2013a;40:36–41.
Kim JS, Oh YS, Kim YI, et al. Combined use of (1)(2)(3)I-metaiodobenzylguanidine (MIBG)
scintigraphy and dopamine transporter (DAT) positron emission tomography (PET) predicts
prognosis in drug-induced Parkinsonism (DIP): a 2-year follow-up study. Arch Gerontol
Geriatr. 2013b;56:124–8.
Kinon BJ, Gilmore JA, Liu H, Halbreich UM. Prevalence of hyperprolactinemia in schizophrenic
patients treated with conventional antipsychotic medications or risperidone.
Psychoneuroendocrinology. 2003;28 Suppl 2:55–68.
Klawans HL, Rubovits R. Effect of cholinergic and anticholinergic agents on tardive dyskinesia.
J Neurol Neurosurg Psychiatry. 1974;37:941–7.
Lang AE. Miscellaneous drug-induced, movement disorders. In: Lang AE, Weiner WJ, editors.
Drug-induced movement disorders. 1st ed. New York: Futura Publishing; 1992.
p. 339–81.
Lang AE, Lozano AM. Parkinson’s disease. First of two parts. N Engl J Med. 1998;339:1044–53.
Lee PH, Yeo SH, Yong SW, Kim YJ. Odour identification test and its relation to cardiac
123I-metaiodobenzylguanidine in patients with drug induced parkinsonism. J Neurol
Neurosurg Psychiatry. 2007;78:1250–2.
LeWitt PA, Walters A, Hening W, McHale D. Persistent movement disorders induced by buspi-
rone. Mov Disord. 1993;8:331–4.
Lopez-Sendon J, Mena MA, de Yebenes JG. Drug-induced parkinsonism. Expert Opin Drug Saf.
2013;12(4):487–96.
Masmoudi K, Gras-Champel V, Masson H, Andrejak M. Parkinsonism and/or cognitive impair-
ment with valproic acid therapy: a report of ten cases. Pharmacopsychiatry. 2006;
39:9–12.
Masmoudi K, Masson H, Gras V, Andrejak M. Extrapyramidal adverse drug reactions associated
with trimetazidine: a series of 21 cases. Fundam Clin Pharmacol. 2012;26:198–203.
Mena MA, de Yebenes JG. Drug-induced parkinsonism. Expert Opin Drug Saf. 2006;5:759–71.
Merello M, Starkstein S, Petracca G, et al. Drug-induced parkinsonism in schizophrenic patients:
motor response and psychiatric changes after acute challenge with L-Dopa and apomorphine.
Clin Neuropharmacol. 1996;19:439–43.
Merello M, Nouzeilles MI, Arce GP, Leiguarda R. Accuracy of acute levodopa challenge for clini-
cal prediction of sustained long-term levodopa response as a major criterion for idiopathic
Parkinson’s disease diagnosis. Mov Disord. 2002;17:795–8.
Montagna P, Gabellini AS, Monari L, Lugaresi E. Parkinsonian syndrome after long-term treat-
ment with clebopride. Mov Disord. 1992;7:89–90.
Montastruc JL, Durrieu G. Drug-induced tremor and acute movement disorders. Therapie.
2004;59:97–103.
Montastruc JL, Llau ME, Rascol O, Senard JM. Drug-induced parkinsonism: a review. Fundam
Clin Pharmacol. 1994;8:293–306.
Montastruc JL, Sommet A, Lacroix I, et al. Pharmacovigilance for evaluating adverse drug reac-
tions: value, organization, and methods. Joint Bone Spine. 2006;73:629–32.
Morgan JC, Sethi KD. Drug-induced tremors. Lancet Neurol. 2005;4:866–76.
114 S. Perez-Lloret et al.
Mott SH, Packer RJ, Vezina LG, et al. Encephalopathy with parkinsonian features in children fol-
lowing bone marrow transplantations and high-dose amphotericin B. Ann Neurol.
1995;37:810–4.
Nguyen N, Pradel V, Micallef J, et al. Drug-induced parkinson syndromes. Therapie. 2004;
59:105–12.
Nouzeilles M, Garcia M, Rabinowicz A, Merello M. Prospective evaluation of parkinsonism and
tremor in patients treated with valproate. Parkinsonism Relat Disord. 1999;5:67–8.
Noyes K, Liu H, Holloway RG. What is the risk of developing parkinsonism following neuroleptic
use? Neurology. 2006;66:941–3.
Olanow CW, Damier P, Goetz CG, et al. Multicenter, open-label, trial of sarizotan in Parkinson
disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin
Neuropharmacol. 2004;27:58–62.
Orti-Pareja M, Jimenez-Jimenez FJ, Vazquez A, et al. Drug-induced tardive syndromes.
Parkinsonism Relat Disord. 1999;5:59–65.
Oterino A, Pascual J. Sumatriptan-induced axial dystonia in a patient with cluster headache.
Cephalalgia. 1998;18:360–1.
Pasricha PJ, Pehlivanov N, Sugumar A, Jankovic J. Drug insight: from disturbed motility to disor-
dered movement–a review of the clinical benefits and medicolegal risks of metoclopramide.
Nat Clin Pract Gastroenterol Hepatol. 2006;3:138–48.
Paulson GW. Historical comments on tardive dyskinesia: a neurologist’s perspective. J Clin
Psychiatry. 2005;66:260–4.
Peluso MJ, Lewis SW, Barnes TR, Jones PB. Extrapyramidal motor side-effects of first- and
second-generation antipsychotic drugs. Br J Psychiatry. 2012;200:387–92.
Perez F, Helmer C, Dartigues JF, et al. A 15-year population-based cohort study of the incidence
of Parkinson’s disease and dementia with Lewy bodies in an elderly French cohort. J Neurol
Neurosurg Psychiatry. 2010;81:742–6.
Perez-Lloret S, Amaya M, Merello M. Pregabalin-induced parkinsonism: a case report. Clin
Neuropharmacol. 2009;32(6):353–4.
Petzinger G, Mayer SA, Przedborski S. Fentanyl-induced dyskinesias. Mov Disord.
1995;10:679–80.
Poewe W. The natural history of Parkinson’s disease. J Neurol. 2006;253 Suppl 7:VII2–6.
Povlsen UJ, Pakkenberg H. Effect of intravenous injection of biperiden and clonazepam in dysto-
nia. Mov Disord. 1990;5:27–31.
Pringsheim T, Wiltshire K, Day L, et al. The incidence and prevalence of Huntington’s disease: a
systematic review and meta-analysis. Mov Disord. 2012;27:1083–91.
Reeves AL, So EL, Sharbrough FW, Krahn LE. Movement disorders associated with the use of
gabapentin. Epilepsia. 1996;37:988–90.
Rehan HS, Chopra D, Kakkar AK. Physician’s guide to pharmacovigilance: terminology and cau-
sality assessment. Eur J Intern Med. 2009;20:3–8.
Remington G, Mamo D, Labelle A, et al. A PET study evaluating dopamine D2 receptor occu-
pancy for long-acting injectable risperidone. Am J Psychiatry. 2006;163:396–401.
Rochon PA, Stukel TA, Sykora K, et al. Atypical antipsychotics and parkinsonism. Arch Intern
Med. 2005;165:1882–8.
Rodnitzky RL. Drug-induced movement disorders. Clin Neuropharmacol. 2002;25:142–52.
Ross RT. Drug-induced parkinsonism and other movement disorders. Can J Neurol Sci.
1990;17:155–62.
Rupniak NM, Jenner P, Marsden CD. Acute dystonia induced by neuroleptic drugs.
Psychopharmacology (Berl). 1986;88:403–19.
Sachdev P. The epidemiology of drug-induced akathisia: part I. Acute akathisia. Schizophr Bull.
1995;21:431–49.
Seijo-Martinez M, del Castro RM, Rodriguez AJ, et al. Prevalence of parkinsonism and Parkinson’s
disease in the Arosa Island (Spain): a community-based door-to-door survey. J Neurol Sci.
2011;304:49–54.
6 Drug-Induced Movement Disorders in Elderly Patients 115
Abbreviations
AD Alzheimer’s disease
DLB Dementia with Lewy bodies
ESRS Extrapyramidal Symptom Rating Scale
FAST Functional Assessment Staging
ICARS International Cerebellar Ataxia Rating Scale
MDS-UPDRS Movement Disorder Society-sponsored version of the UPDRS
MSA Multiple system atrophy
NNIPPS-PPS Natural History and Neuroprotection in Parkinson Plus Syndromes
– Parkinson Plus Scale
PD Parkinson’s disease
PDD Dementia associated with Parkinson’s disease
PEPS Pyramidal and Extrapyramidal Scale
PIGD Postural instability gait difficulty
POS-PP Palliative Care Outcome Scale-Parkinsonism Plus
PSP Progressive supranuclear palsy
PSPRS Progressive Supranuclear Palsy Rating Scale
RSGE-CD Rating Scale for Gait Evaluation in Cognitive Deterioration
SCOPA Scales for Outcomes in Parkinson’s Disease
UMSARS Unified Multiple System Atrophy Rating Scale
UPDRS Unified Parkinson’s Disease Rating Scale
Table 7.1 Examples of conditions that can associate simultaneously dementia and parkinsonism
Neurodegenerative Other
Parkinson’s disease Cerebrovascular disease
Dementia with Lewy bodies Normal-pressure hydrocephalus
Corticobasal degeneration Creutzfeldt-Jakob disease
Multiple system atrophy HIV-associated dementia
Progressive supranuclear palsy Traumatic brain injury
Huntington’s disease Alcoholic dementia
Alzheimer’s disease
Frontotemporal dementia
PD patients at an early stage of disease (Reid et al. 1989; Foltynie et al. 2004), and it
has been suggested that the possible onset of PDD might be predictable by measuring
if PD patients suffer from early cognitive impairment (Pedersen et al. 2013). PD
patients with postural instability gait difficulty (PIGD) are at higher risk to get demen-
tia and also have a faster rate of cognitive decline (Burn et al. 2006; Alves et al. 2006).
Furthermore, a study by McKeith et al. (2006) showed that extrapyramidal motor
symptoms are more impaired (McKeith et al. 2006) and are associated to more rapid
noncognitive disease progression (Williams et al. 2006) in DLB than in AD.
In addition, it has been reported that in patients with AD and parkinsonism, psycho-
sis appears more frequently than in patients without AD and parkinsonism (Zubenko
et al. 1991; Mayeux et al. 1985). By analyzing the parkinsonian signs in AD, it may be
possible to predict the risk to develop a psychosis (Caligiuri et al. 2003).
In Table 7.1, we list the common conditions that can be associated with dementia
and parkinsonism. This list is not exhaustive as many rare metabolic and genetic
conditions can cause dementia with a degree of parkinsonism, but in this chapter we
will not be able to cover these conditions.
We have attempted to subdivide these conditions to neurodegenerative and other
disorders (see Table 7.1).
Scales and questionnaires are commonly used to determine the type of manifesta-
tions of the underlying condition, to measure their severity and frequency, and to
establish their relationships with demographic, historical, functional, and psycho-
social factors. They are needed, for example, to determine the patients’ current
health state and its course over time, to compare groups of patients, to inform the
outcomes of clinical practices and trials, to file and share information, to assign
resources, and to make decisions in Health Policy. For multicenter and interna-
tional collaborations, it is important to harmonize measurements in order to obtain
homogeneous results. A survey by Ramirez Diaz et al. (2005), for example,
emphasized the need of unified tools and evaluation criteria in order to get robust
data collection.
120 C. Rodriguez-Blazquez et al.
In general, scales can be divided into generic, which means that they are usable in
any condition, or specific, meaning that they can just be used for one condition in par-
ticular. Furthermore, scales can be classified in several ways: self-completed or rater-
completed; single-item, multi-item, or composite; unidimensional or multidimensional;
and disease-centered and patient-centered measures. Only a few of the above condi-
tions (Table 7.1) have validated scales including specific assessment of parkinsonism.
In order to review scales for selection of the most appropriate, for a research
study or clinical practice, for instance, some aspects have to be considered. The
most relevant are shown in the Table 7.2 that includes the most commonly consid-
ered and relevant points for description and clinimetric information of a scale. For
example, the systematic reviews on scales performed by the Movement Disorder
Society Task Force have been based on a similar scheme (www.movementdisorders.
org/publications/ebm_reviews/).
Concerning patients with dementia, it has to be considered that they are usually
not fully capable to adequately complete self-assessments or respond to interviews.
That is the reason why, in the setting of dementia, this kind of evaluations needing
the patients’ inference cannot be used or have to be completed by a proxy (care-
giver, health professional), a situation challenging the reliability and validity of the
assessment. However, the rating scales used for evaluation of parkinsonism are rater
based and completed by a health professional who performs the motor examination.
Patient’s collaboration for doing some maneuvers (e.g., tapping fingers or hand,
rapid alternative movements, walking a distance, etc.) is necessary, but usually there
is a response option (e.g., unable to do, cannot perform the task, unable to test, etc.)
adequate for the situation lacking of patient’s collaboration. Therefore, data quality
is usually satisfactory for this kind of assessment.
A brief review of the scales useful to evaluate parkinsonism in patients with
dementia ensues.
In 1987 (Fahn and Elton 1987) the Unified Parkinson’s Disease Rating Scale
(UPDRS) was introduced, which has now become one of the most used scales in
Parkinson’s disease (Ramaker et al. 2002). This scale takes about 10–20 min to
complete and is available from the original publications. It is a public domain scale
that evaluates impairment and disability for basic activities of daily life (Movement
Disorder Society Task Force 2003). It contains 42 items, which are separated into
four domains: (1) mentation, behavior, and mood; (2) activities of daily living; (3)
motor items, which are relevant in the context of the present chapter; and (4) com-
plications. In addition, it covers a modified Hoehn and Yahr Staging (1967) and
the Schwab and England Scale. Besides the importance for this chapter, section
of the “UPDRS” is completed by a health professional and focused on the clini-
cal examination of motor impairment characteristic of parkinsonism, needing not
the input of patients themselves. Concerning the psychometric properties in PD,
7 Scales for Measuring Parkinsonism in Demented Patients 121
internal consistency, inter-rater and test-retest reliability, and construct validity are
satisfactory as a whole (Martinez-Martin and Forjaz 2006), and the scale is sensitive
to changes. The scale has been translated to several languages and has been widely
used in different studies and settings (Movement Disorder Society Task Force
2003). However, some ambiguities in the written text, as well as lack of adequate
instructions for the raters and the missing of many non-motor aspects, promoted the
revision of the scale to a new version (MDS-UPDRS) free of these shortcomings
(Movement Disorders Society Task Force 2003).
Cubo et al. (2000) and Kroonenberg et al. (2006) have reported that the motor
examination of the UPDRS can also be used for patients with progressive
122 C. Rodriguez-Blazquez et al.
supranuclear palsy (PSP). Section III, motor examination, has been used to assess
the motor impairment in multiple system atrophy (MSA) (Seppi et al. 2005) show-
ing similar clinimetric properties in MSA than in PD (Tison et al. 2002a) and a high
correlation with the International Cerebellar Ataxia Rating Scale (ICARS) in MSA
patients (Tison et al. 2002b). UPDRS motor scores have been used to assess extra-
pyramidal symptoms in AD (Ellis et al. 1996; Caligiuri et al. 2003) and to distin-
guish between these DLB patients and AD patients (Kaur et al. 2013) and are
sensitive to changes due to treatment with cholinesterase inhibitors in PDD (Rolinski
et al. 2012).
Modified versions of the UPDRS motor section have been used to characterize
AD patients (Wilson et al. 2000; Park et al. 2006) and to predict the risk of develop-
ing dementia in older people (Louis et al. 2010).
The Progressive Supranuclear Palsy Rating Scale (PSPRS) (Golbe and Ohman-
Strickland 2007) is a useful and practical tool that can be used as a routine measure-
ment for assessment of PSP patients’ disability and disease progression and also may
be used as variable for clinical trials and indicator of prognosis. It takes about 10 min
to fulfill the 28 items divided into 6 main groups. These groups contain daily activities
(by history), behavior, bulbar, ocular motor, limb motor, and gait/midline. Whereas 6
of the total of 28 items are rated on a 3-point scale (0–2), the missing 22 items are
rated on a 5-point scale (0–4). A maximum of 100 points can be obtained. The scale
has an excellent overall reliability, moderate convergent validity, and predictive valid-
ity in relation to subsequent survival (Golbe and Ohman-Strickland 2007).
The Pyramidal and Extrapyramidal Scale (PEPS) especially detects motor symp-
toms in patients with dementia or mild cognitive impairment because of the small
vessel disease and has been developed and validated by Kim et al. (2011a). The
124 C. Rodriguez-Blazquez et al.
scale can be particularly used to follow the disease process. The scale consists of 34
items, which are subdivided into five domains: (1) corticospinal (6/60), (2) corti-
cobulbar (9/60) tract symptoms and signs, (3) extrapyramidal symptoms/signs
(30/60), (4) gait abnormalities (9/60), and (5) gait severity (6/60). The patient can
reach a maximum of 60 points.
The test-retest and the inter-rater reliability coefficients were satisfactory. In
addition, the motor signs scores as measured by the UPDRS were significantly cor-
related with the total and extrapyramidal signs scores from the PEPS (Kim et al.
2011a). Moreover, the motor deficits as assessed by the PEPS correlated with supra-
and infratentorial lesions identified by magnetic resonance imaging (Kim et al.
2011b). The scale has not been used beyond the original authors.
This scale has been especially developed to measure disease severity and progression
in MSA and PSP in order to predict the survival and as a sensitive instrument for clini-
cal trials (Payan et al. 2011). The scale comprises 83 items, summarized into 15
dimensional sub-scores and a total score with a maximum of 309 points. This scale
shows a very appropriate content as well as a good convergent validity. Furthermore,
the internal consistency of the total score and the inter-rater reliability have been found
excellent (Payan et al. 2011). The scale is able to predict survival and is responsive to
changes, although PSP patients show higher progression rates than MSA patients.
Nonetheless, the scale has not been tested beyond the original validation study.
Other Scales
Other PD rating scales that have been used to assess extrapyramidal motor signs in
dementia patients (Ellis et al. 1996) are the Webster scale (Webster 1968) and the
Columbia University Parkinson’s Disease Rating Scale (Yahr et al. 1969). However,
7 Scales for Measuring Parkinsonism in Demented Patients 125
Conclusion
There is a variety of scales assessing the signs that characterize the parkinsonian
syndrome. Most of these scales have been designed to evaluate disorders in which
the motor aspects are relevant and frequently the most typical aspect of the disease
and have not been properly validated in populations experiencing dementia as pre-
dominant feature. However, management and clinical assessment of parkinsonism
is important for a complete evaluation of a patient with dementia and parkinsonism.
As such, development of specific scales which can be reliably and reproducibly
applied to demented parkinsonian patients remains a key unmet need. Further large-
scale studies are required to develop these tools in an international framework.
References
Caligiuri MP, Peavy G, Salmon DP, Galasko DR, Thal LJ. Neuromotor abnormalities and risk for
psychosis in Alzheimer’s disease. Neurology. 2003;61:954–8.
Chouinard G, Margolese HC. Manual for the Extrapyramidal Symptom Rating Scale (ESRS).
Schizophr Res. 2005;76:247–65.
Cubo E, Stebbins GT, Golbe LI, Nieves AV, Leurgans S, Goetz CG, et al. Application of the unified
Parkinson’s disease rating scale in progressive supranuclear palsy: factor analysis of the motor
scale. Mov Disord. 2000;15:276–9.
De Deyn PP, Wirshing WC. Scales to assess efficacy and safety of pharmacologic agents in the
treatment of behavioral and psychological symptoms of dementia. J Clin Psychiatry. 2001;62
Suppl 21:19–22.
Dubois B, Pillon B, Sternic N, Lhermitte F, Agid Y. Age induced cognitive disturbances in
Parkinson’s disease. Neurology. 1990;40:38–41.
Ellis RJ, Caligiuri M, Galasko D, Thal LJ. Extrapyramidal motor signs in clinically diagnosed
Alzheimer disease. Alzheimer Dis Assoc Disord. 1996;10:103–14.
Fahn S, Elton RL. UPDRS program members. Unified Parkinson’s disease rating scale. In: Fahn S,
Marsden CD, Goldstein M, Calne DB, editors. Recent developments in Parkinson’s disease,
vol. 2. Florham Park: Macmillan Healthcare Information; 1987. p. 153–64.
Foltynie T, Brayne CEG, Robbins TW, Barker RA. The cognitive ability of an incident cohort of
Parkinson’s patients in the UK. The CamPaIGN study. Brain. 2004;127:550–60.
Geser F, Wenning GK, Seppi K, Stampfer-Kountchev M, Scherfler C, Sawires M, et al. Progression
of multiple system atrophy (MSA): a prospective natural history study by the european MSA
Study Group (EMSA SG). Mov Disord. 2006;21:179–86.
Goetz CG, Fahn S, Martinez-Martin P, Poewe W, Sampaio C, Stebbins GT, et al. Movement
Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-
UPDRS): process, format, and clinimetric testing plan. Mov Disord. 2007;22:41–7.
Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, et al. Movement
Disorder Society-Sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-
UPDRS): scale presentation and clinimetric testing results. Mov Disord. 2008;23:2129–70.
Goetz CG, Stebbins GT, Chmura TA, Fahn S, Poewe W, Tanner CM. Teaching program for the
Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating
Scale: (MDS-UPDRS). Mov Disord. 2010;25:1190–4.
Goetz CG, Stebbins GT, LaPelle N, Huang J, Tilley BC. MDS-UPDRS non-English translation
program. Mov Disord. 2012;27 Suppl 1:S96.
Golbe LI, Ohman-Strickland PA. A clinical rating scale for progressive supranuclear palsy. Brain.
2007;130:1552–65.
Hearn J, Higginson IJ. Development and validation of a core outcome measure for palliative care:
the palliative care outcome scale. Palliative Care Core Audit Project Advisory Group. Qual
Health Care. 1999;8:219–27.
Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG. The Sydney multicenter study of
Parkinson’s disease: the inevitability of dementia at 20 years. Mov Disord. 2008;23:837–44.
Higginson IJ, Gao W, Saleem TZ, Chaudhuri KR, Burman R, McCrone P, et al. Symptoms and
quality of life in late stage Parkinson syndromes: a longitudinal community study of predictive
factors. PLoS One. 2012;7:e46327.
Hoehn MM, Yahr MD. Parkinsonism: onset, progression and mortality. Neurology. 1967;17:427–42.
Hughes TA, Ross HF, Musa S, Bhattacherjee S, Nathan RN, Mindham RHS, et al. A 10-year study
of the incidence of and factors predicting dementia in Parkinson’s disease. Neurology.
2000;54:1596–602.
Kaur B, Harvey DJ, Decarli CS, Zhang L, Sabbagh MN, Olichney JM. Extrapyramidal signs by
dementia severity in Alzheimer disease and dementia with Lewy bodies. Alzheimer Dis Assoc
Disord. 2013;27(3):226–32. doi:10.1097/WAD.0b013e31826f040d.
Kim SH, Seo SW, Go SM, Chin J, Lee BH, Lee JH, et al. Pyramidal and extrapyramidal scale
(PEPS): a new scale for the assessment of motor impairment in vascular cognitive impairment
associated with small vessel disease. Nab Clin Neurol Neurosurg. 2011a;113:181–7.
7 Scales for Measuring Parkinsonism in Demented Patients 127
Kim SH, Park JS, Ahn HJ, Seo SW, Lee JM, Kim ST, et al. Voxel-based analysis of diffusion tensor
imaging in patients with subcortical vascular cognitive impairment: correlates with cognitive
and motor deficits. J Neuroimaging. 2011b;21:317–24.
Knol W, Keijsers CJ, Jansen PA, van Marum RJ. Systematic evaluation of rating scales for drug-
induced parkinsonism and recommendations for future research. J Clin Psychopharmacol.
2010;30:57–63.
Krismer F, Seppi K, Tison F, Sampaio C, Zangerl A, Peralta C, et al. The unified multiple system
atrophy rating scale: intrarater reliability. Mov Disord. 2012;27:1683–5.
Kroonenberg PM, Oort FJ, Stebbins GT, Leurgans SE, Cubo E, Goetz CG. Motor function in
Parkinson’s disease and supranuclear palsy: simultaneous factor analysis of a clinical scale in
several populations. BMC Med Res Methodol. 2006;6:26.
Louis ED, Tang MX, Schupf N. Mild parkinsonian signs are associated with increased risk of
dementia in a prospective, population-based study of elders. Mov Disord. 2010;25:172–8.
Marinus J, Visser M, Stiggelbout AM, Martin Rabey J, Martinez-Martin P, Bonuccelli U, et al. A
short scale for the assessment of motor impairments and disabilities in Parkinson’s disease: the
SPES/SCOPA. J Neurol Neurosurg Psychiatry. 2004;75:388–95.
Martinez-Martin P, Forjaz MJ. Metric attributes of the unified Parkinson’s disease rating scale 3.0
battery: part I, feasibility, scaling assumptions, reliability, and precision. Mov Disord.
2006;21:1182–8.
Martinez-Martin P, Benito-Leon J, Burguera JA, Castro A, Linazasoro G, Martinez-Castrillo JC,
et al. The SCOPA-motor scale for assessment of Parkinson’s disease is a consistent and valid
measure. J Clin Epidemiol. 2005;58:674–9.
Martinez-Martin P, Osa Ruiz E, Gómez-Conesa A, Olazarán J, The RSGE-CD Validation Group.
A Rating Scale for Gait Evaluation in Cognitive Deterioration (RSGE-CD): validation study.
J Alzheimers Dis. 2012;31:543–53.
Martinez-Martin P, Rodriguez-Blazquez C, Alvarez-Sanchez M, Arakaki T, Bergareche-Yarza A,
Chade A, et al. Expanded and independent validation of the Movement Disorder Society-
Unified Parkinson’s Disease Rating Scale (MDS-UPDRS). J Neurol. 2013;260:228–36.
Mayeux R, Stern Y, Spanton S. Heterogeneity in dementia of the Alzheimer type: evidence of
subgroups. Neurology. 1985;35:453–61.
McKeith IG, Rowan E, Askew K, Naidu A, Allan L, Barnett N, et al. More severe functional
impairment in dementia with Lewy bodies than Alzheimer disease is related to extrapyramidal
motor dysfunction. Am J Geriatr Psychiatry. 2006;14:582–8.
Merello M, Gerschcovich ER, Ballesteros D, Cerquetti D. Correlation between the Movement
Disorders Society Unified Parkinson’s Disease rating scale (MDS-UPDRS) and the Unified
Parkinson’s Disease rating scale (UPDRS) during l-dopa acute challenge. Parkinsonism Relat
Disord. 2011;17:705–7.
Movement Disorder Society Task Force on Rating Scales for Parkinson’s Disease. The Unified
Parkinson’s Disease Rating Scale (UPDRS): status and recommendations. Mov Disord.
2003;18:738–50.
Müller ML, Frey KA, Petrou M, Kotagal V, Koeppe RA, Albin RL, et al. β-amyloid and postural
instability and gait difficulty in Parkinson’s disease at risk for dementia. Mov Disord.
2013;28:296–301.
Novak P, Williams A, Ravin P, Zurkiya O, Abduljalil A, Novak V. Treatment of multiple system
atrophy using intravenous immunoglobulin. BMC Neurol. 2012;12:131.
Pal G, Goetz CG. Assessing bradykinesia in Parkinsonian disorders. Front Neurol. 2013;4:54.
Park M, Comella CL, Leurgans SE, Fan W, Wilson RS, Bennett DA. Association of daytime nap-
ping and Parkinsonian signs in Alzheimer’s disease. Sleep Med. 2006;7:614–8.
Payan CA, Viallet F, Landwehrmeyer BG, Bonnet AM, Borg M, Durif F, et al. Disease severity and
progression in progressive supranuclear palsy and multiple system atrophy: validation of the
NNIPPS-Parkinson Plus Scale. PLoS One. 2011;6:e22293.
Pedersen KF, Larsen JP, Tysnes OB, Alves G. Prognosis of mild cognitive impairment in early
Parkinson disease: the Norwegian ParkWest study. JAMA Neurol. 2013;70:580–6.
128 C. Rodriguez-Blazquez et al.
Ramaker C, Marinus J, Stiggelbout AM, van Hilten BJ. Systematic evaluation of rating scales for
impairment and disability in Parkinson’s disease. Mov Disord. 2002;17:867–76.
Ramirez Diaz SP, Gil Gregorio P, Ribera Casado JM, Reynish E, Ousset PJ, Vellas B, et al. The
need for a consensus in the use of assessment tools for Alzheimer’s disease: the Feasibility
Study (assessment tools for dementia in Alzheimer Centres across Europe), a European
Alzheimer’s Disease Consortium’s (EADC) survey. Int J Geriatr Psychiatry. 2005;20:744–8.
Reid WGJ, Broe GA, Hely MA, Morris JG, Williamson PM, O’Sullivan DJ, et al. The neuropsy-
chology of de novo patients with idiopathic Parkinson’s disease: the effects of age of onset. Int
J Neurosci. 1989;48:205–17.
Reisberg B. Functional assessment staging (FAST). Psychopharmacol Bull. 1988;24(4):653–9.
Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase inhibitors for dementia with Lewy
bodies. Parkinson’s disease dementia and cognitive impairment in Parkinson’s disease.
Cochrane Database Syst Rev. 2012;3, CD006504.
Sabbagh MN, Adler CH, Lahti TJ, Connor DJ, Vedders L, Peterson LK, et al. Parkinson disease
with dementia: comparing patients with and without Alzheimer pathology. Alzheimer Dis
Assoc Disord. 2009;23:295–7.
Seppi K, Yekhlef F, Diem A, Luginger Wolf E, Mueller J, Tison F, et al. Progression of parkinson-
ism in multiple system atrophy. J Neurol. 2005;252:91–6.
Tison F, Yekhlef F, Chrysostome V, Balestre E, Quinn NP, Poewe W, et al. Parkinsonism in multiple
system atrophy: natural history, severity (UPDRS-III), and disability assessment compared
with Parkinson’s disease. Mov Disord. 2002a;17:701–9.
Tison F, Yekhlef F, Balestre E, Chrysostome V, Quinn N, Wenning GK, et al. Application of the
international cooperative ataxia scale rating in multiple system atrophy. Mov Disord. 2002b;17:
1248–54.
Webster DD. Critical analysis of the disability in Parkinson’s disease. Mod Treat. 1968;5:
257–82.
Wenning GK, Tison F, Seppi K, Sampaio C, Diem A, Yekhlef F, et al. Development and validation
of the Unified Multiple System Atrophy Rating Scale (UMSARS). Mov Disord. 2004;19:
1391–402.
Wenning GK, Geser F, Krismer F, Seppi K, Duerr S, Boesch S, et al. The natural history of mul-
tiple system atrophy: a prospective European cohort study. Lancet Neurol. 2013;12:264–74.
Whitwell JL, Master AV, Avula R, Kantarci K, Eggers SD, Edmonson HA, Jack Jr CR, Josephs
KA. Clinical correlates of white matter tract degeneration in progressive supranuclear palsy.
Arch Neurol. 2011;68:753–60.
Williams MM, Xiong C, Morris JC, Galvin JE. Survival and mortality differences between demen-
tia with Lewy bodies vs Alzheimer disease. Neurology. 2006;67:1935–41.
Wilson RS, Bennett DA, Gilley DW, Beckett LA, Schneider JA, Evans DA. Progression of parkin-
sonian signs in Alzheimer’s disease. Neurology. 2000;54:1284–9.
Yahr MD, Duvoisin RC, Schear MJ, Barrett RE, Hoehn MM. Treatment of parkinsonism with
levodopa. Arch Neurol. 1969;21:343–54.
Zubenko GS, Moossy J, Martinez AJ, et al. Neuropathologic and neurochemical correlates of
psychosis in primary dementia. Arch Neurol. 1991;48:619–24.
Chapter 8
Movement Disorders in Alzheimer’s Disease
Alzheimer’s Disease
Alzheimer’s disease (AD) is the most common type of dementia, affecting more
than five million people in the USA (Bateman et al. 2012). AD accounts for over
70 % of dementia cases among individuals over the age of 70 years (Tarawneh and
Holtzman 2012), and its incidence increases exponentially with age (Kukull et al.
2002). Without successful treatment, AD will affect about 200 million individuals
by the year 2050 (Thies and Bleiler 2011). Neuropathological changes are known to
start several decades before the first clinical symptoms (Jack et al. 2010; Price and
Morris 1999; Braak and Braak 1997), especially among those with autosomal domi-
nant AD. Brain changes compatible with dementia are present in about 65 % of
80-year-olds (Villegmane et al. 2011; Rowe et al. 2010).
Diagnosis of AD
Since 2007, two sets of diagnostic criteria have been introduced in clinical practice.
Dubois and coworkers proposed the International Work Group (IWG) criteria for
the diagnosis of AD (Cummings et al. 2013). The novelty of these criteria is that
they require a clinical phenotype characterized by anterograde amnesia; supportive
evidence of biomarkers, such as positron emission tomography (PET) brain amy-
loid scans and brain metabolic activity and structural analysis using PET and mag-
netic resonance imaging, respectively; and genetic and cerebrospinal fluid (CSF)
analysis. Thus, the IWG criteria integrate the assessment of biomarkers to clinical
criteria, providing a more biological approach to diagnosis. The IWG criteria for
probable AD include the core diagnostic criteria for a gradual and progressive
impairment of episodic memory over more than 6 months. The recall deficit does
not improve with cueing or recognition, and the memory deficit may be an isolated
finding or be associated with other cognitive deficits. Additionally, one or more of
the following supportive features are referred for a diagnosis of probable AD: (1)
medial temporal lobe atrophy, (2) low 1–42 beta amyloid or increased total tau in
the CSF, (3) hypometabolism in bilateral temporoparietal regions, or (4) a proven
AD autosomal dominant mutation (Dubois et al. 2010). This strategy also allows
classifying groups into those with prodromal AD (memory deficits, one or more
positive biomarkers, and no impairments on activities of daily living (ADLs)) and
AD dementia, when deficits in ADLs are present. Finally, the use of biomarkers
identifies four diagnostic groups: (1) asymptomatic at-risk state with positive bio-
markers, (2) preclinical AD with an autosomal dominant mutation, (3) prodromal
AD with memory deficits and a biomarker for AD, and (4) AD dementia.
The second set of criteria were recently proposed by McKhann and coworkers
(2011). The authors proposed core clinical criteria for AD which include the pres-
ence of cognitive and neuropsychiatric symptoms that produce impairments in
ADLs representing a clear decline from previous levels of functioning. Cognitive
impairment is diagnosed based on a clinical history assessed with the patient and an
informant, as well as an objective cognitive impairment. The diagnosis of probable
AD also requires anterograde amnesia, poor judgment, impaired visuospatial abili-
ties, impaired language, and personality changes such as apathy and disinhibited
behaviors. For a diagnosis of probable AD, patients not only have to meet the above
criteria but also must have an insidious onset, history of worsening, and initial and
prominent cognitive deficits characterized by anterograde memory deficits, word-
finding deficits, or impaired reasoning.
The above set of criteria also include the major biomarkers for AD, such as a
brain beta amyloid deposition (e.g., low CSF A-beta 42, positive PET amyloid imag-
ing), or biomarkers of neural degeneration (increased CSF tau, hypometabolism
8 Movement Disorders in Alzheimer’s Disease 131
Parkinsonism
motor signs only at a single point during the course of AD, typically at the baseline
visit or less frequently at any point during the disease course, but patients’ lack of
follow-up and evolution of motor signs during disease progression or even its asso-
ciation with different outcomes are difficult to describe.
In one early attempt to describe parkinsonism in AD, we used the motor section
of the UPDRS to divide patients into two groups: (a) Alzheimer’s disease–parkinson-
ism: patients included in this group had rigidity, bradykinesia, and resting tremor;
rigidity plus bradykinesia only; or resting tremor only. Bradykinesia was defined as
a score >1 on the finger tapping, rapid hand movements, and alternating movements
of the hands sections of the UPDRS. Rigidity was defined as a score >1 in the UPDRS
(only cogwheel rigidity was considered). (b) Alzheimer’s disease–extrapyramidal
signs: patients included in this group had extrapyramidal signs other than bradykine-
sia, rigidity, or resting tremor (flexed posture, gait disorders, masked facies). In our
cross-sectional study of 78 patients, we found that 23 % of the patients showed par-
kinsonism, 56 % showed isolated extrapyramidal signs, and only 21 % had no extra-
pyramidal signs. Whereas the frequency of parkinsonism was significantly higher
than in age-comparable normal controls, the prevalence of isolated extrapyramidal
signs was not significantly different. Secondly, parkinsonism in Alzheimer’s disease
failed to improve after the apomorphine test. Thirdly, patients with Alzheimer’s dis-
ease–parkinsonism had significantly more severe deficits on neuropsychological
tasks assessing abstract reasoning, set-shifting abilities, and executive functions than
patients with Alzheimer’s disease but no extrapyramidal signs, whereas patients with
Alzheimer’s disease and isolated extrapyramidal signs had scores in between both
groups. Lastly, patients with Alzheimer’s disease–parkinsonism showed a signifi-
cantly higher frequency of both major depression and dysthymia than patients with
Alzheimer’s disease and no extrapyramidal signs (Merello et al. 1994). The relation-
ship between parkinsonism and psychiatric symptoms in AD was later confirmed in
a larger study on 169 patients meeting diagnostic criteria for AD followed between 1
and 4 years after the baseline evaluation. Patients with apathy at baseline or those
who developed apathy during follow-up had a significant increase in parkinsonism at
follow-up when compared with patients with no apathy at both assessments. The
association between apathy and increasing parkinsonism was unrelated to age, gen-
der, the severity of cognitive deficits, the presence of depression, or use of psychotro-
pic medications. On the other hand, neither the presence of parkinsonism nor
depression at baseline was significantly associated with more severe apathy at fol-
low-up (Starkstein et al. 2009). In addition Park et al. described that the risk of sleepi-
ness was 2.37 times greater in AD participants with parkinsonian features when
compared to the AD participants without parkinsonism (Park et al. 2011).
There is little published information on patterns of progression of parkinsonism
in AD. Several studies have shown that the probability of meeting criteria for par-
kinsonism increases with time (Morris et al. 1989; Stern et al. 1996; Chen et al.
1991). Wilson et al. (2000) reported an average of 4.5 % of annual increases in
bradykinesia, 6.1 % on rigidity, and 8.9 % on gait and posture. While large cohort
studies reported an average annual increase of 1.5 % on UPDRS scores on people
with PD, Wilson et al. reported that in persons with AD, parkinsonism progressed
8 Movement Disorders in Alzheimer’s Disease 133
more than twice as fast (Wilson et al. 2000). The same authors interestingly reported
that there was an average annual increase of 1.39 units in postural tremor (95 % CI,
0.76–2.02), whereas little change was evident in resting tremor (mean, 0.33 units;
95 % CI, 0.12–0.54).
Motor signs in AD are important because they may predict cognitive and func-
tional decline, institutionalization, and death, and as compared to patients with AD
without motor signs, patients with AD with motor signs have higher annual total
cost of care (Murman et al. 2003; Morris et al. 1989; Soininen et al. 1992; Haan
et al. 2002). Scarmeas et al. even found certain selectivity of each motor sign to
determine specific outcome. In that way, presence of tremor was associated with
increased risk for cognitive decline, presence of bradykinesia with increased risk for
functional decline, and presence of postural–gait impairments with increased risk
for institutionalization and death (Scarmeas et al. 2005).
The modification of parkinsonism in AD after dopaminergic therapy presents an
important clinical as well psychopathological implicancies. We have not found clin-
ical improvements after the apomorphine injection in 11 Alzheimer’s disease
patients with parkinsonism suggesting that parkinsonism in AD is not related to
presynaptic nigrostriatal dysfunction (Merello et al. 1994). This may be in agree-
ment with DaTscan studies which showed indemnity of nigrostriatal pathway in AD
patients with parkinsonism (Vaamonde-Gamo et al. 2005). On the other hand, we
have examined the presence of significant regional cerebral blood flow (rCBF) dif-
ferences between AD patients with and without parkinsonism. Nine patients with
AD and parkinsonism showed significantly lower rCBF in the superior frontal,
superior temporal, and parietal regions of the left hemisphere than AD patients
without parkinsonism. Rigidity and bradykinesia independently accounted for the
decreased rCBF in these areas. These findings suggest that the presence of EPS in
AD may result from dysfunction in specific brain regions other than basal ganglia
(Starkstein et al. 1995).
Parkinsonism
DLBD
Dementia
Parkinsonism
AD
Dementia
Parkinsonism
PD-D
Dementia
1 Year span
Years
Fig. 8.1 Temporal relationship between parkinsonism and disease evolution in Parkinson’s
disease-related dementia, diffuse Lewy body dementia, and Alzheimer’s disease
19 %/55 %, impaired chair rise 15 %/29 %, rest tremor 3 %/13 %, postural instabil-
ity 12 %/26 %, and postural tremor 6 %/12 %), logistic regression models consis-
tently demonstrate that all 9 EPSs differentiate DLB from AD, regardless of age,
sex, education, or MMSE, and being masked facies the one that best differentiated
AD from Lewy body (odds ratio, 6.5; P < 0.001; 95 % confidence interval, 3.8–
11.1), Kaur et al. (2013) findings were in agreement with those of Ballard et al. and
Galasko et al. (Ballard et al. 1997; Galasko et al. 1996), which demonstrate that rest
tremor, action tremor, bradykinesia, facial expression, and rigidity subscales of the
UPDRS were helpful in differentiating DLB from AD. Patients with DLB experi-
ence a greater level of functional impairment than subjects with AD of similar cog-
nitive ability largely attributable to parkinsonism (Chap. 10). The loss of
dopaminergic neurons in DLB can be confirmed in vivo with a presynaptic dopa-
mine transporter marker DaTscan, whereas there are no changes in DaTscan in AD
compared with controls (Piggott et al. 1999).
In patients with rapidly progressive dementia and motor signs, Creutzfeldt–Jakob
disease (CJD) should be the first suspected diagnosis. However, the presence of
dementia, myoclonus, and rigidity with a longer course of disease may suggest AD or
even DLBD. In an elegant study, Tschampa et al. (2001) described that myoclonus and
extrapyramidal signs were especially common in patients with CJD and patients with
DLB and in more than 50 % of patients with AD. Extrapyramidal signs in patients with
8 Movement Disorders in Alzheimer’s Disease 135
AD and with DLB were restricted to limb rigidity (>50 % respectively) and parkinson-
ism (58 % in patients with DLB, 8 % in CJD, and 11 % in AD). Patients with CJD
additionally showed hyperkinetic signs (40 %). Nearly all patients with CJD and 37 %
of patients with AD had cerebellar signs, not noted in DLB. Regarding hyperkinetic
movement disturbances, dystonia and athetosis were seen exclusively in CJD patients
(Edler et al. 2009). In other comparative studies, dystonia and athetosis were also seen
in CJD patients only, but dystonia has also been reported to be present at a low fre-
quency of 6 % in DLB patients (Louis et al. 1997; Burkhardt et al. 1988) (Fig. 8.2).
AD
Slow progression
Disease duration >2 years
Initial memory impairment
Dementia
Hypertonia
Myoclonus
Sleep disruption
DLBD Cognitive fluctuations Behavioural
Disease duration >2 years disturbances
Parkinsonism
Paratonia
Dystonia
Cerebellar signs
Pyramidal signs
Akinetic mutism
Seizures
Chorea
CJD
Fig. 8.2 Movement disorders occurring in Alzheimer’s disease, diffuse Lewy body disease, and
Creutzfeldt–Jakob disease
136 S.E. Starkstein and M. Merello
Corticobasal Syndrome
Myoclonus
Pisa Syndrome
Pisa syndrome was originally coined to describe a rare form of tardive dystonia or
acute dystonia associated with the use of neuroleptics. The typical clinical picture is
the tonic lateral flexion of the trunk associated with a mild backward rotation and
not related to musculoskeletal sustained fixed abnormalities. There is a current
debate whether it corresponds to the same entity of persistent lateral deviation and
whether it is associated with camptocormia. It has been described in a Parkinson’s
disease patient, and the sign has been included as supportive criterion for multiple
system atrophy. EMG showed abnormal tonic hyperactivity on the side of the devia-
tion in the paravertebral thoracic muscles and in the abdominal oblique muscles
(Tassorelli et al. 2012). It has been suggested that Pisa syndrome corresponded to a
8 Movement Disorders in Alzheimer’s Disease 137
Paratonia
Dystonia
References
Jack Jr CR, Knopman DS, Jaqust WJ, et al. Hypothetical model of dynamic biomarkers of the
Alzheimer’s pathological cascade. Lancet Neurol. 2010;9:119–28.
Kaur B, Harvey DJ, Decarli CS, et al. Extrapyramidal signs by dementia severity in Alzheimer
disease and dementia with Lewy bodies. Alzheimer Dis Assoc Disord. 2013;27(3):226–32.
Kauwe JS, Wang J, Chakraverty S, et al. Novel presenilin 1 variant (P117A) causing Alzheimer’s
disease in the fourth decade of life. Neurosci Lett. 2008;438:257–9.
Kukull WA, Higdon R, Bowen JD, et al. Dementia and Alzheimer disease incidence: a prospective
cohort study. Arch Neurol. 2002;59:1737–46.
Lleo A, Rey MJ, Castellvi M, et al. Asymmetric myoclonic parietal syndrome in a patient with
Alzheimer’s disease mimicking corticobasal degeneration. Neurologia. 2002;17:223–6.
Louis ED, Klatka LA, Liu Y, Fahn S. Comparison of extrapyramidal features in 31 pathologically
confirmed cases of diffuse Lewy body disease and 34 pathologically confirmed cases of
Parkinson’s disease. Neurology. 1997;48:376–80.
Lowe VJ, Peller PJ, Weigand SD, et al. Application of the National Institute on Aging-Alzheimer’s
Association AD criteria to ADNI. Neurology. 2013;80:2130–7.
Magnuson TM, Roccaforte WH, Wengel SP, Burke WJ. Medication-induced dystonias in nine
patients with dementia. J Neuropsychiatry Clin Neurosci. 2000;12:219–25.
McKeith IG, Perry RH, Fairbairn AF, et al. Operational criteria for senile dementia of Lewy body
type (SDLT). Psychol Med. 1992;22:911–22.
McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s
disease: recommendations from the National Institute on Aging-Alzheimer’s Association
workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement.
2011;7:263–9.
Merello M, Sabe L, Teson A, et al. Extrapyramidalism in Alzheimer’s disease: prevalence, psychi-
atric, and neuropsychological correlates. J Neurol Neurosurg Psychiatry. 1994;57:1503–9.
Miyaoka T, Seno H, Yamamori C, et al. Pisa syndrome due to a cholinesterase inhibitor (donepe-
zil): a case report. J Clin Psychiatry. 2001;62:573–4.
Morris JC, Drazner M, Fulling K, et al. Clinical and pathological aspects of parkinsonism in
Alzheimer’s disease. Arch Neurol. 1989;46:651–7.
Murman DL, Kuo SB, Powell MC, Colenda CC. The impact of parkinsonism on costs of care in
patients with AD and dementia with Lewy bodies. Neurology. 2003;61:944–9.
Park M, Shah RC, Fogg LF, Wyatt JK. Daytime sleepiness in mild Alzheimer’s disease with and
without parkinsonian features. Sleep Med. 2011;12:397–402.
Pavlis CJ, Kutscher EC, Carnahan RM, et al. Rivastigmine-induced dystonia. Am J Health Syst
Pharm. 2007;64:2468–70.
Piggott MA, Marshall EF, Thomas N, et al. Striatal dopaminergic markers in dementia with Lewy
bodies, Alzheimer’s and Parkinson’s diseases: rostrocaudal distribution. Brain. 1999;122:
1449–68.
Price JL, Morris JC. Tangles and plaques in nondemented aging and “preclinical” Alzheimer’s
disease. Ann Neurol. 1999;45:358–68.
Richards M, Stern Y, Marder K, et al. Relationships between extrapyramidal signs and cognitive
function in a community-dwelling cohort of patients with Parkinson’s disease and normal
elderly individuals. Ann Neurol. 1993;33:267–74.
Richards M, Touchon J, Ledesert B, et al. Mild extrapyramidal signs and functional impairment in
ageing. Int J Geriatr Psychiatry. 2002;17:150–3.
Risse SC, Raskind MA, Nochlin D, et al. Neuropathological findings in patients with clinical
diagnoses of probable Alzheimer’s disease. Am J Psychiatry. 1990;147:168–72.
Rowe CC, Ellis KA, Rimajowa M, et al. Amyloid imaging results from the Australian Imaging,
Biomarkers and Lifestyle (AIBL) study of aging. Neurobiol Aging. 2010;31:1275–83.
Scarmeas N, Albert M, Brandt J, et al. Motor signs predict poor outcomes in Alzheimer disease.
Neurology. 2005;64:1696–703.
Sherrington R, Rogaev EI, Liang Y, et al. Cloning of a gene bearing missense mutations in
early-onset familial Alzheimer’s disease. Nature. 1995;375:754–60.
140 S.E. Starkstein and M. Merello
Shih LC, Vanderhorst VG, Lozano AM, et al. Improvement of pisa syndrome with contralateral
pedunculopontine stimulation. Mov Disord. 2013;28:555–6.
Shim YS, Roe CM, Buckles VD, Morris JC. Clinicopathologic study of Alzheimer’s disease:
Alzheimer mimics. J Alzheimers Dis. 2013;35:799–811.
Soininen H, Laulumaa V, Helkala EL, et al. Extrapyramidal signs in Alzheimer’s disease: a 3-year
follow-up study. J Neural Transm Park Dis Dement Sect. 1992;4:107–19.
Spina S, Murrell JR, Huey ED, et al. Corticobasal syndrome associated with the A9D Progranulin
mutation. J Neuropathol Exp Neurol. 2007;66:892–900.
Starkstein SE, Vazquez S, Merello M, et al. SPECT study of parkinsonism in Alzheimer’s disease.
J Neuropsychiatry Clin Neurosci. 1995;7:308–13.
Starkstein SE, Merello M, Brockman S, et al. Apathy predicts more severe parkinsonism in
Alzheimer’s disease. Am J Geriatr Psychiatry. 2009;17:291–8.
Stern Y, Liu X, Albert M, et al. Modeling the influence of extrapyramidal signs on the progression
of Alzheimer’s disease. Arch Neurol. 1996;53:1121–6.
Tarawneh R, Holtzman DM. The clinical problem of symptomatic Alzheimer disease and mild
cognitive impairment. Cold Spring Harb Perspect Med. 2012;2:1–16.
Tassorelli C, Furnari A, Buscone S, et al. Pisa syndrome in Parkinson’s disease: clinical, electro-
myographic, and radiological characterization. Mov Disord. 2012;27:227–35.
Thies W, Bleiler L. 2011 Alzheimer’s disease facts and figures. Alzheimers Dement.
2011;7:208–44.
Tschampa HJ, Neumann M, Zerr I, et al. Patients with Alzheimer’s disease and dementia with
Lewy bodies mistaken for Creutzfeldt-Jakob disease. J Neurol Neurosurg Psychiatry.
2001;71:33–9.
Vaamonde-Gamo J, Flores-Barragan JM, Ibañez R, et al. DaT-SCAN SPECT in the differential
diagnosis of dementia with Lewy bodies and Alzheimer’s disease. Rev Neurol.
2005;41:276–9.
Vahia I, Cohen CI, Prehogan A, Memon Z. Prevalence and impact of paratonia in Alzheimer dis-
ease in a multiracial sample. Am J Geriatr Psychiatry. 2007;15:351–3.
van den Bosch S, Bouckaert F, Peuskens J. Mirtazapine-induced dystonia in a patient with
Alzheimer’s disease. Tijdschr Psychiatr. 2006;48:153–7. Dutch.
Vanacore N, Suzzareddu G, Maggini M, et al. Pisa syndrome in a cohort of Alzheimer’s disease
patients. Acta Neurol Scand. 2005;111:199–201.
Villarejo A, Camacho A, Garcia-Ramos R, et al. Cholinergic-dopaminergic imbalance in Pisa syn-
drome. Clin Neuropharmacol. 2003;26:119–21.
Villegmane VL, Pike KE, Chételat G, et al. Longitudinal assessment of Aβ and cognition in aging
and Alzheimer disease. Ann Neurol. 2011;69:181–92.e.
Wilson RS, Bennett DA, Gilley DW, et al. Progression of Parkinsonian signs in Alzheimer’s dis-
ease. Neurology. 2000;54:1284–9.
Wisniewski T, Dowjat WK, Buxbaum JD, et al. A novel Polish presenilin-1 mutation (P117L) is
associated with familial Alzheimer’s disease and leads to death as early as the age of 28 years.
Neuroreport. 1998;9:217–21.
Zekanowski C, Styczynska M, Peplonska B, et al. Mutations in presenilin 1, presenilin 2 and amy-
loid precursor protein genes in patients with early-onset alzheimer’s disease in Poland. Exp
Neurol. 2003;184:991–6.
Chapter 9
Movement Disorders in Frontotemporal
Dementia
Abbreviations
Background
have little or no insight but is a cause of considerable distress for carers (Mioshi
et al. 2009). The diagnosis hinges on a detailed carer interview to elicit key behav-
ioral features, notably apathy, alterations in social conduct and inhibitory control,
loss of empathy, mental rigidity, stereotypic speech and motor behaviors, and a
change in eating habits toward sweet cravings and a lack of satiety. In addition,
patients may exhibit prominent executive deficits. It has been suggested that mem-
ory is spared in FTD, however recent studies have demonstrated poor performance
of tests of episodic memory (Hornberger et al. 2010). In contrast to Alzheimer’s
disease, orientation is well preserved. In parallel, patients typically have neuroimag-
ing evidence of atrophy in the orbitofrontal cortex, insula, and medial thalamus on
MRI or hypoperfusion in these areas on functional imaging (Schroeter et al. 2008).
Unlike some of the other FTD syndromes, it is difficult to predict pathology;
approximately 50 % of cases have FTLD-tau pathology, while the other 50 % have
FTLD-TDP deposition. Of all variants, bvFTD appears to be most likely associated
with ALS, particularly in those patients harboring the C9ORF72 genetic mutation.
Semantic dementia is characterized by progressive anomia and impaired word
comprehension secondary to dissolution of semantic networks in the anterior tempo-
ral lobe (Hodges et al. 1992). The loss of vocabulary affects mainly nouns in the early
stages and shows a marked familiarity effect, in that even advanced cases can recog-
nize high-frequency words. In contrast, phonology and syntax are relatively pre-
served, and speech remains fluent although simplified with use of higher-frequency
substitutions and filler words such as “thing” and “that place.” Patients initially func-
tion very well at home; however as the disease progresses, object knowledge and use
becomes impaired, and this parallels the language deficits with lower-frequency
objects initially compromised. The anterior temporal lobe atrophy is usually asym-
metrical and predominantly left sided in most cases (Hodges et al. 1992). Deficit in
person recognition (prosopagnosia) is the key feature of the right predominant syn-
drome, which progresses to a cross-modal loss of face, name, and voice recognition
(Evans et al. 1995). Behavioral features, similar to those found in bvFTD, are com-
mon in patients with SD and particularly in those with the right temporal variant.
Pathologically SD is relatively homogenous with the majority of cases showing TDP-
43 pathology, and of all FTD variants, it is most likely to be sporadic.
In contrast to SD, PNFA patient’s speech is strikingly nonfluent with speech
distortion, pauses, groping, and agrammatism (Gorno-Tempini et al. 2011). The
distortion of speech is due to breakdown in motor planning, referred to as speech
apraxia, causing impairment of rhythm and the normal stress patterns of speech.
These deficits occur in the presence of spared word comprehension. Sentence com-
prehension however can be impaired due to problems understanding the grammati-
cal elements of sentences. Word repetition is often impaired due to articulatory
errors. FTLD-tau is most frequently present in these cases. Patients with PNFA
commonly develop Parkinsonism and apraxia which may lead to a change in syn-
dromic diagnosis to corticobasal syndrome (CBS) or progressive supranuclear palsy
(PSP) (Kertesz et al. 2005). In a large longitudinal clinicopathological study Kertesz
and colleagues (Kertesz et al. 2005) identified evolution to CBS or PSP in almost
half of 22 cases of PNFA.
144 E. Devenney and J. Hodges
The neural basis of parkinsonian features in FTD remains unclear. A degree of atrophy
of the basal ganglia and histopathological changes in the striatum have been reported
(Mann et al. 1993). Another study found a significant inverse correlation between the
degree of severity of parkinsonian symptoms and the degree of uptake of a dopamine
transporter ligand, suggesting that striatal dysfunction is a factor in these conditions
(Rinne et al. 2002). This idea was corroborated in a case series of six patients who satis-
fied criteria for both dementia with Lewy bodies (DLB) and FTD at presentation: two
cases came to autopsy, pathological study of these cases revealed gliosis and neuronal
loss in the basal ganglia and substantia nigra with pigment loss in the latter (Claassen
et al. 2008). While Parkinsonism in FTD has been linked to underlying tau pathology
(Hodges et al. 2004), TDP-43 pathology was actually present in each of these cases.
As CBS and PSP are discussed in detail in another chapter, the remainder of this
chapter will focus on the inherited forms of FTD.
Background
Parkinsonism in FTD has been reported in the literature since the turn of the century
but was initially overlooked in favor of the prominent behavioral and language fea-
tures. In 1998 a dementia and Parkinsonism syndrome was linked to chromosome
17q21-22 in an Irish kindred which sparked considerable interest in this area (Lendon
et al. 1998). The first frontotemporal dementia and Parkinsonism consensus consor-
tium met in Ann Arbor in 1996 to discuss the clinical and neuropathological features
of this and 12 other kindreds with FTDP-17, as it then became known (Foster et al.
1997). Despite clinical heterogeneity within and between families, a severe behav-
ioral disorder characteristic of FTD and Parkinsonism without resting tremor was
common to all. Neuropathologically, there was atrophy of the frontal and temporal
cortices, as well as the basal ganglia and depigmentation of the substantia nigra. A
defect in the microtubule-associated protein tau (MAPT) gene on chromosome 17
was discovered 2 years later in a proportion of FTDP-17 families (Hutton et al. 1998),
but it was almost another decade before a mutation in the progranulin gene (GRN)
was discovered to account for the remaining FTDP-17 families (Gass et al. 2006).
Over 40 mutations in the MAPT gene have now been described, all of which
have underlying tau pathology, and account for approximately 30 % of all famil-
ial FTD (Houlden et al. 1999; Poorkaj et al. 2001; Rosso et al. 2003; Stanford
et al. 2004). Over 50 mutations of the GRN gene have been described; these GRN
146 E. Devenney and J. Hodges
Clinical Features
At a clinical level, FTDP-17 associated with MAPT and GRN mutations has com-
mon and contrasting features (Table 9.1). Both show an autosomal dominant pattern
of inheritance (Foster et al. 1997). MAPT mutation carriers can present initially with
either a predominant parkinsonian syndrome or a dementia (Baba et al. 2005;
Tsuboi et al. 2002), with the precise mutation determining the clinical phenotype;
the N279K mutation has a predominant parkinsonian presentation (Wszolek et al.
2000), personality and behavioral changes characterize the P301L and 10 +16 muta-
tion (Kodama et al. 2000; Janssen et al. 2002), and the S305S mutation does not
appear to favor either (Stanford et al. 2000; Reed et al. 2001).
Although the reported age of onset ranges from 25 to 65 years in MAPT carriers,
the majority of patients first exhibit signs in their 40s and live for 3–10 years with the
disease (Boeve and Hutton 2008; Espay and Litvan 2011). An exception to this is the
R406W mutation carriers who have a much longer disease duration with reports of
patient surviving into their 70s (Van Swieten et al. 1999). GRN mutations tend to pres-
ent later than MAPT, with most patients developing symptoms after 50 years of age
with a mean disease duration of 5 years (3–22 years) and a shorter disease duration
in late-onset disease (Van Swieten and Heutink 2008). The MAPT mutation is almost
fully penetrant, although there are reports of asymptomatic individuals living into old
age (Hutton et al. 1998). In contrast, GRN carriers exhibit incomplete penetrance with
10 % of carriers remaining asymptomatic at 70 years (Gass et al. 2006; Boeve and
Hutton 2008). Parkinsonism has been reported in approximately 40 % of GRN cases
(Le Ber et al. 2007). In common with MAPT mutation carriers, the Parkinsonism is
generally symmetrical with bradykinesia, rigidity, and rarely resting tremor and with
modest or little response to levodopa (Tsuboi et al. 2002). Features typically associated
with PSP, including supranuclear gaze palsy, eyelid apraxia, myoclonus, dysphagia,
9 Movement Disorders in Frontotemporal Dementia 147
dysarthria, and pyramidal features, have all been reported in MAPT mutation carriers
(Stanford et al. 2000), while dystonia is infrequent in all mutation carriers.
TDP-43 pathology is common to both patients with FTD in association with GRN
mutations and patients with ALS. Despite this commonality, features of ALS are not
found in GRN carriers which suggests a distinct pathogenetic mechanism in GRN
carriers. ALS features are also rarely found in MAPT carriers. (Seelaar et al. 2007;
Pickering-Brown et al. 2008; Tsuboi 2006). By contrast although apraxia is rare in
MAPT carriers, it can be a feature of GRN carriers. Severe behavioral disorders are
found in the majority of carriers of both mutations, in addition to frontal dysexecutive
cognitive deficits (Boeve and Hutton 2008; Le Ber et al. 2007). In one published GRN
case, the patient developed features of Kluver-Bucy syndrome with prominent hyper-
sexuality, hyperphagia, and psychotic features (Boeve et al. 2006). In general, psy-
chotic features have been reported in up to a quarter of GRN carriers and include
hallucinations and/or delusions which in some cases has led to misdiagnoses of
schizophrenia. Hallucinations include visual (animals and people) reported in 25 % in
one series and tactile (insects crawling over the skin) in another series (Le Ber et al.
2007; Beck et al. 2008). Psychotic features are also reported in MAPT carriers but
appear to be less common (Saito et al. 2002; Spina et al. 2007). Nonfluent aphasia
often accompanies the behavioral and personality deficits in GRN carriers but is rarely
the predominant feature, and semantic dementia is very rare (Pickering-Brown et al.
2008). A dynamic aphasia has also been documented in a number of patients, charac-
terized by a decreased output of speech without evidence of a motor speech disorder
or agrammatism, resulting in mutism in a selection of patients (Beck et al. 2008).
Imaging in FTLD-17
Gray and white matter atrophy patterns can aid in the distinction between the genetic
causes of FTLD-17 and may offer insight into the neural basis of symptomatology
in these diseases. In brief, voxel-based morphometry (VBM) analyses have deter-
mined significant variations in the neuroimaging signatures of GRN and MAPT
mutations. GRN mutation carriers have smaller brain volumes and tend to atrophy
faster than MAPT carriers. The most striking difference is in the hemispheric asym-
metry seen in GRN carriers compared to relatively symmetrical atrophy in MAPT
carriers. As might be expected given the high prevalence of apraxia, GRN carriers
tend to have an atrophy pattern involving the inferior parietal lobe as well as inferior
frontal and temporal lobes. In contrast, MAPT carriers exhibit a symmetrical antero-
medial temporal lobe and orbitofrontal atrophy with involvement of the fornix.
Involvement of the long intrahemispheric association tracts has also been docu-
mented in those with GRN mutations (Rohrer et al. 2010a; Whitwell et al. 2012).
It is now clear that ALS overlaps considerably at a clinical, pathological, and genetic
level with FTD to such an extent that the two disorders are considered to form a
148 E. Devenney and J. Hodges
continuum with pure ALS at one end of the spectrum and pure FTD (without ALS) at
the other (Lillo and Hodges 2009), although most cases appear to fall somewhere in the
middle with around 20 % of ALS cases also meeting criteria for FTD and a much
greater proportion having subtle behavioral and/or cognitive changes (Strong 2008).
The recent discovery of the C9ORF72 genetic mutation has consolidated this concept
(DeJesus-Hernandez et al. 2011; Renton et al. 2011) and extended this overlap to
involve Parkinsonism. Parkinsonism has been reported in up to 30 % of patients with
this mutation, although reports have varied between studies (Snowden et al. 2012;
Boeve et al. 2012; Sha et al. 2012). The Parkinsonism mirrors that found in other inher-
ited and sporadic cases of FTD with bilateral rigidity and bradykinesia without tremor.
It has been proposed that intermediate numbers of repeat copies of the mutation confer
a significant risk of developing Parkinson’s disease (Nuytemans et al. 2013). The
imaging finding in C9ORF72 carriers are variable. A proportion present with frontal
and temporal atrophy typically found in patients with FTD, while others have rela-
tively normal MRI imaging. Atrophy of subcortical structures including the thalamus
and cerebellum are emerging as a diagnostic marker of the C9ORF72 mutation
(Whitwell et al. 2012). A higher burden of TDP-43 pathology and neuronal cell death
in the substantia nigra of mutation carriers compared to noncarriers may explain the
increased incidence of parkinsonian features in this cohort (Cooper-Knock et al. 2013).
Perry Syndrome
Perry syndrome is a rare genetic disorder due to a G71R mutation in the DCTN1
gene, characterized by Parkinsonism, hypoventilation, weight loss, depression, or
psychiatric disease. In one published case, the patient exhibited slowing of vertical
saccades and a marked behavioral disorder typical of bvFTD but without focal atro-
phy on MRI or convincing executive cognitive deficits (Newsway et al. 2010). This
rare disorder is also linked to FTD, as TDP-43 pathology is common to both.
Dystonia has been rarely reported in FTD, aside from a handful of instances where
it has been linked with mutations in the MAPT and GRN gene. It has, however, been
described in FTD patients treated with antipsychotic medications. All three patients
in a case series of FTD patients treated with antipsychotic medications developed a
tardive dystonia which manifested as an antecollis (Czarnecki et al. 2008). In another
case series, up to 30 % of FTD patients developed extrapyramidal features when
treated with antipsychotic medications (Pijnenburg et al. 2003). The dangers of such
medication have been well documented in DLB and linked to dopaminergic dysfunc-
tion (Piggott et al. 1998). It seems logical then that a similar danger exists in FTD
cohorts and accords well with the evidence for striatal dysfunction in FTD patients.
9 Movement Disorders in Frontotemporal Dementia 149
Similar to dystonia, myoclonus has been reported rarely and exclusively in cases
of FTDP-17. In these cases, the myoclonus involved the upper extremities and was
described as small, subtle, and action induced, without detectable activation with
rest stimuli (Caviness and Wszolek 2002).
Apraxia
This section considers the literature on apraxia in FTD particularly in PNFA which,
of the FTD syndromes, shows the greatest overlap with CBS at both a clinical and
pathological level (Kertesz et al. 2005). Apraxia is a core component of CBS which
must be present to make the diagnosis, and is common in PSP (Soliveri et al. 2005;
Mathew et al. 2012). Patients with CBS also exhibit features of PNFA and/or behav-
ioral abnormalities. This can make it very difficult to disentangle the literature on
apraxia in FTD since many mixed cases are labeled as CBS rather than PNFA with
apraxia (Sha et al. 2006).
While apraxia is considered common in PNFA, it has not been well character-
ized. Variations in the definition of apraxia and a lack of consensus regarding the
underlying cortical circuitry involved have probably contributed to the dearth of
studies on this topic. Up until 2003 case reports and case series described the pres-
ence of apraxia in PNFA (Kertesz et al. 1994; Karbe et al. 1993). Others reported an
absence of apraxia in PNFA cases (Béland and Ska 1992; Craenhals et al. 1990).
The first prospective case–control study documented some form of apraxia in 90 %
of all PNFA subjects (Joshi et al. 2003): apraxia was present in 70 % for both pan-
tomime and imitation gestures, while 20 % showed apraxia for imitation only.
Unfortunately, this study had limitations: firstly, it is unclear how patients reaching
criteria for CBS or PSP were excluded. Moreover, evaluation of orofacial apraxia
and apraxia of speech (AOS) was not considered.
A comprehensive review of apraxia in 16 PNFA patients, using a standardized
Apraxia Battery and incorporating VBM analyses, was reported by Rohrer et al.
(2010b): three patients also had CBS and two PSP. Using a surrogate measure for
AOS, they found that all patients had evidence of AOS. More recently it has been
suggested that AOS can be the presenting sign of neurodegenerative disease in the
absence of aphasia referred to as primary progressive AOS (PPAOS) (Josephs et al.
2012). Longitudinal studies are needed to determine if such cases evolve into a
PNFA syndrome. In the Rohrer et al. cohort, orofacial apraxia was often associated
with AOS and the neural basis for both localized to the left inferior frontal gyrus.
Even after excluding patients with CBS and PSP over a quarter of PNFA, patients
exhibited features of limb apraxia.
In GRN mutation carriers, estimates of the rate of apraxia range from 16 to 71 % in
the bvFTD phenotype and 25–60 % in PNFA (Le Ber et al. 2007; Beck et al. 2008).
Approximately 6 % of GRN carriers satisfy criteria for CBS with apraxia, asymmetric
Parkinsonism, and rigidity (Le Ber et al. 2007; Pickering-Brown et al. 2008; Beck et al.
2008), while apraxia alone or in combination with CBS features is present in up to 50 %
150 E. Devenney and J. Hodges
of carriers. This likely reflects the parietal involvement seen in these cases. Apraxia is
not a common feature of the MAPT or the C9ORF72 mutation.
Future Directions
References
Baba Y, Tsuboi Y, Baker MC, Uitti RJ, Hutton ML, Dickson DW, et al. The effect of tau genotype
on clinical features in FTDP-17. Parkinsonism Relat Disord. 2005;11(4):205–8.
Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Lindholm C, et al.
Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome
17. Nature. 2006;442(7105):916–9.
Beck J, Rohrer JD, Campbell T, Isaacs A, Morrison KE, Goodall EF, et al. A distinct clinical, neu-
ropsychological and radiological phenotype is associated with progranulin gene mutations in a
large UK series. Brain. 2008;131(3):706–20.
Béland R, Ska B. Interaction between verbal and gestural language in progressive aphasia: a lon-
gitudinal case study. Brain Lang. 1992;43(3):355–85.
Boeve BF. Links between frontotemporal lobar degeneration, corticobasal degeneration, progres-
sive supranuclear palsy, and amyotrophic lateral sclerosis. Alzheimer Dis Assoc Disord.
2007;21(4):S31–8. Epub 2007/12/20.
Boeve BF, Hutton M. Refining frontotemporal dementia with parkinsonism linked to chromosome
17: introducing FTDP-17 (MAPT) and FTDP-17 (PGRN). Arch Neurol. 2008;65(4):460–4.
Epub 2008/04/17.
Boeve BF, Baker M, Dickson DW, Parisi JE, Giannini C, Josephs KA, et al. Frontotemporal
dementia and parkinsonism associated with the IVS1+ 1G → A mutation in progranulin: a
clinicopathologic study. Brain. 2006;129(11):3103–14.
Boeve BF, Boylan KB, Graff-Radford NR, DeJesus-Hernandez M, Knopman DS, Pedraza O, et al.
Characterization of frontotemporal dementia and/or amyotrophic lateral sclerosis associated with
the GGGGCC repeat expansion in C9ORF72. Brain. 2012;135(Pt 3):765–83. Epub 2012/03/01.
Caviness JN, Wszolek ZK. Myoclonus in pallido-ponto-nigral degeneration. Adv Neurol.
2002;89:35–9.
Claassen DO, Parisi JE, Giannini C, Boeve BF, Dickson DW, Josephs KA. Frontotemporal demen-
tia mimicking dementia with Lewy bodies. Cogn Behav Neurol. 2008;21(3):157–63.
Clinical and neuropathological criteria for frontotemporal dementia. The Lund and Manchester
Groups. J Neurol Neurosurg Psychiatry. 1994;57(4):416–8.
Cooper-Knock J, Frolov A, Highley JR, Charlesworth G, Kirby J, Milano A, et al. C9ORF72
expansions, parkinsonism, and Parkinson disease: a clinicopathologic study. Neurology.
2013;81(9):808–11. Epub 2013/07/26.
9 Movement Disorders in Frontotemporal Dementia 151
Kertesz A, McMonagle P, Blair M, Davidson W, Munoz DG. The evolution and pathology of fron-
totemporal dementia. Brain. 2005;128(Pt 9):1996–2005.
Kodama K, Okada S, Iseki E, Kowalska A, Tabira T, Hosoi N, et al. Familial frontotemporal
dementia with a P301L tau mutation in Japan. J Neurol Sci. 2000;176(1):57–64. Epub
2000/06/24.
Le Ber I, Van Der Zee J, Hannequin D, Gijselinck I, Campion D, Puel M, et al. Progranulin null muta-
tions in both sporadic and familial frontotemporal dementia. Hum Mutat. 2007;28(9):846–55.
Lendon CL, Lynch T, Norton J, McKeel Jr DW, Busfield F, Craddock N, et al. Hereditary dyspha-
sic disinhibition dementia: a frontotemporal dementia linked to 17q21-22. Neurology.
1998;50(6):1546–55.
Lillo P, Hodges JR. Frontotemporal dementia and motor neurone disease: overlapping clinic-
pathological disorders. J Clin Neurosci. 2009;16(9):1131–5.
Mann DM, South PW, Snowden JS, Neary D. Dementia of frontal lobe type: neuropathology and
immunohistochemistry. J Neurol Neurosurg Psychiatry. 1993;56(6):605–14.
Mathew R, Bak TH, Hodges JR. Diagnostic criteria for corticobasal syndrome: a comparative
study. J Neurol Neurosurg Psychiatry. 2012;83(4):405–10.
Mioshi E, Bristow M, Cook R, Hodges JR. Factors underlying caregiver stress in frontotemporal demen-
tia and Alzheimer's disease. Dement Geriatr Cogn Disord. 2009;27(1):76–81. Epub 2009/01/22.
Neary D, Snowden JS, Gustafson L, Passant U, Stuss D, Black S, et al. Frontotemporal lobar
degeneration: a consensus on clinical diagnostic criteria. Neurology. 1998;51(6):1546–54.
Newsway V, Fish M, Rohrer JD, Majounie E, Williams N, Hack M, et al. Perry syndrome due to
the DCTN1 G71R mutation: a distinctive levodopa responsive disorder with behavioral syn-
drome, vertical gaze palsy, and respiratory failure. Mov Disord. 2010;25(6):767–70.
Nuytemans K, Bademci G, Kohli MM, Beecham GW, Wang L, Young JI, et al. C9ORF72 interme-
diate repeat copies are a significant risk factor for Parkinson disease. Ann Hum Genet.
2013;77(5):351–63.
Onari KSH.Anatomishe Beitrage zur Lehre von der Pickschen umschriebenen Grosshirnrinden-Atrophie
(‘Picksche Krankheit’). Z Gesamte Neurol Psych. 1926;101:470–511.
Padovani A, Agosti C, Premi E, Bellelli G, Borroni B. Extrapyramidal symptoms in frontotempo-
ral dementia: prevalence and clinical correlations. Neurosci Lett. 2007;422(1):39–42.
Pickering-Brown SM, Rollinson S, Du Plessis D, Morrison KE, Varma A, Richardson AM, et al.
Frequency and clinical characteristics of progranulin mutation carriers in the Manchester fron-
totemporal lobar degeneration cohort: comparison with patients with MAPT and no known
mutations. Brain. 2008;131(Pt 3):721–31.
Piggott MA, Perry EK, Marshall EF, McKeith IG, Johnson M, Melrose HL, et al. Nigrostriatal
dopaminergic activities in dementia with Lewy bodies in relation to neuroleptic sensitivity:
comparisons with Parkinson’s disease. Biol Psychiatry. 1998;44(8):765–74.
Piguet O, Brooks WS, Halliday GM, Schofield PR, Stanford PM, Kwok JB, et al. Similar early
clinical presentations in familial and non-familial frontotemporal dementia. J Neurol Neurosurg
Psychiatry. 2004;75(12):1743–5. Epub 2004/11/19.
Pijnenburg Y, Sampson E, Harvey R, Fox N, Rossor M. Vulnerability to neuroleptic side effects in
frontotemporal lobar degeneration. Int J Geriatr Psychiatry. 2003;18(1):67–72.
Poorkaj P, Grossman M, Steinbart E, Payami H, Sadovnick A, Nochlin D, et al. Frequency of tau
gene mutations in familial and sporadic cases of non-Alzheimer dementia. Arch Neurol.
2001;58(3):383–7.
Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH, Neuhaus J, et al. Sensitivity of
revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain.
2011;134(Pt 9):2456–77. Epub 2011/08/04.
Ratnavalli E, Brayne C, Dawson K, Hodges JR. The prevalence of frontotemporal dementia.
Neurology. 2002;58(11):1615–21. Epub 2002/06/12.
Reed LA, Wszolek ZK, Hutton M. Phenotypic correlations in FTDP-17. Neurobiol Aging.
2001;22(1):89–107. Epub 2001/02/13.
Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, et al. A hexanucleo-
tide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD.
Neuron. 2011;72(2):257–68.
9 Movement Disorders in Frontotemporal Dementia 153
Rinne JO, Laine M, Kaasinen V, Norvasuo-Heila MK, Nagren K, Helenius H. Striatal dopamine
transporter and extrapyramidal symptoms in frontotemporal dementia. Neurology. 2002;58(10):
1489–93.
Rohrer JD, Ridgway GR, Modat M, Ourselin S, Mead S, Fox NC, et al. Distinct profiles of brain
atrophy in frontotemporal lobar degeneration caused by progranulin and tau mutations.
Neuroimage. 2010a;53(3):1070–6.
Rohrer JD, Rossor MN, Warren JD. Apraxia in progressive nonfluent aphasia. J Neurol. 2010b;
257(4):569–74.
Rosso SM, Donker Kaat L, Baks T, Joosse M, de Koning I, Pijnenburg Y, et al. Frontotemporal
dementia in The Netherlands: patient characteristics and prevalence estimates from a
population-based study. Brain. 2003;126(Pt 9):2016–22.
Saito Y, Geyer A, Sasaki R, Kuzuhara S, Nanba E, Miyasaka T, et al. Early-onset, rapidly progres-
sive familial tauopathy with R406W mutation. Neurology. 2002;58(5):811–3.
Schroeter ML, Raczka K, Neumann J, von Cramon DY. Neural networks in frontotemporal demen-
tia–a meta-analysis. Neurobiol Aging. 2008;29(3):418–26.
Seelaar H, Schelhaas HJ, Azmani A, Küsters B, Rosso S, Majoor-Krakauer D, et al. TDP-43
pathology in familial frontotemporal dementia and motor neuron disease without progranulin
mutations. Brain. 2007;130(5):1375–85.
Sha S, Hou C, Viskontas IV, Miller BL. Are frontotemporal lobar degeneration, progressive supra-
nuclear palsy and corticobasal degeneration distinct diseases? Nat Clin Pract Neurol.
2006;2(12):658–65. Epub 2006/11/23.
Sha SJ, Takada LT, Rankin KP, Yokoyama JS, Rutherford NJ, Fong JC, et al. Frontotemporal
dementia due to C9ORF72 mutations: clinical and imaging features. Neurology. 2012;79(10):
1002–11. Epub 2012/08/10.
Snowden JS, Rollinson S, Thompson JC, Harris JM, Stopford CL, Richardson AM, et al. Distinct
clinical and pathological characteristics of frontotemporal dementia associated with C9ORF72
mutations. Brain. 2012;135(Pt 3):693–708. Epub 2012/02/04.
Soliveri P, Piacentini S, Girotti F. Limb apraxia in corticobasal degeneration and progressive supra-
nuclear palsy. Neurology. 2005;64(3):448–53.
Spina S, Murrell JR, Yoshida H, Ghetti B, Bermingham N, Sweeney B, et al. The novel Tau muta-
tion G335S: clinical, neuropathological and molecular characterization. Acta Neuropathol.
2007;113(4):461–70.
Stanford PM, Halliday GM, Brooks WS, Kwok JB, Storey CE, Creasey H, et al. Progressive supra-
nuclear palsy pathology caused by a novel silent mutation in exon 10 of the tau gene: expansion
of the disease phenotype caused by tau gene mutations. Brain. 2000;123(Pt 5):880–93. Epub
2000/04/25.
Stanford PM, Brooks WS, Teber ET, Hallupp M, McLean C, Halliday GM, et al. Frequency of tau
mutations in familial and sporadic frontotemporal dementia and other tauopathies. J Neurol.
2004;251(9):1098–104.
Strong MJ. The syndromes of frontotemporal dysfunction in amyotrophic lateral sclerosis.
Amyotroph Lateral Scler. 2008;9(6):323–38.
Tsuboi Y. Neuropathology of familial tauopathy. Neuropathology. 2006;26(5):471–4. Epub
2006/11/04.
Tsuboi Y, Baker M, Hutton ML, Uitti RJ, Rascol O, Delisle MB, et al. Clinical and genetic studies
of families with the tau N279K mutation (FTDP-17). Neurology. 2002;59(11):1791–3. Epub
2002/12/11.
Van Swieten JC, Heutink P. Mutations in progranulin (<i>GRN</i>) within the spectrum of clinical
and pathological phenotypes of frontotemporal dementia. Lancet Neurol. 2008;7(10):965–74.
Van Swieten J, Stevens M, Rosso S, Rizzu P, Joosse M, De Koning I, et al. Phenotypic variation in
hereditary frontotemporal dementia with tau mutations. Ann Neurol. 1999;46(4):617–26.
Whitwell JL, Weigand SD, Boeve BF, Senjem ML, Gunter JL, DeJesus-Hernandez M, et al.
Neuroimaging signatures of frontotemporal dementia genetics: C9ORF72, tau, progranulin
and sporadics. Brain. 2012;135(Pt 3):794–806. Epub 2012/03/01.
Wszolek ZK, Uitti RJ, Hutton M. A mutation in the microtubule-associated protein tau in
pallido-nigro-luysian degeneration. Neurology. 2000;54(10):2028–30. Epub 2000/05/24.
Chapter 10
Dementia with Lewy Bodies
A.-C. Vijverman, MD
Department of Movement Disorders (Neurology),
Toronto Western Hospital, Toronto, ON, Canada
C. Tartaglia, MD, FRCPC
Memory Clinic (Neurology), Toronto Western Hospital,
Toronto, ON, Canada
S. Fox, MRCP, PhD (*)
Movement Disorders (Neurology), Toronto Western Hospital,
Toronto, ON, Canada
e-mail: [email protected]
Keywords Dementia with Lewy bodies (DLB) • Lewy body disease (LBD) • Lewy
bodies (LB) • Movement disorders • Dementia • Neuroleptic sensitivity •
Alpha-synuclein (αSyn) • Hallucinations • Cognitive fluctuations
Abbreviations
Introduction
Dementia with Lewy bodies (DLB) is a clinical syndrome characterized by slowly pro-
gressive cognitive, neuropsychiatric, and motor decline. Frederick Lewy was the first to
report abnormal intracytoplasmic protein deposits in a patient with Parkinson’s disease
10 Dementia with Lewy Bodies 157
(PD) in 1912, later called “Lewy body inclusions.” The relationship between the neuro-
pathological finding of cortical Lewy bodies (LBs) and dementia was described almost
40 years later by Okazaki et al. (1961), while it was not until 1984 that Kosaka et al. first
used the term “diffuse Lewy body disease” to describe a new disease entity consisting
of presenile dementia with presence of cortical LBs (Huang and Halliday 2013). Since
then, confusion has arisen as many different names have been used to describe this dis-
order, including diffuse Lewy body disease, Lewy body dementia, senile dementia of
Lewy body type, dementia with Lewy bodies, Lewy body variant of Alzheimer’s dis-
ease, dementia associated with cortical Lewy bodies, and Lewy body disease. In this
chapter, we will use the term “dementia with Lewy bodies” when describing the clinical
syndrome and “Lewy body disease” (LBD) when referring to the pathological disease.
This is in agreement with the 1996 consortium workshop on DLB that published the first
clinical and pathological diagnostic criteria (McKeith et al. 1996). The criteria were
revised in 1999 and 2005, with further clinicopathological correlation studies demon-
strating a need for refinement due to a low sensitivity – though high specificity – of the
previous diagnostic consensus criteria (McKeith et al. 2005). Following the criteria of
2005, a definite diagnosis is reserved for cases with pathological confirmation, whereas
possible and probable DLB are diagnosed in vivo.
DLB shows significant clinical and pathological overlap with other neurodegenera-
tive diseases, including Alzheimer’s disease (AD) and Parkinson’s disease with demen-
tia (PDD). In particular, DLB and PDD are likely the same disorder but with variable
clinical presentation. Many patients with DLB and PDD will have Alzheimer patho-
logical changes at postmortem, thus adding to the clinicopathological confusion.
Epidemiology
Clinical Features
Table 10.1 Clinical consensus criteria for the diagnosis of dementia with Lewy bodies (McKeith
et al. 2005)
Clinical features
Central feature: dementia
Core features
Spontaneous parkinsonian motor signs
Visual hallucinations
Fluctuations with pronounced variations in attention and alertness
Suggestive features
REM sleep behavior disorder
Severe neuroleptic sensitivity
Low dopamine transporter uptake in basal ganglia on SPECT/PET imaging
Supportive features
Repeated falls and syncope
Transient, unexplained loss of consciousness
Severe autonomic dysfunction (orthostatic hypotension, urinary incontinence)
Hallucinations in other modalities
Systematized delusions
Depression
Relative preservation of medial temporal lobe structures on CT/MRI scan
Generalized low uptake and reduced occipital activity on SPECT/PET
Abnormal (low uptake) MIBG myocardial scintigraphy
Prominent slow wave activity and temporal lobe transient sharp waves on EEG
Clinical Diagnosis
Definite DLB: neuropathological confirmation of LBD
Probable DLB
Dementia + 2/3 core features
or
Dementia + 1 core feature + 1 suggestive feature
Possible DLB
Dementia + 1 core feature
or
Dementia + ≥1 suggestive feature
REM rapid eye movements, SPECT single-photon emission computerized tomography, PET
positron-emission tomography, MRI magnetic resonance imaging, CT computerized tomography,
MIBG 123I-metaiodobenzylguanidine, EEG electroencephalography, DLB dementia with Lewy
bodies, LBD Lewy body disease
diagnostic criteria (McKeith et al. 2005) (Table 10.1), the key feature of DLB is
dementia, defined by a cognitive decline substantial enough to interfere with activi-
ties of daily living. The core features of DLB are cognitive fluctuations, visual hal-
lucinations, and spontaneous parkinsonism. Supportive features are REM sleep
behavior disorders (RBD), severe neuroleptic sensitivity, and low dopamine trans-
porter uptake in basal ganglia on single-photon emission computerized tomography
(SPECT) or on positron-emission tomography (PET). Additional suggestive symp-
toms are frequently associated with DLB, although aspecific, and are therefore not
part of the diagnostic criteria.
10 Dementia with Lewy Bodies 159
Motor Manifestations
Spontaneous parkinsonism that is not drug induced is a core feature in the clinical
diagnosis of DLB. Symptoms occur with ~50 % loss of nigrostriatal dopaminergic
neurons (Fearnley and Lees 1991) and are described in up to 50 % of patients with
DLB at diagnosis (McKeith et al. 2004). The parkinsonian features in DLB (Table 10.2)
are more symmetric at onset when compared to the unilateral onset seen in idiopathic
PD (Gnanalingham et al. 1997; Del Ser et al. 2000). The axial bradykinetic-rigid sub-
type, with postural instability and gait difficulties (PIGD), is encountered more fre-
quently than the tremor-dominant (TD) subtype (McKeith et al. 1996; Galasko et al.
1996; Jankovic et al. 1990). Burn et al. found in a cross-sectional study of 107 patients
that 69 % of DLB patients present the PIGD subtype as opposed to 38 % in PD, where
the tremor-dominant (TD) subtype is much more prevalent (Burn et al. 2003).
Extrapyramidal signs and symptoms (EPS) may also occur in later stages of AD
(Lopez et al. 1997). Although not clinicopathologically correlated, Kaur et al. ana-
lyzed data from the US National Alzheimer’s Coordinating Center (Kaur et al.
2013) of 1,826 patients with AD and 130 patients with DLB, characterizing EPS in
both diseases (Table 10.2). Hypomimia and hypophonia occurred in 4.2 % of the
AD group as opposed to more than 48 % in DLB. Likewise, bradykinesia, rigidity,
impaired posture, and postural instability were significantly more prevalent in DLB
when compared to AD (55 % versus 19.3 %). The prevalence of tremor was also
significantly higher in DLB than in AD, 13.9 % versus 3.2 % respectively for resting
tremor in DLB and AD and 12.3 % versus 6.2 % for action/postural tremor.
Myoclonus is a common sign of many dementias. Up to 35 % of patients with DLB
will develop myoclonus (Tartaglia et al. 2012), predominantly occurring in upper
limbs, induced by action and posture but seldom observed in the resting position
(Weiner and Tolosa 2011). It has clinical and electrophysiological characteristics of
cortical myoclonus (Weiner and Tolosa 2011). When associated with rapid cognitive
decline and hallucinations, the Heidenhain variant of sCJD may need to be considered.
Distinguishing diagnostic features in DLB are absence of cortical ribonning on brain
magnetic resonance imaging (MRI), cerebrospinal fluid (CSF) 14-3-3 negativity, and
less common periodic electroencephalographic (EEG) changes (Tartaglia et al. 2012).
Non-motor Manifestations
Cognitive Manifestations
DLB AD PD PDD
Extrapyramidal symptoms Often more symmetric Rare, usually mild in late Initially often asymmetric Initially often asymmetric
stages
PIGD subtypea 69 % – 38 % 88 %
TD subtypea 31 % – 62 % 12 %
Hypomimiab 48.5 % 4.2 %
Hypophoniab 30.8 % 3.4 %
Rigidityb 44.6 % 9.8 %
Impaired posture/gaitb 43.1 % 14.2 %
Bradykinesiab 55.4 % 19.3 %
Impaired chair riseb 28.9 % 15.4 %
Postural instabilityb 26.2 % 12.5 %
Resting tremorb 13.9 % 3.2 %
Action/postural tremorb 12.3 % 6.2 %
Cognitive impairment Early disturbances in attention Early impairment of Impaired executive and Impaired executive and
and visuoperceptive declarative memory visuoperceptive visuoperceptive functions
functions and attention functions
Fluctuations in cognition Prominent, early second to Moderate day to day Mild day to day variations Mild day to day variations
hourly variations variations
Neuropsychiatric symptoms
Visual hallucinations Typical, early, and persistent Sometimes, late course Often present with Often present with anticholin-
anticholinergic ergic dopaminergic drugs
dopaminergic drugs
Delusions Typical Usually present Present Present
Depression Usually present Usually present Usually present Usually present
DLB Dementia with Lewy bodies, AD Alzheimer’s Disease, PD Parkinson’s Disease, PDD Parkinson’s Disease with dementia, PIGD Postural instability and
gait difficulties, TD tremor dominant
a
Prevalence in cross-sectional study of Burn et al. (2003)
b
Prevalence in cross-sectional study of Kaur et al. (2013)
A.-C. Vijverman et al.
10 Dementia with Lewy Bodies 161
striking, so that even with mild cognitive impairment patients seem to be unable to
draw a clock, to copy overlocking pentagons, or to navigate (Cormack et al. 2004).
This neuropsychological finding sometimes allows discrimination with AD, cer-
tainly in the early stages (Yoshizawa et al. 2013). Language is relatively preserved,
although dysprosody is often seen. Dementia characteristics of DLB and PDD are
very similar, and the distinction is mainly made based on the arbitrary 1-year rule
(McKeith et al. 1996). By definition, patients developing dementia prior to parkin-
sonism or during the first year of disease are diagnosed with DLB. In PDD, the
onset of motor symptoms precedes dementia by at least 1 year, generally occurring
only later in the disease course, but encountered in up to 80 % of all PD patients
during the course of their disease (Aarsland et al. 2003).
Fluctuations in attention and alertness are considered part of DLB’s core fea-
tures according to the diagnostic consensus criteria (McKeith et al. 2005). As
opposed to AD, the transient fluctuating episodes last seconds to hours and may
consist of staring spells, confusion, episodes of decreased attention, or disorga-
nized speech with sometimes a confabulatory or delusional quality. The level of
cognition returns to near normal after those spontaneous fluctuations in awareness
that are generally not triggered by discernible environmental factors. EEG shows
variability in theta rhythm during the spells (Yamamoto and Imai 1988; Doran and
Larner 2004). There can also be a loss of alpha activity and occurrence of slow
wave transient activity in the temporal lobes (Briel et al. 1999). In AD, fluctua-
tions also are a common feature, although less prevalent, and they consist of more
daily variations.
Neuropsychiatric Manifestations
Visual hallucinations (VH), misperceptions, and mood alterations are some of the
frequently encountered neuropsychiatric manifestations of DLB. Spontaneous VH
are a core feature of DLB, often present early in the disease, recurring daily and
lasting minutes at a time (Mosimann et al. 2006). The phenomenology of the VH is
typically of 3-dimensional, detailed mute animate subjects in the central field of
vision, similar as in PDD, although dopaminergic medications are frequently the
trigger in the latter. Auditory and tactile hallucinations are much less common but
can occur. In both DLB and PDD, medical causes of delirium should always be
excluded as a trigger. The VH in DLB might be related to alterations in cortical
acetylcholine together with abnormalities in brainstem and visual systems
(Collerton et al. 2005). This is corroborated by the reduction of visual hallucina-
tions that occurs in DLB when cholinesterase inhibitors (AChI) are started (Barber
et al. 2000; Burn et al. 2006). It remains unclear if there is a relationship between
the occurrence of visual hallucinations and the occipital hypometabolism found in
DLB (Imamura et al. 1999).
Misperceptions, characterized by seeing existing objects as animate subjects, are
frequently encountered as well. Other psychotic symptoms may occur in DLB,
including paranoid delusions, often of spouse infidelity and fear of strangers in the
162 A.-C. Vijverman et al.
home. Common mood alterations in DLB include depression and anxiety, although
not specific for DLB (Borroni et al. 2008).
Hypersensitivity to Neuroleptics
Autonomic Manifestations
Autonomic postganglionic neurons are probably one of the earliest regions affected by LB
pathology, but autonomic failure doesn’t occur within the first year of disease (Wenning
et al. 1999). Urinary incontinence and orthostatic hypotension are the most common
symptoms, but adrenergic dysfunction (an impaired pressor response to acute sympathetic
stimulation in physical stress), distal anhidrosis, and urinary urgency or retention are also
frequently seen. Dopaminergic agents can exacerbate signs and symptoms of postural
hypotension and should be stopped unless providing a significant benefit.
Sleep
Pathophysiology
published to date. Such differences arise from variable referral bases, e.g., nursing
homes versus community-based referrals. Thus, there still remains controversy
around the pathological phenotype of dementias in parkinsonian syndromes.
Neurochemical Changes
Diagnostic Tests
There are no objective diagnostic tests available to confirm the clinical diagnosis of
DLB. Currently, the diagnosis is based on clinical assessment and examination in
vivo. Some imaging modalities may assist in the differentiation from other dementia
or parkinsonian syndromes.
Brain MRI, for example, does not show any DLB-specific structural findings, but
the degree of medial temporal lobe atrophy and hippocampal atrophy is less pro-
nounced than in AD on volumetric MRI measures (Barber et al. 2000). Also, MRI
166 A.-C. Vijverman et al.
Management
Pharmacological
Parkinsonism
Rigidity and bradykinesia respond less to levodopa therapy in DLB than in idio-
pathic PD (Molloy et al. 2005; Bonelli et al. 2004), with a benefit in only one third
(Molloy et al. 2005; Bonelli et al. 2004). Moreover, DLB patients are more sensitive
to the side effects of dopaminergic agents, especially dopamine agonists, and can
cause or worsen neuropsychiatric side effects (Molloy et al. 2005). Strategies for
decreasing the incidence of adverse events include starting levodopa at lower doses
and increasing more slowly than in PD. The trial should be longer if well tolerated
as often drugs take several weeks to take effect. Given the predisposition to side
effects and the sensitivity to interactions, the treatment should be stopped if no obvi-
ous clinical response has been observed.
Table 10.3 Therapeutic strategies in dementia with Lewy bodies
168
Treatment options
Symptoms Treatment options Details Comments
Parkinsonism Non-pharmacological Physiotherapy and physical exercise Balance, gait, arm swing
Pharmacological
Levodopa Start very slow (50 mg BID or TID), increase Continue several weeks if well tolerated; stop if no
slowly per ½ tablets apparent benefit (30 % responders).
Side effects: (transient) gastrointestinal upset, visual
hallucinations, delusions, orthostatic hypotension
Dopamine agonists Relatively contraindicated because of risk of worsening
behavioral problems
Anticholinergics Strictly contraindicated because of high risk of increased
cognitive and neuropsychiatric symptoms
Cognitive Non-pharmacological Modifying stressors in the environment. –
impairment Physiotherapy
Active social interactions
Pharmacological
Cholinesterase inhibitors
Rivastigmine Start 1.5 mg BID, increase by 1.5 mg every Side effects: worsening parkinsonism (in 10 %: tremor),
2–4 weeks, maximum 6 mg BID (transient) gastrointestinal upset, cardiac arrhythmia
Donepezil Start 5 mg OD for 4 weeks, then increase to Side effects: gastrointestinal (less with slow increase),
10 mg OD (transient) gastrointestinal upset, cardiac arrhythmia
Galantamine 8 mg OD, increase slowly to max 24 mg OD Effective and well tolerated in a 24-week open-label
study, but no RCTs
Memantine Start at 5 mg OD and increase to 10 mg BID Well tolerated
A.-C. Vijverman et al.
10
Neuropsychiatric symptoms
Visual Non-pharmacological No treatment if not harmful or disturbing –
hallucinations Pharmacological
Atypical neuroleptics
Quetiapine Start at 12.5 mg OD, increase slowly if necessary Sedation
Clozapine Start at 12.5 mg OD, increase per 12.5 mg Sedation
if necessary, max 50 mg OD
Typical neuroleptics – Strictly contraindicated
Depression Non-pharmacological Modifying stressors in the environment –
Pharmacological
Dementia with Lewy Bodies
Treatment options
Symptoms Treatment options Details Comments
RBD Non-pharmacological No treatment if not harmful or disturbing –
Pharmacological
Clonazepam Start 0.25 mg QHS, increase per 0.25 mg Side effects: sedation
to maximum 1 mg QHS
Melatonin Start 3 mg QHS, slowly increase per 3 mg to –
maximum 12 mg QHS
Treatment strategy
Start pharmacological treatment only if symptoms are harmful or disturbing and if non-pharmacological strategies are insufficient
Start low, go slow
Stop treatment if no apparent benefit
Anticholinergics, TCA, and typical neuroleptics are strictly contraindicated
Avoid general anesthesia (Surgery necessary? Possible locoregional anesthesia?) or reduce length and strength of general anesthesia
SSRI selective serotonin reuptake inhibitors, TCA tricyclic antidepressant, EDS excessive daytime sleepiness, RBD REM sleep behavior disorder, RCT randomized con-
trolled trial
A.-C. Vijverman et al.
10 Dementia with Lewy Bodies 171
Autonomic Impairment
Sleep
Non-pharmacological
Although there are no randomized controlled trials or cohort studies evaluating the
benefit of non-pharmacological measures in DLB, such approaches may benefit cog-
nitive and physical skills. The patient should be encouraged to take part in social
activities to maintain active social interactions, stimulating the patient’s arousal and
improving nighttime sleep. On the other hand, as in PD, physical activity is highly
recommended (Goodwin et al. 2008; Keus et al. 2007). Since DLB generally shows
more axial than appendicular EPS, exercises of balance, walking, and arm swing
helps mobility and prolongs independency. Often physiotherapy is needed to help in
mobilizing the patients and teaching them easy exercises to perform on a daily basis.
References
Aarsland D, et al. Prevalence and characteristics of dementia in Parkinson disease: an 8-year pro-
spective study. Arch Neurol. 2003;60(3):387–92.
Aarsland D, et al. Memantine in patients with Parkinson’s disease dementia or dementia with
Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8(7):
613–8.
10 Dementia with Lewy Bodies 173
Barber R, et al. MRI volumetric study of dementia with Lewy bodies: a comparison with AD and
vascular dementia. Neurology. 2000;54(6):1304–9.
Barker WW, et al. Relative frequencies of Alzheimer disease, Lewy body, vascular and frontotem-
poral dementia, and hippocampal sclerosis in the State of Florida Brain Bank. Alzheimer Dis
Assoc Disord. 2002;16(4):203–12.
Bennett DA, et al. Neuropathology of older persons without cognitive impairment from two
community-based studies. Neurology. 2006;66(12):1837–44.
Boeve BF, Silber MH, Ferman TJ. Melatonin for treatment of REM sleep behavior disorder in
neurologic disorders: results in 14 patients. Sleep Med. 2003;4(4):281–4.
Boeve BF, Silber MH, Ferman TJ. REM sleep behavior disorder in Parkinson’s disease and demen-
tia with Lewy bodies. J Geriatr Psychiatry Neurol. 2004;17(3):146–57.
Boeve BF, et al. Pathophysiology of REM sleep behaviour disorder and relevance to neurodegen-
erative disease. Brain. 2007;130(Pt 11):2770–88.
Bogaerts V, et al. A novel locus for dementia with Lewy bodies: a clinically and genetically het-
erogeneous disorder. Brain. 2007;130(Pt 9):2277–91.
Bonelli SB, et al. L-dopa responsiveness in dementia with Lewy bodies, Parkinson disease with
and without dementia. Neurology. 2004;63(2):376–8.
Borroni B, Agosti C, Padovani A. Behavioral and psychological symptoms in dementia with
Lewy-bodies (DLB): frequency and relationship with disease severity and motor impairment.
Arch Gerontol Geriatr. 2008;46(1):101–6.
Braak H, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol
Aging. 2003;24(2):197–211.
Briel RC, et al. EEG findings in dementia with Lewy bodies and Alzheimer’s disease. J Neurol
Neurosurg Psychiatry. 1999;66(3):401–3.
Burn DJ, et al. Extrapyramidal features in Parkinson’s disease with and without dementia and
dementia with Lewy bodies: a cross-sectional comparative study. Mov Disord. 2003;18(8):
884–9.
Burn D, et al. Effects of rivastigmine in patients with and without visual hallucinations in dementia
associated with Parkinson’s disease. Mov Disord. 2006;21(11):1899–907.
Byrne EJ, Burns A, Waite J. Neuroleptic sensitivity in dementia with cortical Lewy bodies. BMJ.
1992;305(6862):1158–9.
Chartier-Harlin MC, et al. Alpha-synuclein locus duplication as a cause of familial Parkinson’s
disease. Lancet. 2004;364(9440):1167–9.
Chung EJ, et al. MIBG scintigraphy for differentiating Parkinson’s disease with autonomic dysfunction
from Parkinsonism-predominant multiple system atrophy. Mov Disord. 2009;24(11):1650–5.
Collerton D, Perry E, McKeith I. Why people see things that are not there: a novel Perception and
Attention Deficit model for recurrent complex visual hallucinations. Behav Brain Sci.
2005;28(6):737–57; discussion 757–94.
Colloby S, O’Brien J. Functional imaging in Parkinson’s disease and dementia with Lewy bodies.
J Geriatr Psychiatry Neurol. 2004;17(3):158–63.
Cormack F, et al. Pentagon drawing and neuropsychological performance in Dementia with Lewy
Bodies, Alzheimer’s disease, Parkinson’s disease and Parkinson’s disease with dementia. Int J
Geriatr Psychiatry. 2004;19(4):371–7.
Costa DC, et al. Dementia with Lewy bodies versus Alzheimer’s disease: role of dopamine trans-
porter imaging. Mov Disord. 2003;18 Suppl 7:S34–8.
Dalfo E, et al. Abnormal metabotropic glutamate receptor expression and signaling in the cerebral
cortex in diffuse Lewy body disease is associated with irregular alpha-synuclein/phospholipase
C (PLCbeta1) interactions. Brain Pathol. 2004;14(4):388–98.
Del Ser T, et al. Dementia with Lewy bodies: findings from an international multicentre study. Int
J Geriatr Psychiatry. 2000;15(11):1034–45.
Dolder CR, Davis LN, McKinsey J. Use of psychostimulants in patients with dementia. Ann
Pharmacother. 2010;44(10):1624–32.
Doran M, Larner AJ. EEG findings in dementia with Lewy bodies causing diagnostic confusion
with sporadic Creutzfeldt-Jakob disease. Eur J Neurol. 2004;11(12):838–41.
174 A.-C. Vijverman et al.
Duda JE. Pathology and neurotransmitter abnormalities of dementia with Lewy bodies. Dement
Geriatr Cogn Disord. 2004;17 Suppl 1:3–14.
Emre M, et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med.
2004;351(24):2509–18.
Fearnley JM, Lees AJ. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain.
1991;114(Pt 5):2283–301.
Ferman TJ, et al. REM sleep behavior disorder and dementia: cognitive differences when com-
pared with AD. Neurology. 1999;52(5):951–7.
Fodero-Tavoletti MT, et al. In vitro characterization of Pittsburgh compound-B binding to Lewy
bodies. J Neurosci. 2007;27(39):10365–71.
Francis PT, Perry EK. Cholinergic and other neurotransmitter mechanisms in Parkinson’s disease,
Parkinson’s disease dementia, and dementia with Lewy bodies. Mov Disord. 2007;22 Suppl
17:S351–7.
Friedman JH, Fernandez HH. Atypical antipsychotics in Parkinson-sensitive populations. J Geriatr
Psychiatry Neurol. 2002;15(3):156–70.
Galasko D, et al. Clinical and neuropathological findings in Lewy body dementias. Brain Cogn.
1996;31(2):166–75.
Gnanalingham KK, et al. Motor and cognitive function in Lewy body dementia: comparison with
Alzheimer’s and Parkinson’s diseases. J Neurol Neurosurg Psychiatry. 1997;62(3):243–52.
Gomperts SN, et al. Brain amyloid and cognition in Lewy body diseases. Mov Disord.
2012;27(8):965–73.
Goodwin VA, et al. The effectiveness of exercise interventions for people with Parkinson’s disease:
a systematic review and meta-analysis. Mov Disord. 2008;23(5):631–40.
Haik S, et al. Dementia with Lewy bodies in a neuropathologic series of suspected Creutzfeldt-
Jakob disease. Neurology. 2000;55(9):1401–4.
Halliday GM, et al. Neuropathology of immunohistochemically identified brainstem neurons in
Parkinson’s disease. Ann Neurol. 1990;27(4):373–85.
Hu XS, et al. 18 F-fluorodopa PET study of striatal dopamine uptake in the diagnosis of dementia
with Lewy bodies. Neurology. 2000;55(10):1575–7.
Huang Y, Halliday G. Can we clinically diagnose dementia with Lewy bodies yet? Transl
Neurodegener. 2013;2(1):4.
Imamura T, et al. Visual hallucinations and regional cerebral metabolism in dementia with Lewy
bodies (DLB). Neuroreport. 1999;10(9):1903–7.
Jankovic J, et al. Variable expression of Parkinson’s disease: a base-line analysis of the DATATOP
cohort. The Parkinson Study Group. Neurology. 1990;40(10):1529–34.
Kaur B, et al. Extrapyramidal signs by dementia severity in Alzheimer disease and dementia with
Lewy bodies. Alzheimer Dis Assoc Disord. 2013;27:226–32.
Keus SH, et al. Evidence-based analysis of physical therapy in Parkinson’s disease with recom-
mendations for practice and research. Mov Disord. 2007;22(4):451–60; quiz 600.
Klunk WE, et al. Two-year follow-up of amyloid deposition in patients with Alzheimer’s disease.
Brain. 2006;129(Pt 11):2805–7.
Lobotesis K, et al. Occipital hypoperfusion on SPECT in dementia with Lewy bodies but not AD.
Neurology. 2001;56(5):643–9.
Lopez OL, et al. Extrapyramidal signs in patients with probable Alzheimer disease. Arch Neurol.
1997;54(8):969–75.
Mata IF, et al. Glucocerebrosidase gene mutations: a risk factor for Lewy body disorders. Arch
Neurol. 2008;65(3):379–82.
McKeith I, et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ.
1992;305(6855):673–8.
McKeith IG, et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with
Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology.
1996;47(5):1113–24.
McKeith I, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-
blind, placebo-controlled international study. Lancet. 2000;356(9247):2031–6.
10 Dementia with Lewy Bodies 175
Ridha BH, Josephs KA, Rossor MN. Delusions and hallucinations in dementia with Lewy bodies:
worsening with memantine. Neurology. 2005;65(3):481–2.
Rolinski M, et al. Cholinesterase inhibitors for dementia with Lewy bodies. Parkinson’s disease
dementia and cognitive impairment in Parkinson’s disease. Cochrane Database Syst Rev.
2012;3, CD006504.
Samuel W, et al. Better cognitive and psychopathologic response to donepezil in patients prospec-
tively diagnosed as dementia with Lewy bodies: a preliminary study. Int J Geriatr Psychiatry.
2000;15(9):794–802.
Scatton B, et al. Reduction of cortical dopamine, noradrenaline, serotonin and their metabolites in
Parkinson’s disease. Brain Res. 1983;275(2):321–8.
Schellenberg GD, et al. Association of an apolipoprotein CII allele with familial dementia of the
Alzheimer type. J Neurogenet. 1987;4(2–3):97–108.
Schenck CH, et al. Chronic behavioral disorders of human REM sleep: a new category of parasom-
nia. 1986, classical article. Sleep. 2002;25(2):293–308.
Sechi G, et al. Acute hyponatremia and neuroleptic malignant syndrome in Parkinson’s disease.
Prog Neuropsychopharmacol Biol Psychiatry. 1996;20(3):533–42.
Shiozaki K, et al. Alterations of muscarinic acetylcholine receptor subtypes in diffuse Lewy body
disease: relation to Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1999;67(2):209–13.
Sidransky E, Lopez G. The link between the GBA gene and parkinsonism. Lancet Neurol.
2012;11(11):986–98.
Siepel FJ, et al. (123I)FP-CIT SPECT in suspected dementia with Lewy bodies: a longitudinal case
study. BMJ Open. 2013;3(4). pii: e002642. doi: 10.1136/bmjopen-2013-002642.
Singleton AB, et al. Alpha-synuclein locus triplication causes Parkinson’s disease. Science.
2003;302(5646):841.
Spina E, Scordo MG. Clinically significant drug interactions with antidepressants in the elderly.
Drugs Aging. 2002;19(4):299–320.
Suzuki M, et al. Impaired myocardial 123I-metaiodobenzylguanidine uptake in Lewy body dis-
ease: comparison between dementia with Lewy bodies and Parkinson’s disease. J Neurol Sci.
2006;240(1–2):15–9.
Taki J, et al. Significance of 123I-MIBG scintigraphy as a pathophysiological indicator in the
assessment of Parkinson’s disease and related disorders: it can be a specific marker for Lewy
body disease. Ann Nucl Med. 2004;18(6):453–61.
Tan A, Salgado M, Fahn S. Rapid eye movement sleep behavior disorder preceding Parkinson’s
disease with therapeutic response to levodopa. Mov Disord. 1996;11(2):214–6.
Tartaglia MC, et al. Clinical overlap between Jakob-Creutzfeldt disease and Lewy body disease.
Can J Neurol Sci. 2012;39(3):304–10.
Tateno F, et al. Alpha-synuclein in the cerebrospinal fluid differentiates synucleinopathies
(Parkinson Disease, dementia with Lewy bodies, multiple system atrophy) from Alzheimer
disease. Alzheimer Dis Assoc Disord. 2012;26(3):213–6.
Tsuang D, et al. APOE epsilon4 increases risk for dementia in pure synucleinopathies. JAMA
Neurol. 2013;70(2):223–8.
Walker Z, et al. Differentiation of dementia with Lewy bodies from Alzheimer’s disease using a
dopaminergic presynaptic ligand. J Neurol Neurosurg Psychiatry. 2002;73(2):134–40.
Wang CS, et al. Twin pairs discordant for neuropathologically confirmed Lewy body dementia.
J Neurol Neurosurg Psychiatry. 2009;80(5):562–5.
Weiner WJ, Tolosa E. Hyperkinetic movement disorders. Preface. Handb Clin Neurol. 2011;100:ix.
Wenning GK, et al. Time course of symptomatic orthostatic hypotension and urinary incontinence
in patients with postmortem confirmed parkinsonian syndromes: a clinicopathological study.
J Neurol Neurosurg Psychiatry. 1999;67(5):620–3.
Williams MM, et al. Survival and mortality differences between dementia with Lewy bodies vs
Alzheimer disease. Neurology. 2006;67(11):1935–41.
Yamamoto T, Imai T. A case of diffuse Lewy body and Alzheimer’s diseases with periodic syn-
chronous discharges. J Neuropathol Exp Neurol. 1988;47(5):536–48.
10 Dementia with Lewy Bodies 177
Ye L, et al. In vitro high affinity alpha-synuclein binding sites for the amyloid imaging agent PIB
are not matched by binding to Lewy bodies in postmortem human brain. J Neurochem.
2008;105(4):1428–37.
Yoshizawa H, Vonsattel JP, Honig LS. Early neuropsychological discriminants for Lewy body
disease: an autopsy series. J Neurol Neurosurg Psychiatry. 2013;84:1326–30.
Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidcence studies of
dementia with Lewy bodies. Age Ageing. 2005;34(6):561–6.
Chapter 11
Dementia in Parkinson’s Disease
and Atypical Parkinsonism
features, while hallucinations and delusions occur extremely rarely. Tau pathology
in the cortex and subcortical structures seems to correlate with these cognitive phe-
notypes; however, the understanding of the pathophysiology as well as treatment
options is limited. In contrast, dementia in multiple system atrophy (MSA) still
counts as one of the exclusion criteria for its diagnosis. Recent studies have high-
lighted that executive dysfunction does occur in MSA, but etiopathogenesis and
treatments are still to be researched.
Introduction
Age is the most prominent risk factor for PDD (Kempster et al. 2010; Levy et al.
2002; Williams-Gray et al. 2009) independently from the age of PD onset. PDD
also correlates with the severity of motor disability, and the aging factor may be
11 Dementia in Parkinson’s Disease and Atypical Parkinsonism 181
additive to the severity of the motor dysfunction (Levy et al. 2000, 2002). Other
factors such as visual hallucinations, and the PD phenotype (e.g., prominent axial
rigidity and bradykinesia as opposed to tremor), confer risk for the development of
PDD (Emre et al. 2007a; Jankovic and Poewe 2012; Galvin et al. 2006). Mild cogni-
tive impairment (MCI) has also been associated with an increased risk of develop-
ing PDD (Litvan et al. 2012; Levy et al. 2002).
Cognitive Deficits
The most common cognitive deficits in PDD include those in attention, executive
functioning, and visuospatial processing. Attentional performance may fluctuate,
leading to a variable level of function and a major impact upon activities of daily
living (Bronnick et al. 2006). In tests such as the letter cancelation test and others,
PDD patients are slower, tend to fluctuate more, and incur on a higher number of
errors than Alzheimer’s dementia (AD) patients, whereas the profile of PDD seems
to be similar to that seen in dementia with Lewy bodies (DLB). Executive dysfunc-
tion has been widely recognized in PDD and the most used tests have been verbal
fluency, digit span backward, Wisconsin Card Sorting Test, Stroop Test, and Trail
Making Test (Kudlicka et al. 2011). Studies assessing visuospatial function have
shown greater deficits in PDD than AD patients (Jeannerod and Jacob 2005). It has
been shown that visual perception, space-motion perception, and object-form per-
ception are globally more impaired in PDD patients than non-demented PD patients,
and AD patients (Mosimann et al. 2004).
In PDD, short-term memory is impaired, both for initial learning and immediate
recall. Traditionally, amnesic deficits in PD have been considered to be mainly of
retrieval, rather than encoding and storage. However, memory loss in PDD has been
associated more with a frontally mediated retrieval deficit thus again a deficit in
executive functioning, than to an intrinsic defect (Emre et al. 2007a). A large meta-
analysis has shown that verbal fluency impairment is more pronounced than that
seen in PD non-demented patients; also, semantic fluency seems to be more com-
promised than phonemic fluency (Henry and Crawford 2004). Significant language
deficits are not typically seen in PDD and occur much less frequently than in AD
(Pillon et al. 1993). When present, language deficits in PDD are more likely associ-
ated to executive dysfunction-related problems in sentence processing rather than
an intrinsic core language deficit (Grossman et al. 2012).
Neuropsychiatric Features
depression occur with approximately the same frequency in PDD and AD (40–
58 %) patients (Aarsland et al. 2001). Anxiety occurs at a similar frequency (30–
49 %) as depressed mood, and these symptoms may frequently coexist in the same
patient (Bronnick et al. 2007). Irritable mood, anger, and aggression are common in
AD, but uncommon in PDD (Engelborghs et al. 2005). Prominent apathy may occur
in PDD but also in other forms of dementia, including frontotemporal dementia,
progressive supranuclear palsy, AD, and DLB. Furthermore, some individuals may
exhibit an impulse control disorder that is characterized by compulsive gambling,
eating, purchasing, sexual behavior, and/or a dopamine dysregulation syndrome
(Weintraub 2008). The etiology of impulse control disorders in PD is thought to be
due to stimulation of hypersensitive ventral striatal-frontal connections by dopami-
nergic therapy rather than a direct consequence of the neurodegenerative process
that is specific to PD and PDD (Weintraub 2008; Weintraub et al. 2013).
Etiopathogenesis of PDD
Neuropathology
pathogenesis of PDD and a possible synergy with α-syn pathology (Masliah et al.
2001; Clinton et al. 2010). Indeed, clinically, patients with PDD and AD pathology
have shorter disease duration, older age at onset of motor symptoms, and shortened
survival times compared to PDD patients without concomitant AD pathology
(Compta et al. 2009, 2011; Irwin et al. 2012, 2013; Halliday and McCann 2010;
Halliday et al. 2008).
In summary, the progression of Lewy body and neurite pathology from subcorti-
cal areas into limbic and cortical structures seems to be the major determinant of the
development of dementia in most individuals with PDD; however, other pathologies
such as that of AD may be implicated in the underlying neuropathology of PDD.
Genetics
Genetic factors may also play an important role in the development of cognitive
impairment in PDD. Some monogenic forms of PD have been associated with
dementia such as those resulting from pathogenic mutations of the α-synuclein gene
(SNCA), whereas others such as mutations in leucine-rich repeat kinase 2 (LRRK2)
or the parkin gene do not seem to be as strongly linked to PDD and DLB (Poulopoulos
et al. 2012; Sidransky et al. 2009). Heterozygous mutations in the b-glucocerebrosidase
(GBA) gene are associated with an increased risk of PD or DLB, and GBA-linked
PD is associated with a higher risk and an earlier age of onset of dementia, as well
as higher levels of cortical and limbic α-syn pathology than noncarrier patients with
PD (Sidransky et al. 2009; Alcalay et al. 2012; Clark et al. 2009; Nalls et al. 2013;
Neumann et al. 2009; Tsuang et al. 2012). Moreover, polymorphisms of DYRK1A,
which encodes a kinase that phosphorylates proteins such as α-synuclein and amy-
loid precursor protein, have been associated with PDD and DLB (Jones et al. 2012).
The APOE ε4 allele has been established as a risk factor for AD and may also
confer an increased risk of dementia in PD, but further studies are needed (Morley
et al. 2012; Tsuang et al. 2013; Wider et al. 2012). The H1/H1 haplotype of the
MAPT gene, encoding for protein tau, has been associated with an increased risk of
some tauopathies, for example, progressive supranuclear palsy (Hoglinger et al.
2011). Interestingly, this variation in MAPT has been associated with PD as well;
however, the risk for PDD as associated with the H1/H1 haplotype in PD has been
less well studied (Neumann et al. 2009; Tsuang et al. 2013; Wider et al. 2012;
Hoglinger et al. 2011). Other possible associations such as the BDNF (Met/Met)
homozygote genotype need further confirmation (Guerini et al. 2009).
Diagnosis of PDD
At the outset it should be clarified that frank dementia in the initial stages of PD is
not considered a typical feature of the disease (Massano and Bhatia 2012). This is
particularly relevant in the differential diagnosis with DLB, in which the cognitive
184 M. Stamelou and K. Bhatia
features are similar to PDD. According to the research criteria for DLB, patients
should have an onset of dementia within 1 year after the onset of motor symptoms,
while in PDD dementia, this occurs at least 1 year after PD onset (McKeith 2006).
Similarly, cognitive dysfunction at early stages should alert the clinician for other
differential diagnoses in particular when other atypical features are present such as
progressive supranuclear palsy (Litvan et al. 1996). Moreover, clinicians should
always search for possible secondary causes if cognitive impairment in Parkinson’s
disease is noted, such as side effects from dopaminergic treatment.
The identification of patients with PDD but even more so of PD patients with
MCI, and thus patients at high risk to develop dementia, is crucial for manage-
ment and prognostic reasons but also for future therapeutic research. Clinical
criteria for PDD have recently been proposed, which however lack clinicopatho-
logical validation (Tables 11.1 and 11.2) (Emre et al. 2007a). Identification of a
dementia syndrome, defined as impairment in at least two of the four core cogni-
tive domains (e.g., attention, executive functioning, visuospatial functioning, and
free recall memory), is needed for diagnosis. Impairment should be severe enough
to affect daily social, occupational, or personal care independent of the effects of
motor or autonomic symptoms. Behavioral symptoms such as apathy, depressed
or anxious mood, hallucinations, delusions, or excessive daytime sleepiness sup-
port the diagnosis of cognitive impairment. The criteria are highly specific when
all eight items are met, but their sensitivity is low (Martinez-Martin et al. 2011;
Dujardin et al. 2010; Barton et al. 2012). However, the criteria are more sensitive
for PDD diagnosis than is the fourth edition of the diagnostic and statistical man-
ual of mental disorders (DSM-4) (Emre et al. 2007a). Moreover, diagnostic crite-
ria for MCI in PD have been proposed recently (Table 11.3) (Litvan et al. 2012).
Possible biomarkers for MCI are currently under research (Duncan et al. 2013;
Yarnall et al. 2013).
Treatment of PDD
Table 11.1 Features of dementia associated with Parkinson’s disease (Emre et al. 2007a)
I. Core features 1. Diagnosis of Parkinson’s disease according to Queen Square Brain Bank
criteria (Gibb and Lees 1988)
2. A dementia syndrome with insidious onset and slow progression,
developing within the context of established Parkinson’s disease and
diagnosed by history, clinical, and mental examination, defined as:
Impairment in more than one cognitive domain
Representing a decline from premorbid level
Deficits severe enough to impair daily life (social, occupational, or
personal care), independent of the impairment ascribable to motor or
autonomic symptoms
II. Associated 1. Cognitive features:
clinical features Attention: Impaired. Impairment in spontaneous and focused attention,
poor performance in attentional tasks; performance may fluctuate
during the day and from day to day
Executive functions: Impaired. Impairment in tasks requiring initiation,
planning, concept formation, rule finding, set shifting, or set mainte-
nance; impaired mental speed (bradyphrenia)
Visuospatial functions: Impaired. Impairment in tasks requiring visuospa-
tial orientation, perception, or construction
Memory: Impaired. Impairment in free recall of recent events or in tasks
requiring learning new material, memory usually improves with cueing,
and recognition is usually better than free recall
Language: Core functions largely preserved. Word finding difficulties and
impaired comprehension of complex sentences may be present
2. Behavioral features:
Apathy: Decreased spontaneity, loss of motivation, interest, and effortful
behavior
Changes in personality and mood including depressive features and anxiety
Hallucinations: Mostly visual, usually complex, formed visions of people,
animals, or objects
Delusions: Usually paranoid, such as infidelity or phantom boarder
(unwelcome guests living in the home) delusions
Excessive daytime sleepiness
III. Features which Coexistence of any other abnormality which may by itself cause cognitive
do not exclude impairment, but judged not to be the cause of dementia, e.g., presence
PDD, but make of relevant vascular disease in imaging
the diagnosis Time interval between the development of motor and cognitive symptoms
uncertain not known
IV. Features Cognitive and behavioral symptoms appearing solely in the context of
suggesting other other conditions such as the following:
conditions or Acute confusion due to either systemic diseases or abnormalities or drug
diseases as intoxication, major depression according to DSM IV
cause of mental Features compatible with “probable vascular dementia” criteria according
impairment, to NINDS-AIREN (dementia in the context of cerebrovascular disease
which, when as indicated by focal signs in neurological exam such as hemiparesis,
present, make it sensory deficits, and evidence of relevant cerebrovascular disease by
impossible to brain imaging and a relationship between the two as indicated by the
reliably presence of one or more of the following: onset of dementia within
diagnose PDD 3 months after a recognized stroke, abrupt deterioration in cognitive
functions, and fluctuating, stepwise progression of cognitive deficits)
186 M. Stamelou and K. Bhatia
Table 11.2 Criteria for the diagnosis of probable and possible PDD (Emre et al. 2007a)
Probable PDD (A) Core features: Both must be present
(B) Associated clinical features:
Typical profile of cognitive deficits including impairment in at least two of
the four core cognitive domains (impaired attention which may fluctuate,
impaired executive functions, impairment in visuospatial functions, and
impaired free recall memory which usually improves with cueing)
The presence of at least one behavioral symptom (apathy, depressed or
anxious mood, hallucinations, delusions, excessive daytime sleepiness)
supports the diagnosis of probable PDD; lack of behavioral symptoms,
however, does not exclude the diagnosis
(C) None of the group III features present
(D) None of the group IV features present
Possible PDD (A) Core features: Both must be present
(B) Associated clinical features:
Atypical profile of cognitive impairment in one or more domains, such as
prominent- or receptive-type (fluent) aphasia or pure storage-failure type
amnesia (memory does not improve with cueing or in recognition tasks)
with preserved attention
Behavioral symptoms may or may not be present
or
(C) One or more of the group III features present
(D) None of the group IV features present
Table 11.3 Criteria for the diagnosis of PD-MCI (Litvan et al. 2012)
I. Inclusion criteria
Diagnosis of Parkinson’s disease as based on the UK PD Brain Bank Criteria (Gibb and Lees
1988)
Gradual decline, in the context of established PD, in cognitive ability reported by either the
patient or informant or observed by the clinician
Cognitive deficits on either formal neuropsychological testing or a scale of global cognitive
abilities (detailed in section III)
Cognitive deficits are not sufficient to interfere significantly with functional independence,
although subtle difficulties on complex functional tasks may be present
II. Exclusion criteria
Diagnosis of PD dementia based on MDS Task Force proposed criteria (Emre et al. 2007a)
Other primary explanations for cognitive impairment (e.g., delirium, stroke, major depression,
metabolic abnormalities, adverse effects of medication, or head trauma)
Other PD-associated comorbid conditions (e.g., motor impairment or severe anxiety, depres-
sion, excessive daytime sleepiness, or psychosis) that, in the opinion of the clinician, signifi-
cantly influence cognitive testing
III. Specific guidelines for PD-MCI level I and level II categories
(A) Level I (abbreviated assessment)
Impairment on a scale of global cognitive abilities validated for use in PD or
Impairment on at least two tests, when a limited battery of neuropsychological tests is
performed (i.e., the battery includes less than two tests within each of the five cognitive
domains, or less than five cognitive domains are assessed)
(B) Level II (comprehensive assessment)
Neuropsychological testing that includes two tests within each of the five cognitive domains
(i.e., attention and working memory, executive, language, memory, and visuospatial)
Impairment on at least two neuropsychological tests, represented by either two impaired
tests in one cognitive domain or one impaired test in two different cognitive domains
Impairment on neuropsychological tests may be demonstrated by:
Performance approximately 1–2 SDs below appropriate norms or
Significant decline demonstrated on serial cognitive testing or
Significant decline from estimated premorbid levels
IV. Subtype classification for PD-MCI (optional, requires two tests for each of the five cognitive
domains assessed and is strongly suggested for research purposes)
PD-MCI single-domain—abnormalities on two tests within a single cognitive domain (specify
the domain), with other domains unimpaired or
PD-MCI multiple-domain—abnormalities on at least one test in two or more cognitive
domains (specify the domains)
The term “atypical parkinsonism” usually refers to disorders that present with par-
kinsonism and some features which are atypical for PD such as, for example, sym-
metric parkinsonism, symptoms unresponsive to dopaminergic treatment, and
additional signs (e.g., among others alien limb phenomenon, vertical gaze palsy,
early falls). Classically, disorders that belong to the atypical parkinsonian disorders
are progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), multi-
ple system atrophy (MSA), and DLB (which has been discussed in PDD), which
188 M. Stamelou and K. Bhatia
are distinct pathological entities. Of those, PSP and CBD belong neuropathologi-
cally to the family of tauopathies, whereas MSA and DLB belong to the
α-synucleinopathies.
Introduction
Progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD) are two
distinct pathological entities (Litvan et al. 1996; Armstrong et al. 2013). The classi-
cal PSP phenotype is characterized by postural instability and early falls, early cog-
nitive dysfunction, and abnormalities of vertical gaze and is referred to as
Richardson’s syndrome (RS) (Williams et al. 2005). The classical CBD phenotype
consists of asymmetric parkinsonism, cortical signs (e.g., apraxia, cortical sensory
loss, alien limb), and possibly other signs such as dystonia and myoclonus and is
referred to as corticobasal syndrome (CBS). However, patients with PSP and CBD
may present with phenotypes other than the classical ones: patients with PSP pathol-
ogy may present as PSP-parkinsonism, CBS, pure akinesia with gait freezing, pro-
gressive nonfluent aphasia, and others; (Lee et al. 2011; Ling et al. 2010; Stamelou
et al. 2012; Williams and Lees 2009a, b) patients with CBD pathology may present
with RS, a frontotemporal dementia (FTD) phenotype, and others (Armstrong et al.
2013; Ling et al. 2010; Stamelou et al. 2012; Kouri et al. 2011). Thus, patients with
PSP and CBD may present with overlapping clinical features including cognitive
ones and also with phenotypes of other dementia syndromes mainly FTD.
In contrast to PD, cognitive and neuropsychiatric features may be the presenting
symptom of PSP and CBD (Donker Kaat et al. 2007). Dementia is evident in both
disorders in clinical practice; however, well-designed, prospective prevalence stud-
ies of dementia in these patients are lacking. From a report of 67 patients, 85 %
showed evidence of cognitive impairment, although dementia criteria were not
applied (Donker Kaat et al. 2007). Similarly, “cognitive problems” were reported as
the initial presenting complaint in 15 % of a prevalent sample of 187 cases (Nath
et al. 2003). Cognitive and neuropsychiatric features have an important impact on
QoL in PSP and CBD (Winter et al. 2011).
In contrast to PDD described above, clinical risk factors to develop dementia in these
disorders are not well studied. For PSP, certain phenotypes such as PSP with parkin-
sonism seem to develop dementia later than the classical phenotype RS, (Williams
11 Dementia in Parkinson’s Disease and Atypical Parkinsonism 189
et al. 2005) but large prospective studies are missing. The time of onset of the symp-
toms and the duration of disease do not seem to influence the course of dementia in
these disorders, which however may correlate with age and clinical motor disability
scores (Brown et al. 2010). The evolution of cognitive impairment in relation to the
motor symptoms varies greatly not only for the different phenotypes of PSP and
CBD but also within the classical phenotypes RS and CBS (Brown et al. 2010).
Given the rarity of these disorders, most research refers to clinically diagnosed
rather than pathology-confirmed PSP and CBD patients. No prospective longitudi-
nal studies with pathological confirmation are available.
In PSP, prominent deficits are described on tests of attention and executive func-
tion, with verbal fluency being particularly severely affected, as well as deficits in
both verbal and nonverbal memory with a relative preservation of recognition. PSP
patients have impairments in processing speed and cognitive flexibility (Dubois
et al. 1988). In one study, the neurocognitive performance of 200 patients with RS
revealed primary executive dysfunction (e.g., 74 % impaired on the Frontal
Assessment Battery, 55 % impaired on Initiation/Perseveration subscale of the
Dementia Rating Scale), with milder difficulties in memory, construction, and nam-
ing (Gerstenecker et al. 2013; Mimura et al. 1997).
These deficits have been confirmed also in larger clinical studies (Brown et al.
2010).
Patients with CBD typically demonstrate difficulties that reflect impairment of
frontal-subcortical and posterior cortical visuospatial association areas, with execu-
tive problem solving and pre-motor coordination of praxis tasks that are not typically
observed in patients with PSP or AD (Pillon et al. 1995) (Massman et al. 1996).
Executive dysfunction and language difficulties characterize the cognitive profile,
while memory is generally preserved. Difficulty with limb apraxias and relatively
intact memory was found when compared to AD patients (Massman et al. 1996).
Language difficulties including verbal fluency and anomia are common in CBD.
Neuropathology
In contrast to PDD, there are no formal criteria for the diagnosis of dementia in PSP
and CBD. Clinical diagnosis of PSP relies on widely accepted criteria, which, how-
ever, are based on retrospective studies and lack prospective validation. In the cur-
rent criteria for clinical diagnosis of PSP, cognitive dysfunction is mentioned as a
supportive criterion only (Litvan et al. 1996). In the recently published clinical cri-
teria of CBD, several cognitive and neuropsychiatric domains are taken into account,
to diagnose possible or probable CBD (Armstrong et al. 2013).
In terms of useful cognitive tests, a recent meta-analysis of 141 cognitive tests
used in parkinsonian disorders showed that only 16 were found to be highly useful.
Inferior performance on phonemic and semantic verbal fluency, the Trail Making
Test, and the Wisconsin Card Sorting Test was moderately to very useful in separat-
ing PSP from PD and MSA. Cognitive testing could not differentiate CBS from
other parkinsonian disorders, although sequential orobuccal apraxia was very use-
ful. Obviously, these tests must be interpreted in conjunction with other clinical
characteristics to be helpful diagnostically (Lee et al. 2012).
Therapeutic trials to enhance cognitive function in PSP and CBD are rare and usu-
ally included a few number of patients, in an open-label fashion. A double-blinded
placebo-controlled trial of donepezil (an acetylcholinesterase inhibitor) showed
mild cognitive benefits and deleterious side effects, which exacerbated the parkin-
sonian features of PSP and dramatically worsened the functional status (Litvan et al.
2001). A small study on five PSP patients with rivastigmine showed a possible mild
benefit (Liepelt et al. 2010). Thus, there are currently no evidence-based treatments
for dementia in PSP and CBD, and more research on the pathophysiology underly-
ing PSP and CBD is required to find safe and tolerable pharmacological alterna-
tives. In terms of possible disease-modifying or neuroprotective treatments, two
large double-blind studies using the GSK-3b inhibitor tideglusib, and the microtu-
bule stabilizer davunetide, showed no clinical improvement after 1 year, in none of
the outcome measures including cognitive ones (Höglinger et al. 2013).
References
Aarsland D, Larsen JP, Tandberg E, Laake K. Predictors of nursing home placement in Parkinson’s
disease: a population-based, prospective study. J Am Geriatr Soc. 2000;48(8):938–42.
Aarsland D, Cummings JL, Larsen JP. Neuropsychiatric differences between Parkinson’s disease
with dementia and Alzheimer’s disease. Int J Geriatr Psychiatry. 2001;16(2):184–91.
Aarsland D, Zaccai J, Brayne C. A systematic review of prevalence studies of dementia in
Parkinson’s disease. Mov Disord. 2005;20(10):1255–63.
Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia
or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet
Neurol. 2009;8(7):613–8.
Alcalay RN, Caccappolo E, Mejia-Santana H, et al. Cognitive performance of GBA mutation car-
riers with early-onset PD: the CORE-PD study. Neurology. 2012;78(18):1434–40.
Armstrong MJ, Litvan I, Lang AE, et al. Criteria for the diagnosis of corticobasal degeneration.
Neurology. 2013;80(5):496–503.
Backman L, Nyberg L, Soveri A, et al. Effects of working-memory training on striatal dopamine
release. Science (New York, NY). 2011;333(6043):718.
Ballard C, Ziabreva I, Perry R, et al. Differences in neuropathologic characteristics across the
Lewy body dementia spectrum. Neurology. 2006;67(11):1931–4.
Ballard CG, Chalmers KA, Todd C, et al. Cholinesterase inhibitors reduce cortical Abeta in demen-
tia with Lewy bodies. Neurology. 2007;68(20):1726–9.
Barone P, Burn DJ, van Laar T, Hsu C, Poewe W, Lane RM. Rivastigmine versus placebo in hyper-
homocysteinemic Parkinson’s disease dementia patients. Mov Disord. 2008;23(11):1532–40.
Barton B, Grabli D, Bernard B, et al. Clinical validation of Movement Disorder Society-
recommended diagnostic criteria for Parkinson’s disease with dementia. Mov Disord. 2012;
27(2):248–53.
Braak H, Rub U, Jansen Steur EN, Del Tredici K, de Vos RA. Cognitive status correlates with
neuropathologic stage in Parkinson disease. Neurology. 2005;64(8):1404–10.
Bronnick K, Ehrt U, Emre M, et al. Attentional deficits affect activities of daily living in dementia-
associated with Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2006;77:1136–42.
11 Dementia in Parkinson’s Disease and Atypical Parkinsonism 193
Geda YE, Boeve BF, Negash S, et al. Neuropsychiatric features in 36 pathologically confirmed
cases of corticobasal degeneration. J Neuropsychiatry Clin Neurosci. 2007;19(1):77–80.
Gerstenecker A, Mast B, Duff K, Ferman TJ, Litvan I. Executive dysfunction is the primary cogni-
tive impairment in progressive supranuclear palsy. Arch Clin Neuropsychol. 2013;28(2):
104–13.
Gibb WR, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s
disease. J Neurol Neurosurg Psychiatry. 1988;51(6):745–52.
Gilman S, Wenning GK, Low PA, et al. Second consensus statement on the diagnosis of multiple
system atrophy. Neurology. 2008;71(9):670–6.
Grossman M, Gross RG, Moore P, et al. Difficulty processing temporary syntactic ambiguities in
Lewy body spectrum disorder. Brain Lang. 2012;120(1):52–60.
Guerini FR, Beghi E, Riboldazzi G, et al. BDNF Val66Met polymorphism is associated with cog-
nitive impairment in Italian patients with Parkinson’s disease. Eur J Neurol. 2009;16(11):
1240–5.
Halliday GM, McCann H. The progression of pathology in Parkinson’s disease. Ann N Y Acad Sci.
2010;1184:188–95.
Halliday G, Hely M, Reid W, Morris J. The progression of pathology in longitudinally followed
patients with Parkinson’s disease. Acta Neuropathol. 2008;115(4):409–15.
Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG. The Sydney multicenter study of
Parkinson’s disease: the inevitability of dementia at 20 years. Mov Disord. 2008;23:837–44.
Henry JD, Crawford JR. Verbal fluency deficits in Parkinson’s disease: a meta-analysis. J Int
Neuropsychol Soc. 2004;10(4):608–22.
Hoglinger GU, Melhem NM, Dickson DW, et al. Identification of common variants influencing
risk of the tauopathy progressive supranuclear palsy. Nat Genet. 2011;43(7):699–705.
Höglinger GU, Huppertz HJ, Wagenpfeil S, et al. Del Ser T; for the TAUROS MRI Investigators.
Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a random-
ized trial. Mov Disord. 2014. doi:10.1002/mds.25815.
Hurtig HI, Trojanowski JQ, Galvin J, et al. Alpha-synuclein cortical Lewy bodies correlate with
dementia in Parkinson’s disease. Neurology. 2000;54(10):1916–21.
Irwin DJ, White MT, Toledo JB, et al. Neuropathologic substrates of Parkinson disease dementia.
Ann Neurol. 2012;72:587–98.
Irwin DJ, Lee VM, Trojanowski JQ. Parkinson’s disease dementia: convergence of alpha-synuclein,
tau and amyloid-beta pathologies. Nat Rev Neurosci. 2013;14(9):626–36.
Jankovic J, Poewe W. Therapies in Parkinson’s disease. Curr Opin Neurol. 2012;25(4):433–47.
Jean L, Bergeron ME, Thivierge S, Simard M. Cognitive intervention programs for individuals
with mild cognitive impairment: systematic review of the literature. Am J Geriatr Psychiatry.
2010;18(4):281–96.
Jeannerod M, Jacob P. Visual cognition: a new look at the two-visual systems model.
Neuropsychologia. 2005;43(2):301–12.
Jellinger KA. Morphological substrates of parkinsonism with and without dementia: a retrospec-
tive clinico-pathological study. J Neural Transm Suppl. 2007;72:91–104.
Jellinger KA, Seppi K, Wenning GK, Poewe W. Impact of coexistent Alzheimer pathology on the
natural history of Parkinson’s disease. J Neural Transm (Vienna, Austria : 1996).
2002;109(3):329–39.
Jones EL, Aarsland D, Londos E, Ballard C. A pilot study examining associations between
DYRK1A and alpha-synuclein dementias. Neurodegener Dis. 2012;10(1–4):229–31.
Kempster PA, O’Sullivan SS, Holton JL, Revesz T, Lees AJ. Relationships between age and late
progression of Parkinson’s disease: a clinico-pathological study. Brain. 2010;133(Pt 6):
1755–62.
Kim HJ, Jeon BS, Kim YE, et al. Clinical and imaging characteristics of dementia in multiple
system atrophy. Parkinsonism Relat Disord. 2013;19(6):617–21.
Kotzbauer PT, Cairns NJ, Campbell MC, et al. Pathologic accumulation of alpha-synuclein and
Abeta in Parkinson disease patients with dementia. Arch Neurol. 2012;69(10):1326–31.
11 Dementia in Parkinson’s Disease and Atypical Parkinsonism 195
McKeith I, Del Ser T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a ran-
domised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):
2031–6.
Mimura M, White RF, Albert ML. Corticobasal degeneration: neuropsychological and clinical cor-
relates. J Neuropsychiatry Clin Neurosci. 1997;9(1):94–8.
Morley JF, Xie SX, Hurtig HI, et al. Genetic influences on cognitive decline in Parkinson’s disease.
Mov Disord. 2012;27(4):512–8.
Mosimann UP, Mather G, Wesnes KA, O’Brien JT, Burn DJ, McKeith IG. Visual perception in
Parkinson disease dementia and dementia with Lewy bodies. Neurology. 2004;63(11):
2091–6.
Nalls MA, Duran R, Lopez G, et al. A multicenter study of glucocerebrosidase mutations in
dementia with Lewy bodies. JAMA Neurol. 2013;70(6):727–35.
Nath U, Ben-Shlomo Y, Thomson RG, Lees AJ, Burn DJ. Clinical features and natural history of
progressive supranuclear palsy: a clinical cohort study. Neurology. 2003;60(6):910–6.
Neumann J, Bras J, Deas E, et al. Glucocerebrosidase mutations in clinical and pathologically
proven Parkinson’s disease. Brain. 2009;132(Pt 7):1783–94.
Oertel W, Poewe W, Wolters E, et al. Effects of rivastigmine on tremor and other motor symptoms
in patients with Parkinson’s disease dementia: a retrospective analysis of a double-blind trial
and an open-label extension. Drug Saf. 2008;31(1):79–94.
Ondo WG, Shinawi L, Davidson A, Lai D. Memantine for non-motor features of Parkinson’s dis-
ease: a double-blind placebo controlled exploratory pilot trial. Parkinsonism Relat Disord.
2011;17(3):156–9.
Paris AP, Saleta HG, de la Cruz Crespo Maraver M, et al. Blind randomized controlled study of the
efficacy of cognitive training in Parkinson’s disease. Mov Disord. 2011;26(7):1251–8.
Perry EK, Curtis M, Dick DJ, et al. Cholinergic correlates of cognitive impairment in Parkinson’s
disease: comparisons with Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1985;48(5):
413–21.
Pillon B, Deweer B, Agid Y, Dubois B. Explicit memory in Alzheimer’s, Huntington’s, and
Parkinson’s diseases. Arch Neurol. 1993;50(4):374–9.
Pillon B, Blin J, Vidailhet M, et al. The neuropsychological pattern of corticobasal degeneration:
comparison with progressive supranuclear palsy and Alzheimer’s disease. Neurology. 1995;
45(8):1477–83.
Poewe W, Wolters E, Emre M, et al. Long-term benefits of rivastigmine in dementia associated
with Parkinson’s disease: an active treatment extension study. Mov Disord. 2006;21(4):
456–61.
Poulopoulos M, Levy OA, Alcalay RN. The neuropathology of genetic Parkinson’s disease. Mov
Disord. 2012;27(7):831–42.
Rosenthal E, Brennan L, Xie S, et al. Association between cognition and function in patients with
Parkinson disease with and without dementia. Mov Disord. 2010;25(9):1170–6.
Schmitt FA, Farlow MR, Meng X, Tekin S, Olin JT. Efficacy of rivastigmine on executive function
in patients with Parkinson’s disease dementia. CNS Neurosci Ther. 2010;16(6):330–6.
Seppi K, Weintraub D, Coelho M, et al. The Movement Disorder Society Evidence-Based Medicine
Review Update: treatments for the non-motor symptoms of Parkinson’s disease. Mov Disord.
2011;26 Suppl 3:S42–80.
Shimada H, Hirano S, Shinotoh H, et al. Mapping of brain acetylcholinesterase alterations in Lewy
body disease by PET. Neurology. 2009;73:273–8.
Sidransky E, Nalls MA, Aasly JO, et al. Multicenter analysis of glucocerebrosidase mutations in
Parkinson’s disease. N Engl J Med. 2009;361(17):1651–61.
Stamelou M, Alonso-Canovas A, Bhatia KP. Dystonia in corticobasal degeneration: a review of the
literature on 404 pathologically proven cases. Mov Disord. 2012;27(6):696–702.
Svenningsson P, Westman E, Ballard C, Aarsland D. Cognitive impairment in patients with
Parkinson’s disease: diagnosis, biomarkers, and treatment. Lancet Neurol. 2012;11(8):
697–707.
11 Dementia in Parkinson’s Disease and Atypical Parkinsonism 197
Tsuang D, Leverenz JB, Lopez OL, et al. GBA mutations increase risk for Lewy body disease with
and without Alzheimer disease pathology. Neurology. 2012;79(19):1944–50.
Tsuang D, Leverenz JB, Lopez OL, et al. APOE epsilon4 increases risk for dementia in pure
synucleinopathies. JAMA Neurol. 2013;70(2):223–8.
Tsuboi Y, Josephs KA, Boeve BF, et al. Increased tau burden in the cortices of progressive supra-
nuclear palsy presenting with corticobasal syndrome. Mov Disord. 2005;20(8):982–8.
Weintraub D. Dopamine and impulse control disorders in Parkinson’s disease. Ann Neurol.
2008;64 Suppl 2:S93–100.
Weintraub D, Papay K, Siderowf A. Screening for impulse control symptoms in patients with de
novo Parkinson disease: a case-control study. Neurology. 2013;80(2):176–80.
Wenning GK, Jellinger KA. The role of alpha-synuclein in the pathogenesis of multiple system
atrophy. Acta Neuropathol. 2005;109(2):129–40.
Wenning GK, Litvan I, Jankovic J, et al. Natural history and survival of 14 patients with cortico-
basal degeneration confirmed at postmortem examination. J Neurol Neurosurg Psychiatry.
1998;64(2):184–9.
Wesnes KA, McKeith I, Edgar C, Emre M, Lane R. Benefits of rivastigmine on attention in demen-
tia associated with Parkinson disease. Neurology. 2005;65(10):1654–6.
Whitehouse PJ, Hedreen JC, White 3rd CL, Price DL. Basal forebrain neurons in the dementia of
Parkinson disease. Ann Neurol. 1983;13(3):243–8.
Wider C, Ross OA, Nishioka K, et al. An evaluation of the impact of MAPT, SNCA and APOE on
the burden of Alzheimer’s and Lewy body pathology. J Neurol Neurosurg Psychiatry. 2012;
83:424–9.
Williams DR, Lees AJ. Progressive supranuclear palsy: clinicopathological concepts and diagnos-
tic challenges. Lancet Neurol. 2009a;8(3):270–9.
Williams DR, Lees AJ. How do patients with parkinsonism present? A clinicopathological study.
Intern Med J. 2009b;39(1):7–12.
Williams DR, de Silva R, Paviour DC, et al. Characteristics of two distinct clinical phenotypes in
pathologically proven progressive supranuclear palsy: Richardson’s syndrome and PSP-
parkinsonism. Brain. 2005;128(Pt 6):1247–58.
Williams-Gray CH, Evans JR, Goris A, et al. The distinct cognitive syndromes of Parkinson’s
disease: 5 year follow-up of the CamPaIGN cohort. Brain. 2009;132:2958–69.
Winter Y, Spottke AE, Stamelou M, et al. Health-related quality of life in multiple system atrophy
and progressive supranuclear palsy. Neurodegener Dis. 2011;8(6):438–46.
Yarnall A, Rochester L, Burn DJ. The interplay of cholinergic function, attention, and falls in
Parkinson’s disease. Mov Disord. 2011;26:2496–503.
Yarnall AJ, Rochester L, Baker MR, et al. Short latency afferent inhibition: a biomarker for mild
cognitive impairment in Parkinson’s disease? Mov Disord. 2013;28(9):1285–8.
Chapter 12
Vascular Dementia and Parkinsonism
Cerebrovascular Disease
The classic acute manifestation of small artery disease is the lacunar stroke: a
sudden neurological deficit caused by the occlusion of a small artery, generating a
small subcortical or brainstem stroke which is between 1 and 15 mm in size. The
most common lacunar syndromes include pure motor hemiplegia, pure hemisensory
deficit or pure sensorimotor hemisyndrome (all of which can be complete, incom-
plete, or alternate, depending on the location of the stroke), ataxia hemiparesis, and
also dysarthria and clumsy hand syndrome (often associated with thalamic infarc-
tion). The so-called silent small vessel disease will often go unnoticed and asymp-
tomatic even under neurological scrutiny. Nevertheless, small vessel occlusion
might lead to cumulative neuronal loss leading to progressive neurological deficit,
including extrapyramidal features and cognitive decline.
Otto Ludwig Binswanger (October, 1852 to July, 1929) had a long-standing career
in clinical neuroscience between receiving his medical degree in 1877 and being
appointed rector of Jena University in 1911. During his career he worked in neurol-
ogy, psychology, and psychiatry and contributed with more than 100 papers encom-
passing subjects from epilepsy to hysteria. It was through his work with
neuropathology that he described eight patients suffering from progressive deterio-
ration of their motor and cognitive capacities, including “aphasic disturbances(…),
hemiamblyopia or hemianopia, hemiparesis with loss of the sense of pressure, posi-
tion or touch(…) combined with the slow and relentless deterioration of intellectual
performances.” He called this syndrome “encephalitis subcorticalis chronica pro-
gressive” because at postmortem examination he found that these patients had “a
pronounced atrophy of the hemispheric white matter, either restricted to one or
more gyri in one brain area or of several hemispheric regions affected with variable
severity; (…) these changes are most clearly found in the area of the occipital and
temporal lobes, so that temporal and occipital horns are widened into bag-like
202 L. Silveira-Moriyama et al.
cavities, while the anterior portion of the lateral ventricle shows relatively little
enlargement and the frontal white matter is almost unaffected by the disease pro-
cess. (…) The cortex does not show any remarkable macroscopic change apart from
a slight narrowing. Invariably, these cases show severe atheroma of the cerebral
arteries it is very likely that the subcortical loss of fibres is caused by a deficiency of
the blood supply resulting from arteriosclerosis.”
The concept that vascular disease could cause progressive neurological disease
was new, and it was Alzheimer who coined the term “Binswanger encephalitis”
when characterizing this process further in patients of his own. Alzheimer described
the microscopic pathology adding that “One can show in the white matter, the pres-
ence of more or less numerous foci which produce wide areas of secondary degen-
eration… Usually the foci are also to be found in the internal capsule, the lenticular
nucleus, the thalamus, and particularly in the pons in the region of the pyramidal
tract… caused by a particularly severe arteriosclerosis of the long vessels deep in
the white matter with intense atrophy of the white matter” (Pearce 1997).
MacDonald Critchley (February, 1900 to October, 1997) was a neurology con-
sultant and later dean at the Institute of Neurology in Queen Square and during his
prolific academic life published extensively in the various fields of neurology. In
1929, he described what he called “arteriosclerotic parkinsonism” which he
described as a “symptomatic variant of paralysis agitans.” Unlike Binswanger’s
clinicopathological work, Critchley’s was a clinical description of a series of
patients which he saw critically in comparison with the sparse literature on vascular
causes of parkinsonism (then including syphilis and larger strokes) and whose clini-
cal features included short-stepping gait, rigidity, and masked faces, often associ-
ated with dementia, pseudobulbar palsy, pyramidal signs, and cerebellar and
sphincter dysfunction (Critchley 1929). Due to the lack of pathological confirma-
tion, and the difficulty in establishing diagnostic criteria, Critchley’s proposal was
heavily criticized although he bravely rejected the idea of a retraction. One of the
main criticisms was that patients did not necessarily presented with classic bradyki-
nesia, to which Critchley responded proposing that the syndrome could be better
referred to as arteriosclerotic pseudoparkinsonism (Critchley 1981).
As with similar neurological conditions which were progressively differentiated
over the twentieth century, the clinical and neuropathological substrates of the con-
ditions described by Binswanger and MacDonald Critchley were further refined by
advances in neuroimaging (especially the advent of CT and MRI imaging, largely
available in the last decades) and neuropathology (including immunohistochemistry
for the various proteins involved in neurodegeneration). Various names have been
used to refer to parkinsonism caused by CVD, and more recently “vascular parkin-
sonism” seems to be the choice among various international centers (see Table 12.2
for historical evolution of the terms).
Although currently it is largely believed that atherosclerosis can cause progres-
sive dementia, vascular parkinsonism remains a somewhat controversial subject; the
differentiation between these two conditions among themselves and their symbiotic
coexistence with other forms of neurodegeneration remains mysterious. Given that
neurodegenerative conditions are most likely the end-spectrum clinical manifesta-
tions of widely prevalent neurodegenerative processes (including cell death associ-
ated with the deposition of characteristic proteins such as alpha-synuclein,
12 Vascular Dementia and Parkinsonism 203
beta-amyloid, and tau) and that small vessel disease is almost ubiquitous in the
elderly, the differentiation between vascular and primary neurodegeneration is very
challenging both on the research and clinical settings (Gorelick and Pantoni 2013),
and overall all processes contribute to neurological disability (Fig. 12.1). Overall, in
individual cases one process might be grossly more abundant then the other. For
example, in cases of clear-cut idiopathic Parkinson’s disease (PD) fulfilling Queen
Square Brain Bank criteria (Gibb and Lees 1988) and possibly with antemortem
confirmation of nigrostriatal deficit (i.e., SPECT or PET scan), often there is some
degree of small artery disease in the brain imaging by MRI or CT. Although CVD
does not fully justify the entirety of symptoms, which are traditionally attributed to
the neurodegenerative process, there is recent evidence that the neuronal damage
caused by the vascular disease plays an important role contributing to disability, in
particular when it comes to axial symptoms of PD (Bohnen et al. 2011).
Primary neuronal
degeneration
High likelihood of
Ageing chronic progressive
neurological deficit
Cerebrovascular
disease
Fig. 12.1 Contributing factors for chronic neurological deficit. The interplay of natural mecha-
nisms of ageing (cell apoptosis, decreased neuronal plasticity, etc.) and cumulative neurodegenera-
tive processes (such as the intracellular deposition of abnormal proteins, cell death, glial
dysfunction, etc.) and cerebrovascular disease of the various types contributes to the development
of chronic neurological deficit
Table 12.3 Main types of cognitive impairment associated with cerebrovascular disease
Name Main features
Subcortical ischemic Chronically progressive cognitive loss associated with a cumulative effect
vascular of various small ischemic foci caused by small vessel disease. Also
dementia known as small vessel dementia, lacunar state, Binswanger disease,
Binswanger encephalopathy
Multi-infarct Cognitive decline associated with various successive cortical strokes
dementia
Strategic infarct Cognitive decline arising from stroke affecting cognitive area (e.g.,
dementia angular gyrus, thalamus, basal forebrain, etc.)
Mixed dementia Combination of significant cerebrovascular disease associated with
underlying neurodegeneration, often Alzheimer’s disease or TPD43
proteinopathy
Vascular mild Cognitive decline not fulfilling clinical criteria for dementia which is
cognitive associated with significant cerebrovascular disease on brain imaging
impairment
Others Includes specific vascular diseases with associated cognitive decline
including CADASIL (cerebral autosomal dominant arteriopathy with
subcortical infarcts and leukoencephalopathy)
Infarcts can be ischemic or hemorrhagic strokes
12 Vascular Dementia and Parkinsonism 205
Despite various limitations (Tang et al. 2004; Román 2004; Ballard et al. 2004), the
NINDS-AIREN (Román et al. 1993) remains largely used to diagnose vascular
dementia. The NINDS-AIREN criteria classify subjects into definite, probable, and
possible vascular dementia based on main features demonstrating (a) dementia, (b)
cerebrovascular disease, and (c) a link between these two, which are summarized in
Table 12.4. In addition, exclusion criteria and features which make VaD more or less
likely are also provided (Table 12.5). The presence of all features supporting VaD and
no exclusion makes vascular dementia “probable.” Less concrete evidence might still
justify a diagnosis of “possible” vascular dementia, while postmortem confirmation of
significant cerebrovascular disease without evidence of significant neurodegeneration
(i.e., tau and beta-amyloid pathology) closes a diagnosis of “definite” VaD.
In addition to the NINDS-AIREN criteria, the Hachinski ischemic score
(Hachinski and Lassen 1974) seen in Table 12.6 might be useful in clinical practice
as a rough guide to estimate the likelihood of VaD in a patient with cognitive deficit
(Brewster et al. 2012). A meta-analysis of pathologically confirmed cases showed
that using a cutoff of ≤4 for AD and ≥7 for VaD, as originally proposed, yielded a
sensitivity of 89.0 % a and a specificity of 89.3 % to differentiate VaD from AD
(Moroney et al. 1997).
The clinical diagnosis of vascular parkinsonism remains controversial, although
progressive evidence shows that it is a separate clinical entity from neurodegenerative
parkinsonism and that it has its own natural history (see Fig. 12.2). It affects older
subjects than neurodegenerative parkinsonism in general, with a tendency to start on
the eighth decade of life, and, in comparison with Parkinson’s disease, patients pres-
ent with gait difficulties and cognitive decline quicker after disease onset. In addition,
gait problems in PD are usually indicative of advanced disease, coinciding with
206 L. Silveira-Moriyama et al.
Table 12.5 Additional criteria for vascular dementia (VaD) by NINDS-AIREN criteria
Criteria Details
Exclusion criteria (a) Factors precluding neuropsychological testing: disturbance of
consciousness, delirium, psychosis, severe aphasia, or major senso-
rimotor impairment
(b) Systemic disorders or other brain diseases that could account for
deficits
Features which (a) Early onset of memory deficit and progressive worsening of memory
make VaD and other cognitive functions such as language (transcortical sensory
uncertain or aphasia), motor skills (apraxia), and perception (agnosia), in the
unlikely absence of corresponding focal lesions on brain imaging
(b) Absence of focal neurologic signs, other than cognitive disturbance
(c) Absence of cerebrovascular lesions on brain CT or MRI
Features consistent (a) Early presence of a gait disturbance (small-step gait or marche à petits
with probable pas, or magnetic, apraxic-ataxic, or parkinsonian gait)
VaD (b) History of unsteadiness and frequent, unprovoked falls
(c) Early urinary frequency, urgency, and other urinary symptoms not
explained by urologic disease
(d) Pseudobulbar palsy
(e) Personality and mood changes, abulia, depression, emotional inconti-
nence, or other subcortical deficits including psychomotor retardation
and abnormal executive function
Summarized from Román et al. (1993)
admittance to nursing home, which is one of the most important prognostic markers
in parkinsonism. In VP, gait disturbance and cognitive decline tend to present earlier
in the disease (on average, halfway through the disease course), and patients are only
admitted to nursing home much later and closer to the end of the disease course.
Multiple system atrophy and progressive supranuclear palsy, which are important dif-
ferential diagnoses, present much earlier in life and progress faster than VP.
In a clinicopathological study, the clinical features at presentation varied accord-
ing to the speed of onset and the underlying vascular pathological state, i.e., the
presence of strategic (lacunar) infarction and diffuse subcortical ischemic vascular
12 Vascular Dementia and Parkinsonism 207
Multiple falls)
Multiple system atrophy
Parkinson’s disease
Nursing home
Vascular parkinsonism
60 70 80
Age (years)
Fig. 12.2 Natural history of vascular parkinsonism. Milestones of disease progression in patho-
logically proven parkinsonian syndromes. Bars represent the time between average disease onset
and average age at death (average disease duration). The vertical lines indicate the average time to
reach each milestone (Adapted from Glass et al. (2012))
disease (Zijlmans et al. 2004a). Certain clinical features are common in VP, includ-
ing bilateral or acute onset and the presence of an early shuffling gait, falls, cogni-
tive impairment, urinary incontinence, and corticospinal or pseudobulbar signs or
symptoms. Zijlmans et al. (2004a) proposed the first criteria for the clinical diagno-
sis of VP based on a systematic clinicopathological investigation and emphasized
that these criteria would need to be evaluated both prospectively and retrospectively
against patients with pathologically established other forms of parkinsonism to ana-
lyze sensitivity, specificity, and positive and negative predictor values (see
Table 12.7). Testing olfactory function may be helpful in differentiating vascular
parkinsonism from Parkinson’s disease, since in contrast to Parkinson’s disease,
olfactory function may be preserved (Katzenschlager et al. 2004). Likewise, the
presence of a rather symmetrical FP-CIT uptake reduction in the basal ganglia may
help to distinguish VP from PD (Zijlmans et al. 2007).
The main approach to the management of VaD or VP regards the risk factors for
progression of vascular disease. These follow the guidelines for cerebrovascular
disease in general and should target the main form of CVD present in each specific
patient. Pharmacotherapy allied with lifestyles changes can be successful in reduc-
ing the rate of progression of the underlying CVD, and might be neuroprotective.
Although isolated studies have shown that individual antihypertensive drugs might
208 L. Silveira-Moriyama et al.
Table 12.7 Proposed clinical criteria for the diagnosis of vascular parkinsonism
Criteria Details
Parkinsonism Bradykinesia (slowness of initiation of voluntary movement with
progressive reduction in speed and amplitude of repetitive actions in
either upper limb or lower limb, including the presence of reduced
step length)
and at least 1 of the following:
Rest tremor
Muscular rigidity
Postural instability not caused by primary visual, vestibular,
cerebellar, or proprioceptive dysfunction
Cerebrovascular Evidence of relevant cerebrovascular disease by brain imaging (CT or MRI)
disease The presence of focal signs or symptoms that are consistent with stroke
A relationship between An acute or delayed progressive onset with infarcts in or near areas that
these 2 disorders can increase the basal ganglia motor output (GPe [globus pallidus
In practice: pars externa] or SNc [substantia nigra pars compacta]) or decrease
the thalamocortical drive directly (VL [ventral lateral] nuclei of the
thalamus, large frontal lobe infarct). At onset, parkinsonism
consists of a contralateral bradykinetic rigid syndrome or shuffling
gait within 1 year after a stroke (VPa)
An insidious onset of parkinsonism with extensive subcortical white
matter lesions, bilateral symptoms at onset, and the presence of
early shuffling gait or early cognitive dysfunction (VPi); the
“classical clinical type”
Exclusion criteria History of repeated head injury
Encephalitis
Neuroleptic treatment at onset of symptoms
The presence of cerebral tumor or communicating hydrocephalus
Other alternative explanations for Parkinson syndrome
Summarized from Zijlmans et al. (2004a)
decrease cognitive decline associated with CVD (Peters et al. 2008), it is likely this
is a class effect rather than specific. Although dyslipidemia has been strongly asso-
ciated with cognitive decline, there is not yet enough evidence that statins can
reduce cognitive decline per se (McGuiness et al. 2010). While experimental evi-
dence suggests it might decrease amyloidogenesis, clinical trials to date failed to
show a neuroprotective effect (Sabbagh and Sparks 2012). There is some evidence
that the use of nonsteroidal anti-inflammatory drugs might decrease the risk of both
dementia and parkinsonism (Noyce et al. 2012), but not nearly enough to recom-
mend its use. There is not enough evidence for the use of either antiplatelets, anti-
coagulants, or surgical vascular treatment (Witt et al. 2007) to prevent incidence or
progression of VaD or VP, but the normal recommendations for these treatments in
the context of CVD do apply.
There is only sparse evidence to support the use of symptomatic treatment in
VP and VaD, and a case-by-case consideration is the usual route. The response
of the parkinsonian symptoms to levodopa is controversial. Most works on VP
12 Vascular Dementia and Parkinsonism 209
Conclusion
References
Ballard CG, Burton EJ, Barber R, Stephens S, Kenny RA, Kalaria RN, O’brien JT. NINDS AIREN
neuroimaging criteria do not distinguish stroke patients with and without dementia. Neurology.
2004;63(6):983–8.
Ballard C, Sauter M, Scheltens P, He Y, Barkhof F, van Straaten EC, van der Flier WM, Hsu C, Wu
S, Lane R. Efficacy, safety and tolerability of rivastigmine capsules in patients with probable
vascular dementia: the VantagE study. Curr Med Res Opin. 2008;24(9):2561–74.
Black S, Román GC, Geldmacher DS, Salloway S, Hecker J, Burns A, Perdomo C, Kumar D, Pratt
R, Donepezil 307 Vascular Dementia Study Group. Efficacy and tolerability of donepezil in
vascular dementia: positive results of a 24-week, multicenter, international, randomized,
placebo-controlled clinical trial. Stroke. 2003;34(10):2323–30.
Bohnen NI, Muller ML, Zarzhevsky N, Koeppe RA, Bogan CW, Kilbourn MR, et al. Leucoaraiosis,
nigrostriatal denervation and motor symptoms in Parkinson’s disease. Brain. 2011;134(Pt 8):
2358–65.
Bradley WG, Daroff RB, Fenichel G, Jankovic J. Neurology in clinical practice. Philadelphia:
Butterworth-Heinemann/Elsevier Saunders; 2008.
Brewster PW, Mcdowell I, Moineddin R, Tierney MC. Differential prediction of vascular dementia
and Alzheimer’s disease in nondemented older adults within 5 years of initial testing.
Alzheimers Dement. 2012;8(6):528–35.
Critchley M. Arteriosclerotic parkinsonism. Brain. 1929;52:23–83.
Critchley M. Arteriosclerotic pseudo-parkinsonism. Tunbridge Wells: Pitman Medical; 1981.
Demirkiran M, Bozdemir H, Sarica Y. Vascular parkinsonism: a distinct, heterogeneous clinical
entity. Acta Neurol Scand. 2001;104:63–7.
Erkinjuntti T, Kurz A, Gauthier S, Bullock R, Lilienfeld S, Damaraju CV. Efficacy of galantamine
in probable vascular dementia and Alzheimer’s disease combined with cerebrovascular dis-
ease: a randomized trial. Lancet. 2002;359:1283–90.
Erkinjuntti T, Roman G, Gauthier S. Treatment of vascular dementia–evidence from clinical trials
with cholinesterase inhibitors. J Neurol Sci. 2004;226:63–6.
Fitzgerald PM, Jankovic J. Lower body parkinsonism: evidence for vascular etiology. Mov Disord.
1989;4:249–60.
Gibb WR, Lees AJ. The relevance of the Lewy body to the pathogenesis of idiopathic Parkinson’s
disease. J Neurol Neurosurg Psychiatry. 1988;51:745–52.
Glass P. Clinical features of vascular parkinsonism in pathologically confirmed cases. MSc thesis,
UCL Institute of Neurology. London, 2012.
Glass PG, Lees AJ, Bacellar A, Zijlmans J, Katzenschlager R, Silveira-Moriyama L. The clinical
features of pathologically confirmed vascular parkinsonism. J Neurol Neurosurg Psychiatry.
2012;83(10):1027–9.
Gorelick PB, Pantoni L. Advances in vascular cognitive impairment. Stroke. 2013;44(2):307–8.
Hachinski VC, Lassen NA. Multi-infarct dementia: a cause of mental deterioration in the elderly.
Lancet. 1974;2:207–10.
Hachinski VC, Iliff LD, Zilhka E, et al. Cerebral blood flow in dementia. Arch Neurol. 1975;32:
632–7.
Jellinger KA. Vascular parkinsonism-neuropathological findings. Acta Neurol Scand. 2002;105:
414–5.
Kavirajan H, Schneider LS. Efficacy and adverse effects of cholinesterase inhibitors and meman-
tine in vascular dementia: a meta-analysis of randomized controlled trials. Lancet Neurol.
2007;6:782–92.
Katzenschlager R, Zijlmans J, Evans A, Watt H, Lees AJ. Olfactory function distinguishes vascu-
lar parkinsonism from Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2004;75:1749–52.
Mcshane R, Areosa AS, Minakaran N. Memantine for dementia. Cochrane Database Syst Rev.
2011;(3):CD003154.pub1.
12 Vascular Dementia and Parkinsonism 211
Mcguiness B, Ohare J, Craig D, et al. Statins for the treatment of dementia. Cochrane Database
Syst Rev. 2010;(8):CD007514.
Moroney JT, Bagiella F, Desmond DW, et al. Meta-analysis of the Hachinski Ischemic Score in
pathologically verified dementias. Neurology. 1997;49:1096–105.
Noyce AJ, Bestwick JP, Silveira-Moriyama L, Hawkes CH, Giovannoni G, Lees AJ, Schrag A.
Meta-analysis of early nonmotor features and risk factors for Parkinson disease. Ann Neurol.
2012;72(6):893–901.
Pearce JMS. Binswanger’s “encephalitis subcorticalis chronica progressiva”. J Neurol Neurosurg
Psychiatry. 1997;63:308. doi:10.1136/jnnp.63.3.308.
Peters R, Beckett N, Forette F, et al. Incident dementia and blood pressure lowering in the
Hypertension in the Very Elderly Trial cognitive function assessment (HYVET-COG): a
double-blind, placebo controlled trial. Lancet Neurol. 2008;7:683–9.
Rampello L, Alvano A, Battaglia G, Raffaele R, Vecchio I, Malaguarnera M. Different clinical and
evolutional patterns in late idiopathic and vascular parkinsonism. J Neurol. 2005;252:1045–9.
Román CG, Tatemichi TK, Erkinjuntti T, et al. Vascular dementia: diagnostic criteria for research
studies. Report of the NINCDS-AIREN International Workshop. Neurology. 1993;43:250–60.
Román GC. Facts, myths, and controversies in vascular dementia. J Neurol Sci. 2004;226(1–2):
49–52.
Sabbagh MN, Sparks DL. Statins to treat Alzheimer’s disease: an incomplete story. Expert Rev
Neurother. 2012;12(1):27–30. doi:10.1586/ern.11.171.
Santangelo G, Vitale C, Trojano L, de Gaspari D, Bilo L, Antonini A, Barone P. Differential neu-
ropsychological profiles in Parkinsonian patients with or without vascular lesions. Mov Disord.
2010;25:50–6.
Seitz DP, Adunuri N, Gill SS, Gruneir A, Herrmann N, Rochon P. Antidepressants for agitation
and psychosis in dementia. Cochrane Database Syst Rev. 2011;(2):CD008191.
Tang WK, Chan SS, Chiu HF, Ungvari GS, Wong KS, Kwok TC, Mok V, Wong KT, Richards PS,
Ahuja AT. Impact of applying NINDS-AIREN criteria of probable vascular dementia to clini-
cal and radiological characteristics of a stroke cohort with dementia. Cerebrovasc Dis.
2004;18(2):98–103.
Vale TC, Barbosa MT, Caramelli P, Cardoso F. Vascular parkinsonism and cognitive impairment.
Dement Neuropsychol. 2012;6(3):137–44.
Wilcock G, Möbius HJ, Stöffler A, MMM 500 group. A double-blind, placebo-controlled, multi-
centre study of memantine in mild to moderate vascular dementia (MMM 500). Int Clin
Psychopharmacol. 2002;17:197–305.
Winikates J, Jankovic J. Clinical correlates of vascular parkinsonism. Arch Neurol. 1999;56:
98–102.
Witt K, Borsch K, Daniels C, et al. Neuropsychological consequences of endarterectomy and
endovascular angioplasty with stent placement for treatment of symptomatic carotid stenosis.
A prospective randomized study. J Neurol. 2007;254:1524–32.
Zijlmans JC, Daniel SE, Hughes AJ, Revesz T, Lees AJ. Clinicopathological investigation of vas-
cular parkinsonism, including clinical criteria for diagnosis. Mov Disord. 2004a;19:630–40.
Zijlmans JC, Katzenschlager R, Daniel SE, Lees AJ. The L-dopa response in vascular parkinson-
ism. J Neurol Neurosurg Psychiatry. 2004b;75:545–7.
Zijlmans J, Evans A, Fontes F, et al. [123I] FP-CIT spect study in vascular parkinsonism and
Parkinson’s disease. Mov Disord. 2007;22:1278–85.
Chapter 13
Progressive Apraxia of Speech and Primary
Progressive Aphasias
Part I Clinical
Apraxia of Speech
Apraxia of speech (AOS) is a type of motor speech disorder that occurs as a result
of a defect in the planning and programming of speech production (Darley et al.
1975). Apraxia of speech has characteristics that are distinct from dysarthria,
another type of motor speech disorder. Dysarthria, unlike AOS, reflects a defect of
neuromuscular function (Darley et al. 1969). The salient features of AOS are sound
production errors that may include any combination of distorted sound substitu-
tions, additions, prolongations, and truncations. Articulation is effortful and is asso-
ciated with groping and uncoordinated movements of buccolingual structures.
Apraxia of speech has been most commonly associated with cerebrovascular
events in which the onset is acute and severity maximum. More recently, however,
there has been increased interest in AOS that is progressive in nature (Duffy 2006,
2013). Progressive AOS (PAOS), unlike AOS due to cerebrovascular events, is char-
acterized by a general worsening of AOS features; errors in speech production
become more frequent and more severe over time (Table 13.1). As a result, verbal
communication becomes more problematic. Ultimately, patients with PAOS become
anarthric and are no longer able to verbally communicate.
Table 13.1 Clinical definitions and demographics based on a cohort of 100 subjects
Mean age Gender
Clinical syndrome Definition at onset dominance
Primary progressive Apraxia of speech is the only feature 69 Female
apraxia of speech present and is characterized by
(PPAOS) distorted sound production
Progressive apraxia ofApraxia of speech is the most prominent 67 Male
speech (PAOS) feature. Additional motor, cognitive, or
language features are present but are
less prominent
Agrammatic variant of Agrammatic aphasia is the only feature 66 Equal
primary progres- present or the most prominent feature.
sive aphasia Apraxia of speech may be present but is
(agPPA) less prominent
Semantic variant of Syndrome characterized by poor naming 64 Equal
primary progres- and loss of word meaning.
sive aphasia
(svPPA)
Logopenic variant of Syndrome characterized by poor naming 63 Equal
primary progres- without loss of word meaning, poor
sive aphasia sentence repetition, and phonological
(lvPPA) errors
Corticobasal syndrome Syndrome characterized by asymmetric N/A N/A
(CBS) ideomotor apraxia, dystonia, parkinson-
ism, and cortical sensory loss. Apraxia
of speech and agrammatic aphasia may
be present.
Syndrome + motor Features of apraxia of speech or agram- N/A N/A
neuron disease matic aphasia are accompanied by
features of motor neuron disease, such
as spastic and flaccid dysarthria, tongue
and limb fasciculation, weakness,
hyperreflexia, and Babinski sign
PPAOS
PAOS
AOS+
AOS PSPS Agrammatic aphasia
CBS
agPPA
Fig. 13.1 demonstrates how patients presenting with apraxia of speech variants progress over time.
AOS apraxia of speech, agPPA agrammatic variant of primary progressive aphasia, PAOS progressive
apraxia of speech (i.e., AOS > > other cognitive and motor features), PPAOS primary progressive apraxia
of speech (AOS only), CBS corticobasal syndrome, PSPS progressive supranuclear palsy syndrome
216 K.A. Josephs and J.L. Whitwell
Progressive apraxia of speech may coexist with other cognitive and motor deficits.
In some instances, the PAOS is the most prominent feature of the presenting syn-
drome. In such instances, the chief complaint is usually that of speech problems;
other presenting features are more subtle and at times may only have been identified
during the neurological and/or speech and language evaluation. In other instances,
when AOS coexists with other cognitive and/or motor deficits, the other features of
the syndrome overshadow the AOS. Typically, when this occurs, the chief complaint
is related to one of the other symptoms and the AOS tends to be mild. Two syn-
dromes that are worth discussing in detail in which PAOS commonly occurs early in
the disease course are primary progressive aphasia (PPA) and CBS.
The term primary progressive aphasia (PPA) is used in situations in which there is a
progressive disorder characterized by impairment of specific aspects of language
function (Mesulam 1982, 2001). These include grammar, naming, single word com-
prehension, sentence comprehension, and repetition. Three subtypes of PPA are
now recognized (Table 13.1).
The first subtype of PPA is the agrammatic variant (agPPA) (Gorno-Tempini et al.
2011). Agrammatic PPA is a syndrome in which the most salient feature is that of
agrammatism in language production. This may be observed in spoken or written
form. In agrammatic PPA, function words and articles may be conspicuously absent,
e.g., “Man fishing with wife on dock. Boat goes by she waves to them.” Importantly,
patients with agPPA commonly also exhibit PAOS (Ogar et al. 2005). Hence, agPPA
is typically characterized by the presence of agrammatic aphasia of greater severity
than the accompanying PAOS (Josephs et al. 2013a). Examination shows little defi-
cits in naming and in sentence repetition for content words, as well as difficulty
understanding complex sentence structures, e.g., “put the brown newspaper under
the blue book in the middle drawer.” Importantly, given that PPA is a disorder of
language, a diagnosis of PPA should only be made when language impairment, not
AOS, is the most prominent feature of the syndrome (Mesulam 2003).
13 Progressive Apraxia of Speech and Primary Progressive Aphasias 217
The second subtype of PPA is the semantic variant (svPPA) (Gorno-Tempini et al.
2011; Warrington 1975). In svPPA, the speech output is fluent and may even be
excessively verbose. However, with more careful attention to what is actually being
said, it becomes apparent that the patient is being circumloquacious in order to
avoid certain words that no longer has specific meaning to the patient. Therefore,
specific words may be absent in general conversation and replaced with more gen-
eral words for the item or object being described, e.g., the patient may say animal
instead of hyena or flower instead of hibiscus. There may also be reference to the
fact that the patient no longer understands the meaning of certain words; the patient
may no longer know what the word lapel means and hence will not be able to tell
how a lapel is different from a collar. Examination shows poor naming and loss of
single word meaning and comprehension (Hodges and Patterson 2007). Unlike in
agPPA, AOS rarely, if ever, coexists with svPPA.
The third subtype of PPA is the logopenic variant (lvPPA) (Gorno-Tempini et al.
2004, 2011). In lvPPA, the speech output may be fluent, but it may also be character-
ized by hesitancy as the patient pauses and searches for words. However, unlike in
svPPA, patients with lvPPA can easily recognize the item when the name is provided
by the examiner, and hence the patient has not lost word meaning. Memory loss is
also often a common complaint in patients with lvPPA. Like in svPPA, patients with
lvPPA perform poorly on naming tasks but there is no loss of word meaning.
Examination also reveals poor repetition of sentences with loss of words or replace-
ment of exact content words. One additional feature of lvPPA is the production of
sound errors known as phonological or phonemic errors. With phonological errors,
one sound is produced for another, e.g., sesipic instead of specific. Importantly,
unlike in AOS, the sounds are not distorted. It can be difficult at times to distinguish
AOS errors from phonological errors, resulting in patients with lvPPA being misdi-
agnosed as having PAOS and vice versa. Apraxia of speech rarely occurs in lvPPA.
Corticobasal Syndrome
Patients with CBS, who also have AOS, are more likely to show right limb asymmetry,
i.e., the right side is the more affected side, and often also have agrammatic aphasia.
In CBS in which AOS or agrammatic aphasia are present, the presenting complaint is
usually that of limb dysfunction, as opposed to speech difficulties, although both may
be mentioned as being experienced by the patient.
Apraxia of speech can be associated with movement disorders, although the move-
ment disorder identified tends to be a syndrome as opposed to being an isolated fea-
ture. We previously discussed the association of AOS and CBS at presentation;
patients with AOS may also show features of CBS and PSPS with disease progres-
sion. When this occurs, the patient is usually 5 years out from onset. It is rare, how-
ever, for the classic syndrome of PSPS to co-occur with AOS at presentation
(Whitwell et al. 2013a). When PSPS features develop in patients that originally pres-
ent with PPAOS or PAOS, they tend to be milder than what is typically observed in
PSPS that starts off with balance problems and eye movement abnormalities.
However, slowing of vertical eye movements, balance problems, and falls become
more prevalent and severe over time in patients presenting with PPAOS or PAOS.
Similarly, patients with PPAOS or PAOS may show the emergence of ideomotor
apraxia after many years (Josephs et al. 2006a, 2012). Of the three PPA syndromes,
agPPA is the one most likely to also develop features of CBS and PSPS over time
(Kertesz et al. 2005). In fact, ideomotor apraxia has been shown to be more common
in agPPA than lvPPA (Adeli et al. 2013). Parkinsonian features such as rigidity and
13 Progressive Apraxia of Speech and Primary Progressive Aphasias 219
bradykinesia are relatively common later on in the disease course in PPAOS and
PAOS. In PPA, almost 30 % of patients present with at least one parkinsonian sign,
such as bradykinesia, rigidity, tremor, postural instability, or gait disturbance (Kremen
et al. 2011). Parkinsonian features are more common in agPPA than lvPPA and are
almost absent in svPPA (Graff-Radford et al. 2012; Kremen et al. 2011). Dystonia is
much less common but may also be observed later on in the disease course.
Hyperkinetic movements can also occur, although resting tremor is rare. Asymmetric
limb myoclonus can occur later on in the disease course in patients who later develop
the CBS. Stereotypies and ticks have not been emphasized in PPA, PPAOS, or PAOS.
Part II Anatomy
Routine clinical studies in patients with PAOS have not been very helpful in identi-
fying the anatomic correlate of PAOS. However, studies using research-based imag-
ing techniques performed using groups of patients have demonstrated that PAOS is
associated with the premotor cortex. Structural magnetic resonance imaging (MRI)
studies using voxel-level techniques, such as voxel-based morphometry, in patients
with PPAOS have shown focal patterns of grey matter atrophy involving the supe-
rior premotor and supplemental motor cortices (Josephs et al. 2012; Whitwell et al.
2013a). White matter atrophy is also observed in the premotor cortex, underlying
regions of grey matter loss, but tends to also involve inferior premotor regions.
Detailed analysis of white matter tracts using diffusion tensor imaging (DTI) has
shown that PPAOS is associated with degeneration of premotor aspects of the supe-
rior longitudinal fasciculus and the body of the corpus callosum (Josephs et al.
2012). Hypometabolism on 18-F fluorodeoxyglucose positron emission tomogra-
phy (FDG-PET) scans is also observed in the superior premotor cortices in patients
with PPAOS (Fig. 13.2a), although hypometabolism on FDG-PET can be very mild,
or even absent, in some patients (Josephs et al. 2012).
Imaging findings in patients with PPAOS overlap to some degree with those asso-
ciated with PSPS, with both syndromes showing atrophy and hypometabolism in the
premotor cortex and degeneration of the superior longitudinal fasciculus and body of
the corpus callosum (Whitwell et al. 2011, 2013a; Knake et al. 2010; Josephs et al.
2008). However, in contrast to PPAOS, patients with PSPS typically show greater
involvement of the prefrontal cortex and striking atrophy in the midbrain (Whitwell
et al. 2013a). Mild midbrain atrophy has however been observed in patients with
PPAOS, likely reflecting the fact that these patients often develop clinical features of
PSPS (Whitwell et al. 2013a). In patients with both PAOS and agrammatic aphasia,
where the AOS is the more dominant feature, VBM and FDG-PET studies show
involvement of the superior premotor cortex extending into the middle and inferior
premotor regions (Fig. 13.2b) (Josephs et al. 2013a). This finding suggests that the
220 K.A. Josephs and J.L. Whitwell
Fig. 13.2 demonstrates focal hypometabolism of the left superior premotor cortex in a patient
with PPAOS (a) with extension of the hypometabolism into middle and inferior premotor cortices
in a patient with PAOS (AOS > agrammatic aphasia) (b)
Each of the three variants of PPA shows distinct regional abnormalities within the
frontotemporal and parietal cortices. In agPPA, routine brain MRI scans reveal atro-
phy of the left inferior frontal lobe with widening of the left perisylvian fissure
(Fig. 13.3a). Research studies demonstrate atrophy and hypometabolism on FDG-
PET throughout the left premotor cortex (Fig. 13.3b) but also with involvement of
the left insula, striatum, lateral temporal lobes, and often other regions in the left
frontal and parietal lobes (Josephs et al. 2006a, 2010; Gorno-Tempini et al. 2004;
13 Progressive Apraxia of Speech and Primary Progressive Aphasias 221
Grossman et al. 2004; Rohrer et al. 2009). Patterns observed in patients with agPPA
are typically more widespread than those observed in patients with PAOS. Relatively
widespread patterns of white matter tract degeneration have also been observed in
agPPA, involving the superior longitudinal fasciculus, corpus callosum, anterior
cingulate, inferior frontal-occipital fasciculus, and temporal lobe tracts such as the
a b
c d
Fig. 13.3 demonstrates the abnormalities that can be identified on MRI and FDG-PET in patients
with the different variants of PPA. In (a), a patient with agPPA shows widening of the left perisyl-
vian area, while in (b), FDG-PET shows premotor cortical hypometabolism, as well as hypome-
tabolism in left temporal and even parietal cortices. On MRI (c) there is left > right anterior medial
temporal lobe atrophy which is also seen on the FDG-PET scan (d) in a patient with svPPA. A
patient with the lvPPA variant shows left parietal atrophy on MRI (e), while FDG-PET shows left
temporal and parietal hypometabolism (f)
222 K.A. Josephs and J.L. Whitwell
e f
lobes (Fig. 13.3e). Group MRI studies and FDG-PET (Fig. 13.3f) show striking
involvement of the left lateral temporal and parietal lobes but can also show involve-
ment of the precuneus, frontal lobes, and even the occipital lobes (Gorno-Tempini
et al. 2004; Josephs et al. 2010; Madhavan et al. 2013; Ridgway et al. 2012). The
medial temporal lobe is usually relatively spared. The right temporal and parietal
lobes may be abnormal but are nearly always less affected than the left. White mat-
ter tract degeneration can be observed on DTI affecting temporal and parietal tracts,
including the left inferior longitudinal fasciculus, uncinate fasciculus, and superior
longitudinal fasciculus (Armstrong et al. 2013; Graff-Radford et al. 2012).
Pathological Overview
Over the last decade, many investigators interested in AOS and PPA have assessed
the pathological processes associated with these different syndromes (Josephs et al.
2005, 2006a, b; Mesulam 2001; Kertesz et al. 1994, 2005; Davies et al. 2005; Galton
et al. 2000; Graff-Radford et al. 1990; Greene et al. 1996; Hodges et al. 2004; Knibb
et al. 2006; Knopman et al. 1990; Lang 1992; Lippa et al. 1991; Wechsler et al.
1982). Unfortunately, it is very difficult to interpret some of these older studies
since subjects were not separated by phenotype or syndrome and instead were all
lumped as PPA, including those with PAOS and PPAOS. Regardless, these older
224 K.A. Josephs and J.L. Whitwell
studies demonstrated that the pathological processes associated with PPA were
not homogeneous. Taken together, these studies demonstrated that many different
pathologies, including Alzheimer’s disease (Kertesz et al. 2005; Galton et al. 2000;
Greene et al. 1996; Knibb et al. 2006), accounted for PPA. Another problem with
older studies was the fact that modern immunohistochemistry techniques were not
performed, and even if they were performed, they were incomplete, at least com-
pared to more recent studies.
With the advent of immunohistochemistry, and recent discoveries of different
proteins that are associated with dementia in general, pathologists have a better grasp
on the biochemistry and the pathological processes that are associated with these
syndromes. Therefore, current classification first separates Alzheimer’s disease from
non-Alzheimer’s disease pathologies. Non-Alzheimer’s disease pathologies are then
further separated under the broad categories of synucleinopathies, tauopathies, and
TDP-43 proteinopathies (Dickson 2003). Synucleinopathies are not discussed in this
chapter but include multiple system atrophy and Lewy body diseases (Dickson
2003); Lewy bodies are rarely found in PPA patients (Caselli et al. 2002). Further
subclassification of the tauopathies exists and includes diseases in which tau is the
predominant abnormal protein identified histologically (Josephs et al. 2011).
Therefore, tauopathies include progressive supranuclear palsy (PSP) (Hauw et al.
1994), corticobasal degeneration (CBD) (Dickson et al. 2002), Pick’s disease with
Pick’s bodies (PiD) (Dickson 2001), and frontotemporal dementia with Parkinsonism
linked to chromosome 17 (FTDP-17) (Ghetti et al. 2003). Similarly the TDP-43
proteinopathies are subclassified in types A–D (Mackenzie et al. 2011). For both
tauopathies and TDP-43 proteinopathies, subclassification is based on the distribu-
tion of the inclusions, as well as the morphological appearances of the inclusions.
Clinicopathological Associations
Part IV Genetics
There are no genetic abnormalities that completely account for any one of the syn-
dromes discussed in this chapter. However, there have been reports of patients with
a genetic abnormality, namely, a mutation in the progranulin gene on chromosome
17, that have presented with PPA (Mesulam et al. 2007; Mukherjee et al. 2006;
Rohrer et al. 2010; Kelley et al. 2009). Most of these reports did not further classify
the PPA into one of the three variants. However, patients with progranulin gene
226 K.A. Josephs and J.L. Whitwell
mutations are almost always fluent and hence have features that overlap with svPPA
and lvPPA. To our knowledge, there are no definitive, well-characterized patients
with PPAOS or PAOS that have had a mutation in the progranulin gene. Recently,
there have also been a handful of patients with PPA that have been found to have a
repeat expansion in the C9ORF72 gene (Simon-Sanchez et al. 2012; Snowden et al.
2006, 2012; Mahoney et al. 2012). Other investigators with large cohorts of patients
with C9ORF72 repeat expansions, however, have not identified patients with PPA
(Boeve et al. 2012). There are no reports of the repeat expansion accounting for the
PPAOS or PAOS phenotypes. Most patients with FTDP-17, i.e., with a mutation in
the microtubule associated protein tau gene, have not commonly presented with any
of the clinical syndromes discussed in this chapter. There are a few reports of
patients presenting as svPPA, although it is unclear whether these patients were
truly svPPA or whether they had a behavioral presentation in which naming was
also affected as part of the behavioral syndrome (Pickering-Brown et al. 2008).
Summary
There are many different clinical syndromes in which a motor speech disorder or apha-
sia is the defining feature. In some instances, the motor speech disorders and aphasia
co-occur. However, it is important not to lump all these features under the heading of
PPA. In fact, the presence of AOS is strongly predictive of tau pathologies, the pres-
ence of flaccid and spastic dysarthrias of MND and TDP type B pathology, svPPA of
TDP type C pathology, and lvPPA of Alzheimer’s disease. Understanding how to sepa-
rate these clinical features and syndromes will have important prognostic value and
future therapeutic benefit. Parkinsonism is frequent in agPPA and lvPPA, and further
imaging and pathological studies are necessary to better characterize this feature.
References
Acosta-Cabronero J, Patterson K, Fryer TD, Hodges JR, Pengas G, et al. Atrophy, hypometabo-
lism and white matter abnormalities in semantic dementia tell a coherent story. Brain.
2012;134:2025–35.
Adeli A, Whitwell JL, Duffy JR, Strand EA, Josephs KA. Ideomotor apraxia in agrammatic and
logopenic variants of primary progressive aphasia. J Neurol. 2013;260:1594–600.
Agosta F, Henry RG, Migliaccio R, Neuhaus J, Miller BL, et al. Language networks in semantic
dementia. Brain. 2010;133:286–99.
Armstrong MJ, Litvan I, Lang AE, Bak TH, Bhatia KP, et al. Criteria for the diagnosis of cortico-
basal degeneration. Neurology. 2013;80:496–503.
Assal F, Laganaro M, Remund CD, Ragno Paquier C. Progressive crossed-apraxia of speech as a
first manifestation of a probable corticobasal degeneration. Behav Neurol. 2012;25:285–9.
Boeve BF, Boylan KB, Graff-Radford NR, DeJesus-Hernandez M, Knopman DS, et al.
Characterization of frontotemporal dementia and/or amyotrophic lateral sclerosis associated
with the GGGGCC repeat expansion in C9ORF72. Brain. 2012;135:765–83.
13 Progressive Apraxia of Speech and Primary Progressive Aphasias 227
Borroni B, Garibotto V, Agosti C, Brambati SM, Bellelli G, et al. White matter changes in
corticobasal degeneration syndrome and correlation with limb apraxia. Arch Neurol. 2008;65:
796–801.
Boxer AL, Geschwind MD, Belfor N, Gorno-Tempini ML, Schauer GF, et al. Patterns of brain
atrophy that differentiate corticobasal degeneration syndrome from progressive supranuclear
palsy. Arch Neurol. 2006;63:81–6.
Broussolle E, Tommasi M, Mauguiere F, Chazot G. Progressive anarthria with secondary parkin-
sonism: a clinico-pathological case report. J Neurol Neurosurg Psychiatry. 1992;55:577–80.
Caselli RJ, Windebank AJ, Petersen RC, Komori T, Parisi JE, et al. Rapidly progressive aphasic
dementia and motor neuron disease. Ann Neurol. 1993;33:200–7.
Caselli RJ, Beach TG, Sue LI, Connor DJ, Sabbagh MN. Dement Geriatr Cogn Disord. 2002;
14:55–8.
Chan D, Fox NC, Scahill RI, Crum WR, Whitwell JL, et al. Patterns of temporal lobe atrophy in
semantic dementia and Alzheimer’s disease. Ann Neurol. 2001;49:433–42.
Chan D, Anderson V, Pijnenburg Y, Whitwell J, Barnes J, et al. The clinical profile of right tempo-
ral lobe atrophy. Brain. 2009;132:1287–98.
Coon EA, Sorenson EJ, Whitwell JL, Knopman DS, Josephs KA. Predicting survival in frontotem-
poral dementia with motor neuron disease. Neurology. 2011;76:1886–93.
Czell D, Andersen PM, Neuwirth C, Morita M, Weber M. Progressive aphasia as the presenting
symptom in a patient with amyotrophic lateral sclerosis with a novel mutation in the OPTN
gene. Amyotroph Lateral Scler Frontotemporal Degener. 2013;14:138–40.
da Rocha AJ, Valerio BC, Buainain RP, Ferraz ME, da Silva CJ, et al. Motor neuron disease associ-
ated with non-fluent rapidly progressive aphasia: case report and review of the literature. Eur
J Neurol. 2007;14:971–5.
Darley FL, Aronson AE, Brown JR. Differential diagnostic patterns of dysarthria. J Speech Hear
Res. 1969;12:246–69.
Darley FL, Aronson AE, Brown JR. Motor speech disorders. Philadelphia: W.B. Saunders; 1975.
Davies RR, Hodges JR, Kril JJ, Patterson K, Halliday GM, et al. The pathological basis of seman-
tic dementia. Brain. 2005;128:1984–95.
Deramecourt V, Lebert F, Debachy B, Mackowiak-Cordoliani MA, Bombois S, et al. Prediction of
pathology in primary progressive language and speech disorders. Neurology. 2010;74:42–9.
Dickson DW. Neuropathology of Pick’s disease. Neurology. 2001;56:S16–20.
Dickson DW. Neurodegeneration: the molecular pathology of dementia and movement disorders.
Basel: ISN Neuropath Press; 2003.
Dickson DW, Bergeron C, Chin SS, Duyckaerts C, Horoupian D, et al. Office of Rare Diseases neu-
ropathologic criteria for corticobasal degeneration. J Neuropathol Exp Neurol. 2002;61:935–46.
Duffy J. Apraxia of speech in degenerative neurologic disease. Aphasiology. 2006;20:511–27.
Duffy JR. Motor speech disorders: substrates, differential diagnosis, and management. St Louis:
Mosby; 2013.
Duffy JR, Peach RK, Strand EA. Progressive apraxia of speech as a sign of motor neuron disease.
Am J Speech Lang Pathol. 2007;16:198–208.
Edwards-Lee T, Miller BL, Benson DF, Cummings JL, Russell GL, et al. The temporal variant of
frontotemporal dementia. Brain. 1997;120(Pt 6):1027–40.
Galantucci S, Tartaglia MC, Wilson SM, Henry ML, Filippi M, et al. White matter damage in pri-
mary progressive aphasias: a diffusion tensor tractography study. Brain. 2011;134:3011–29.
Galton CJ, Patterson K, Xuereb JH, Hodges JR. Atypical and typical presentations of Alzheimer’s
disease: a clinical, neuropsychological, neuroimaging and pathological study of 13 cases.
Brain. 2000;123(Pt 3):484–98.
Galton CJ, Patterson K, Graham K, Lambon-Ralph MA, Williams G, et al. Differing patterns of
temporal atrophy in Alzheimer’s disease and semantic dementia. Neurology. 2001;57:216–25.
Ghetti B, Hutton M, Wszolek Z. Frontotemporal dementia and parkinsonism linked to chromosome
17 associated with tau gene mutations (FTDP-17T). In: Dickson D, editor. Neurodegeneration:
the molecular pathology of dementia and movement disorders. Basel: ISN Neuropath Press;
2003. p. 86–102.
228 K.A. Josephs and J.L. Whitwell
Gorno-Tempini ML, Dronkers NF, Rankin KP, Ogar JM, Phengrasamy L, et al. Cognition and
anatomy in three variants of primary progressive aphasia. Ann Neurol. 2004;55:335–46.
Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M, et al. Classification of primary
progressive aphasia and its variants. Neurology. 2011;76:1006–14.
Graff-Radford NR, Damasio AR, Hyman BT, Hart MN, Tranel D, et al. Progressive aphasia in a
patient with Pick’s disease: a neuropsychological, radiologic, and anatomic study. Neurology.
1990;40:620–6.
Graff-Radford J, Duffy JR, Strand EA, Josephs KA. Parkinsonian motor features distinguish the
agrammatic from logopenic variant of primary progressive aphasia. Parkinsonism Relat Disord.
2012;18:890–2.
Greene JD, Patterson K, Xuereb J, Hodges JR. Alzheimer disease and nonfluent progressive apha-
sia. Arch Neurol. 1996;53:1072–8.
Groschel K, Hauser TK, Luft A, Patronas N, Dichgans J, et al. Magnetic resonance imaging-based
volumetry differentiates progressive supranuclear palsy from corticobasal degeneration.
Neuroimage. 2004;21:714–24.
Grossman M, McMillan C, Moore P, Ding L, Glosser G, et al. What’s in a name: voxel-based
morphometric analyses of MRI and naming difficulty in Alzheimer’s disease, frontotemporal
dementia and corticobasal degeneration. Brain. 2004;127:628–49.
Grossman M, Powers J, Ash S, McMillan C, Burkholder L, et al. Disruption of large-scale neural
networks in non-fluent/agrammatic variant primary progressive aphasia associated with fronto-
temporal degeneration pathology. Brain Lang. 2013;127:106–20.
Hauw JJ, Daniel SE, Dickson D, Horoupian DS, Jellinger K, et al. Preliminary NINDS neuro-
pathologic criteria for Steele-Richardson-Olszewski syndrome (progressive supranuclear
palsy). Neurology. 1994;44:2015–9.
Hodges JR, Patterson K. Semantic dementia: a unique clinicopathological syndrome. Lancet
Neurol. 2007;6:1004–14.
Hodges JR, Davies RR, Xuereb JH, Casey B, Broe M, et al. Clinicopathological correlates in fron-
totemporal dementia. Ann Neurol. 2004;56:399–406.
Huey ED, Pardini M, Cavanagh A, Wassermann EM, Kapogiannis D, et al. Association of ideomo-
tor apraxia with frontal gray matter volume loss in corticobasal syndrome. Arch Neurol.
2009;66:1274–80.
Josephs KA, Duffy JR. Apraxia of speech and nonfluent aphasia: a new clinical marker for corti-
cobasal degeneration and progressive supranuclear palsy. Curr Opin Neurol. 2008;21:
688–92.
Josephs KA, Boeve BF, Duffy JR, Smith GE, Knopman DS, et al. Atypical progressive supranu-
clear palsy underlying progressive apraxia of speech and nonfluent aphasia. Neurocase.
2005;11:283–96.
Josephs KA, Duffy JR, Strand EA, Whitwell JL, Layton KF, et al. Clinicopathological and imag-
ing correlates of progressive aphasia and apraxia of speech. Brain. 2006a;129:1385–98.
Josephs KA, Petersen RC, Knopman DS, Boeve BF, Whitwell JL, et al. Clinicopathologic analysis
of frontotemporal and corticobasal degenerations and PSP. Neurology. 2006b;66:41–8.
Josephs KA, Whitwell JL, Dickson DW, Boeve BF, Knopman DS, et al. Voxel-based morphometry
in autopsy proven PSP and CBD. Neurobiol Aging. 2008;29:280–9.
Josephs KA, Whitwell JL, Knopman DS, Boeve BF, Vemuri P, et al. Two distinct subtypes of right
temporal variant frontotemporal dementia. Neurology. 2009a;73:1443–50.
Josephs KA, Stroh A, Dugger B, Dickson DW. Evaluation of subcortical pathology and clinical
correlations in FTLD-U subtypes. Acta Neuropathol. 2009b;118:349–58.
Josephs KA, Duffy JR, Fossett TR, Strand EA, Claassen DO, et al. Fluorodeoxyglucose F18 posi-
tron emission tomography in progressive apraxia of speech and primary progressive aphasia
variants. Arch Neurol. 2010;67:596–605.
Josephs KA, Hodges JR, Snowden JS, Mackenzie IR, Neumann M, et al. Neuropathological back-
ground of phenotypical variability in frontotemporal dementia. Acta Neuropathol.
2011;122:137–53.
Josephs KA, Duffy JR, Strand EA, Machulda MM, Senjem ML, et al. Characterizing a neurode-
generative syndrome: primary progressive apraxia of speech. Brain. 2012;135:1522–36.
13 Progressive Apraxia of Speech and Primary Progressive Aphasias 229
Josephs KA, Duffy JR, Strand EA, Machulda MM, Senjem ML, et al. Syndromes dominated by
apraxia of speech show distinct characteristics from agrammatic PPA. Neurology.
2013a;81:337–45.
Josephs KA, Whitwell JL, Murray ME, Parisi JE, Graff-Radford NR, et al. Corticospinal tract
degeneration associated with TDP-43 type C pathology and semantic dementia. Brain.
2013b;136:455–70.
Kelley BJ, Haidar W, Boeve BF, Baker M, Graff-Radford NR, et al. Prominent phenotypic vari-
ability associated with mutations in Progranulin. Neurobiol Aging. 2009;30:739–51.
Kertesz A, Hudson L, Mackenzie IR, Munoz DG. The pathology and nosology of primary progres-
sive aphasia. Neurology. 1994;44:2065–72.
Kertesz A, McMonagle P, Blair M, Davidson W, Munoz DG. The evolution and pathology of
frontotemporal dementia. Brain. 2005;128:1996–2005.
Knake S, Belke M, Menzler K, Pilatus U, Eggert KM, et al. In vivo demonstration of microstruc-
tural brain pathology in progressive supranuclear palsy: a DTI study using TBSS. Mov Disord.
2010;25:1232–8.
Knibb JA, Xuereb JH, Patterson K, Hodges JR. Clinical and pathological characterization of pro-
gressive aphasia. Ann Neurol. 2006;59:156–65.
Knopman DS, Mastri AR, Frey 2nd WH, Sung JH, Rustan T. Dementia lacking distinctive histo-
logic features: a common non-Alzheimer degenerative dementia. Neurology. 1990;40:251–6.
Koyama M, Yagishita A, Nakata Y, Hayashi M, Bandoh M, et al. Imaging of corticobasal degen-
eration syndrome. Neuroradiology. 2007;49:905–12.
Kremen SA, Mendez MF, Tsai P, Teng E. Extrapyramidal signs in the primary progressive apha-
sias. Am J Alzheimers Dis Other Demen. 2011;26:72–7.
Lang AE. Cortical basal ganglionic degeneration presenting with “progressive loss of speech out-
put and orofacial dyspraxia”. J Neurol Neurosurg Psychiatry. 1992;55:1101.
Lippa CF, Cohen R, Smith TW, Drachman DA. Primary progressive aphasia with focal neuronal
achromasia. Neurology. 1991;41:882–6.
Mackenzie IR, Baborie A, Pickering-Brown S, Du Plessis D, Jaros E, et al. Heterogeneity of ubiq-
uitin pathology in frontotemporal lobar degeneration: classification and relation to clinical phe-
notype. Acta Neuropathol (Berl). 2006;112:539–49.
Mackenzie IR, Neumann M, Baborie A, Sampathu DM, Du Plessis D, et al. A harmonized classi-
fication system for FTLD-TDP pathology. Acta Neuropathol. 2011;122:111–3.
Madhavan A, Whitwell JL, Weigand SD, Duffy JR, Strand EA, et al. FDG PET and MRI in logo-
penic primary progressive aphasia versus dementia of the Alzheimer’s type. PLoS One.
2013;8:e62471.
Mahoney CJ, Beck J, Rohrer JD, Lashley T, Mok K, et al. Frontotemporal dementia with the
C9ORF72 hexanucleotide repeat expansion: clinical, neuroanatomical and neuropathological
features. Brain. 2012;135:736–50.
Mahoney CJ, Malone IB, Ridgway GR, Buckley AH, Downey LE, et al. White matter tract signa-
tures of the progressive aphasias. Neurobiol Aging. 2013;34:1687–99.
Mesulam MM. Slowly progressive aphasia without generalized dementia. Ann Neurol. 1982;11:
592–8.
Mesulam MM. Primary progressive aphasia. Ann Neurol. 2001;49:425–32.
Mesulam MM. Primary progressive aphasia–a language-based dementia. N Engl J Med.
2003;349:1535–42.
Mesulam M, Johnson N, Krefft TA, Gass JM, Cannon AD, et al. Progranulin mutations in primary
progressive aphasia: the PPA1 and PPA3 families. Arch Neurol. 2007;64:43–7.
Mesulam M, Wicklund A, Johnson N, Rogalski E, Leger GC, et al. Alzheimer and frontotemporal
pathology in subsets of primary progressive aphasia. Ann Neurol. 2008;63:709–19.
Mukherjee O, Pastor P, Cairns NJ, Chakraverty S, Kauwe JS, et al. HDDD2 is a familial frontotem-
poral lobar degeneration with ubiquitin-positive, tau-negative inclusions caused by a missense
mutation in the signal peptide of progranulin. Ann Neurol. 2006;60:314–22.
Mummery CJ, Patterson K, Price CJ, Ashburner J, Frackowiak RS, et al. A voxel-based morphom-
etry study of semantic dementia: relationship between temporal lobe atrophy and semantic
memory. Ann Neurol. 2000;47:36–45.
230 K.A. Josephs and J.L. Whitwell
Abstract Although it has been nearly 50 years since its first description of clinical
and radiological, there is considerable uncertainty about the diagnosis of normal
pressure hydrocephalus because it shares the semiotics with the group of dementias.
Hakim’s triad (impaired gait, initially type clumsiness of the lower limbs followed
over time by inability to ambulate or maintain an erect posture; cognitive impair-
ment, initially limited to worsening deficits in memory fixation and execution of
complex actions; the urinary disorder, initially type “urgent urination” and then
complete urinary incontinence) characterizes the progressive course of the adult
chronic hydrocephalus. The clinical onset is typically nonspecific, subtle, and most
often monosymptomatic. The first diagnostic procedure is a head CT scan and/or
brain MRI, which shows an abnormal dilatation of the lateral ventricles and the
third ventricle, associated to variable brain atrophy. Not all subjects will develop a
set of symptoms, since the altered cerebrospinal fluid dynamics can remain stable
for many years or get progressively worse until the appearance of the clinical triad
of normal pressure hydrocephalus. The test of intrathecal infusion (Katzman test)
carried out at constant speed with the introduction of saline solution into the lumbar
subarachnoid space and the concomitant detection of cerebrospinal fluid pressure
establishes that patients with outflow resistance ranging from 12 to 19 mmHg/ml/
min can improve clinically after surgery. This method requires extensive and pro-
longed experience of the center of application and the use of computer systems. The
withdrawal of lumbar cerebrospinal fluid provides for the evacuation of 30–50 cc of
P. Missori, MD (*)
Department of Neurology and Psychiatry, Policlinico “Umberto I”,
“Sapienza” University of Rome, Rome, Italy
e-mail: [email protected]
A. Daniele, MD, PhD
Institute of Neurology, Policlinico “A. Gemelli”, Catholic University of the Sacred Heart,
Rome, Italy
C. Colosimo, MD
Department of Neurology and Psychiatry, “Sapienza” University of Rome, Rome, Italy
cerebrospinal fluid by lumbar puncture under local anesthesia, preceded and fol-
lowed by gait assessment and neuropsychological tests. It also uses the continuous
withdrawal of CSF with an intrathecal catheter placed for 3 days, in order to drain
approximately 135 ml/24 h and the aim of reducing false negatives. After surgery,
the patient is usually able to regain a good quality of life, with independence in daily
living activities. The duration of such postsurgical improvement is variable, but
patients may improve again readjusting the opening pressure of the programmable
valve, although a high comorbidity index is strictly related to a poor outcome.
Historical Remarks
the pressure of the CSF was in the normal range, the disease was named “normal
pressure” hydrocephalus (NPH). For some years, the term was unchanged, but due
to the unknown origin of the disease, the term “idiopathic” NPH was added (Black
1980; Greenberg et al. 1977; Shenkin et al. 1975). During the second half of the
twentieth century, the diagnostic and therapeutic procedures of withdrawal of CSF
became more common after the introduction of spinal catheters and continuous CSF
drainage (Aitken and Drake 1964; Matera and Althabe 1962; Vourc’h 1963; Vourc’h
and Rougerie 1960). The advent of computed tomography represented a milestone
of the radiological diagnosis (Gunasekera and Richardson 1977; Jacobs and Kinkel
1976), with a transposition of the Evans’ index values for hydrocephalus’ diagnosis
from the pneumoencephalography (Gawler et al. 1976; Synek et al. 1976). A further
progress came with the “Charles Miller Fisher test,” in which clinical improvement
persisted many months after a single 20–30 ml CSF tap in patients with NPH (Fisher
1978). This procedure was considered more effective if extended to more days (Di
Lauro et al. 1986). Katzman was the first to suggest an infusion of physiologic solu-
tion into the lumbar subarachnoid space, with monitoring of CSF pressure (Katzman
and Hussey 1970). After infusion of physiologic solution into the lumbar subarach-
noid space and monitoring CSF pressure, the capacity to absorb additional physi-
ologic solution was reduced in patients with NPH and the CSF pressure quickly
increased. The last remark in this history is the guidelines for the diagnosis and
management of idiopathic normal pressure hydrocephalus which came to light from
an international group of researchers in 2005 (Marmarou et al. 2005b). In a five-step
document, all the solved and unsolved questions were addressed, to offer an essen-
tial reference from which prospective randomized studies could originate.
Introduction
The CSF is produced by the choroid plexuses of the lateral and fourth ventricles
and from the capillary ultrafiltrate of the Virchow-Robin spaces, at a rate varying
between 0.2 and 0.6 ml per minute or 600 and 700 ml per day (Wright 1978). This
amount of fluid is reabsorbed through three main systems: the arachnoid villi into
the cerebral venous sinuses, the subarachnoid space along thoracic and lumbosacral
spinal rootlets, and the olfactory pathway (Edsbagge et al. 2004; Johnston 2003;
Luedemann et al. 2002; Mollanji et al. 2002). The equilibrium between produc-
tion and reabsorption allows normal ventricle size during life, and every patho-
logical condition which alters this CSF balance can produce progressive ventricular
enlargement, up to hydrocephalus. This simple concept is challenged by some
brain degenerative disorders (i.e., Alzheimer’s disease, progressive supranuclear
palsy, frontotemporal dementia, vascular dementia, dementia with Lewy bodies,
Parkinson’s disease), in which a progressive tissue loss is replaced by CSF. In these
patients, the atrophy of the brain is the primary event and the increase of the amount
of CSF results from neuronal loss. The characteristic secondary ventricular enlarge-
ment resembles moderate hydrocephalus, but the clinical picture must be carefully
234 P. Missori et al.
evaluated together with neurological examination and brain imaging, since the cor-
rect diagnosis of NPH is promoted by additional tests. Other conditions which may
resemble the clinical picture of NPH (either in early stages or later in the course of
the primary disease) include secondary post-traumatic or posthemorragic hydro-
cephalus, systemic lupus erythematosus, and rarely neurosarcoidosis (Honda et al.
2004; Westhout and Linskey 2008; Wikkelsø et al. 1982). In these conditions, the
clinical course is characterized by faster progression, unlike in NPH patients. Other
rare conditions in which an NPH can occur are myotonic dystrophy and Paget’s
disease (Christensen 1988; Delavallée and Raftopoulos 2006; Martin et al. 1985;
Moiyadi et al. 2006; Riggs et al. 1985; Roohi et al. 2005).
Epidemiology
Few studies reported a possible familial association in NPH (Portenoy et al. 1984). The
description of a large family with four members showing a late-onset primary NPH
across three generations suggests the opportunity to search for underdiagnosed and
asymptomatic NPH in adult family members of patients with NPH (Takahashi et al.
2011). An autosomal dominant transmission has been hypothesized. Another study
reported two sisters with clinical and radiological features of NPH, who underwent
shunt placement with clinical improvement (Cusimano et al. 2011). In both sisters, an
epsilon3-epsilon3 genotype of the apolipoprotein E (ApoE) on chromosome 19 was
detected. Familial aggregation of NPH among first-degree relatives is suggested by an
incidence of 7.1 and 0.7 % among control relatives (McGirr and Cusimano 2012).
Clinical Picture
The most characteristic picture in patients with NPH is the Hakim’s triad: gait dis-
turbance, urinary incontinence, and cognitive decline (Adams et al. 1965). Gait dis-
turbances may occur as the first symptom and vary in the course. In early phases, the
patient may complain of a slow, wide-based walking with small steps and sudden
drops, charged to the shoes or uneven floor (McHugh 1964; Missori et al. 2010;
Roger et al. 1950). In later stages, a cane or wheeled walker may be needed. In late
stages, due to tendency to fall, the patient is completely unable to walk. Differentiating
between Parkinson’s disease and NPH can be challenging for the practicing clini-
cian; in NPH usually the motor impairment is mainly confined in the lower limbs
(lower-body parkinsonism) and the response to levodopa is poor or absent, but in
some cases, the differential diagnosis may not be so straightforward (Table 14.2).
Dopamine transporter brain imaging (DaTSCAN) using single-photon emission
tomography (SPECT) may be useful, since it shows an abnormal picture in all cases
of PD, while the uptake is usually normal in NPH. Disturbances of micturition usu-
ally appear in association with other symptoms of NPH. In early stages NPH patients
often complain daily need to urinate more often than usual, sometimes with urgency.
When the disease worsens, occasional incontinence happens. This symptom is very
harmful to patients and relatives, and in males the attention is often focused on pros-
tatic function and sometimes surgery is even carried out to treat urinary symptoms.
In patients with NPH SPECT, studies showed that urinary dysfunction may be
closely related with right frontal cerebral hypoperfusion, the most common
urodynamic abnormality being detrusor overactivity (Sakakibara et al. 2008, 2012).
14 Normal Pressure Hydrocephalus 237
Table 14.2 Differences between normal pressure hydrocephalus and Parkinson’s disease gait
Features Normal pressure hydrocephalus Parkinson’s disease
Early stages May occur as the first symptom Usually not the first symptom
Clumsiness of the lower limbs Subtle, if present
Loss of unilateral arm swing
Middle stages Wide-based walking Normal or narrow-based
walking
Small steps Small steps
Stooped stance Stooped stance
Sudden drops Sometimes, festination
Charged to the shoes or uneven floor and freezing
Outward rotated feet and a diminished
height of the steps
Frequent freezing
Late stages Tendency to fall Tendency to fall
Inability to ambulate or maintain Inability to ambulate
an erect posture or maintain an erect posture
Frequent freezing Frequent festination and
freezing
Speed Slow Slow
Stride length Reduced Reduced
Rhythmicity Reduced Reduced
Symmetry Reduced Reduced
Improvement with Mild Moderate
external clues
Response to levodopa None or mild Moderate
Cognitive and behavioral symptoms are key clinical features of NPH and may
appear in early disease stages (Hashimoto et al. 2010). In individual patients with
NPH, however, the cognitive impairment may vary largely, ranging from a mild
cognitive decline to a moderate or severe dementia in other patients.
Various neuropsychological studies have supported the view suggesting that
patients with NPH may frequently show a “subcortical” pattern of cognitive impair-
ment (Cummings and Benson 1984; Devito et al. 2005), which is often seen in
pathologies affecting various subcortical structures and is mainly characterized
by the occurrence of mental slowness, deficits of attention and frontal executive
functions (planning, problem-solving, set-shifting), relatively mild deficits of long-
term memory, and behavioral changes (apathy most frequently, depressive symp-
toms in some patients). Since deficits of executive functions frequently arise from
a dysfunction of the frontal lobes, which are abundantly connected to various sub-
cortical structures (Alexander et al. 1986), the term “frontal-subcortical” cogni-
tive impairment/dementia has also been proposed. Such most frequently reported
“frontal-subcortical” pattern of cognitive impairment observed in patients with
NPH does usually not include aphasic, apraxic, or agnostic deficits, which are by
contrast common in patients with “cortical” dementias (in primis, Alzheimer’s dis-
ease). As to long-term memory deficits, it has been shown that patients with NPH
238 P. Missori et al.
in early disease stages have usually a relatively preserved ability to store informa-
tion in long-term memory systems (at variance with patients with Alzheimer’s dis-
ease since early disease stages), but are usually impaired mostly in the retrieval of
stored information from long-term memory systems and therefore may benefit from
external cues aimed at facilitating such retrieval (Iddon et al. 1999). This pattern
of memory impairment of NPH patients may be shared by other conditions with a
“frontal-subcortical” pattern of cognitive impairment, such as Parkinson’s disease
(Taylor et al. 1986). In addition, deficits of spatial long-term memory (Iddon et al.
1999), constructional praxic abilities (Goodman and Meyer 2001), and psychomo-
tor speed (Kaye et al. 1990) have been detected in patients affected by NPH.
In a recent study (Saito et al. 2011) aimed at further investigating cognitive
functioning in patients affected by idiopathic NPH, 32 patients with NPH who sig-
nificantly improved after surgery were recruited and underwent a quite extensive
neuropsychological assessment before (n = 32) and 1 year (n = 26) after CSF shunt
surgery. Moreover, a group of patients with Alzheimer’s disease (n = 32) matched to
the NPH group as for several clinical and demographic variables (including overall
cognitive functioning as measured by performance on the mini-mental state exami-
nation, MMSE) and a group of healthy controls (HCs) (n = 30) were recruited. The
results of such study show that patients with NPH showed a significantly worse
performance as compared to healthy controls on most cognitive tasks (MMSE,
tasks of verbal and spatial short-term memory, phonological and semantic verbal
fluency, episodic verbal memory, several tasks assessing visual discrimination, and
the Frontal Assessment Battery (FAB) assessing executive functions), while no sig-
nificant difference was found between the NPH and HCs on a task of object nam-
ing. Moreover, the NPH group performed significantly worse than the AD group
on some cognitive tasks (spatial short-term memory, phonological verbal fluency,
FAB, some tasks assessing visual discrimination), while the AD group was more
impaired than the NPH group on specific cognitive variables assessing episodic
verbal memory. Although deficits of frontal cognitive functions (including execu-
tive functions) were the most prominent in the NPH group, this study pointed out
that memory deficits and visuoperceptual and visuospatial deficits may also occur
in patients with NPH. This recent study (Saito et al. 2011) suggests that, although
deficits of frontal cognitive functions (including executive functions) due to a dis-
ruption of frontal-subcortical circuits are the most prominent in patients with NPH,
memory deficits and visuoperceptual and visuospatial deficits (due to a disruption
of neural circuits involving more “posterior” cortical and subcortical structures)
may also occur in patients with NPH.
As to other behavioral symptoms (Bloom and Kraft 1998; Kito et al. 2009; Koch
et al. 2009), besides apathy and depression, it has been less commonly reported that
patients with NPH may show manic and psychotic symptoms (Kwentus and Hart
1987; Rice and Gendelman 1973), including aggressivity, hypersexuality, and delu-
sional jealousy (Yusim et al. 2008).
A distinguishing feature of NPH is the relatively slow but progressive clinical
evolution (12–24 months), as the patient becomes unable to carry out everyday
activities and to look after himself. However, some patients may show a slower
14 Normal Pressure Hydrocephalus 239
a b
Fig. 14.1 (a) A 72-year-old female with gait instability. CT scan shows Evans’ index of
39.62 mm/131.88 mm = 0.30. (b) The same patient after 55 months complaining short memory
impairment, recurrent falls, and urgent urination. On control CT scan the frontal horns appear more
enlarged with Evans’ index increase: 43.02 mm/132.84 mm = 0.32
clinical progression both for very mild symptoms and for misdiagnosis. In these
patients, neuroimaging commonly shows a ventriculomegaly. When acute or sub-
acute symptoms of intracranial pressure (headache, alteration of consciousness, cra-
nial nerve deficits, vomiting, dizziness) occur in a short period of time (1–2 weeks)
associated to brain scan of ventricular enlargement, a secondary hydrocephalus
other than NPH or a cerebrovascular disease should be considered as the causative
condition. In the cases in which a previous neuroradiological imaging is available,
a comparison of the Evans’ index between the old and new examination allows a
more secure and reliable assessment of the ventricular progression and of clinical
diagnosis (Figs. 14.1 and 14.2).
Various grading scales have been reported for assessment of patients with NPH.
The use of functional scales based on the degree of disability or stroke rehabilita-
tion scales have been proposed, but these scales were not devised to assess changes
in the severity of main symptoms of NPH (Klinge et al. 2005). The need to develop
assessment scales to be largely shared in the clinical evaluation of patients with NPH
prompted the proposal of two new grading scales (Hellström et al. 2012; Kubo et al.
2008). A major advantage of both scales is the possibility to compare the neurologi-
cal conditions before and after each diagnostic or therapeutic procedure. Cognitive,
gait, and urinary symptoms are assessed in both scales (Hellström et al. 2012; Kubo
et al. 2008), while the examination of balance only in one (Hellström et al. 2012).
In the Swedish scale (Hellström et al. 2012), three different professionals evaluate
240 P. Missori et al.
a b
Fig. 14.2 (a) A 61-year-old female with short-term memory deficit. MRI shows Evans’ index
of 41.1 mm/142 mm = 0.28. (b) The same patient after 16 years with mild cognitive impair-
ment, progressive gait disturbance, and occasional urinary incontinence: Evans’ index of
51.2 mm/140.1 mm = 0.36. Note the secondary brain atrophy with widening of the Sylvian cisterns
separately the three main groups (cognitive, gait, urinary) of symptoms, and scores
may range from 0 to 100, where 100 is the performance of an age-matched healthy
population. Such scale discriminates well between levels of severity. Following sur-
gery, a patient is considered improved if a >5-point increment can be detected on the
scale. In the Japanese scale (Kubo et al. 2008), the clinical assessment is performed
by two independent physicians. Patients with NPH were assessed with this scale
after a CSF tap test and after shunt surgery. For the triad of symptoms, scores may
range from 0 to 4, with higher scores indicating more severe symptoms. Patients
who obtain an improvement in at least 1 of the 3 domains are judged to have a clini-
cally significant improvement. Reliability and validity in predicting shunt respon-
siveness were assessed for this scale.
Neuroradiological Evaluation
a b
Fig. 14.3 (a) A 67-year-old male with slight cognitive impairment, progressive gait disturbance,
and occasional urinary incontinence. Neuroradiological examinations from July 1993 show pro-
gressive ventricular enlargement. In the last MRI (June 2012), the Evans’ index is
59.4 mm/137.60 mm = 0.43, and on T2 coronal image at the level of the posterior commissure, the
callosal angle is more than 90°. After a positive tap test, a programmable shunt was inserted. Ten
months after surgery the patient has a lasting clinical benefit on gait disturbance and urinary incon-
tinence. (b) A 64-year-old female with gait instability and urinary incontinence. On MRI the
Evans’ index shows 47.4 mm/127.1 mm = 0.37. On coronal T1 image at the level of the posterior
commissure, the callosal angle is more than 90°. Two years after surgery the patient is asymptom-
atic. The callosal angle must be interpreted in association to the clinical symptoms and all the
neuroradiological findings
the cranium at the level of the Monro’s foramens in the frontal horns (Evans 1942).
A value equal or more than 0.3 support the diagnosis of probable NPH (Ishikawa
et al. 2008; Marmarou et al. 2005b), but a lower value cannot exclude this diagnosis
(Naruse and Matsuoka 2013). After shunt for NPH, the majority of the patients
assessed by the Evans’ index showed no ventricular changes, despite a satisfactory
clinical improvement (Meier et al. 2003). Since remarkable differences were found
in the Evans’ index calculated at different planes on CT scans, its value has been
questioned and not considered an ideal tool for diagnosis of NPH (Ambarki et al.
2010; Toma et al. 2011a). Other simple criteria which support a radiological diagno-
sis of NPH include a tight high-convexity and medial subarachnoid spaces, enlarged
Sylvian fissures, a small callosal angle (under 90°) on the coronal section of MRI,
and periventricular signal changes (Fig. 14.3) (Hashimoto et al. 2010; Ishii et al.
2008; Kitagaki et al. 1998; Sasaki et al. 2008). The aqueductal CSF stroke volume,
which is the mean volume of CSF passing through the Sylvian aqueduct during
cardiac systole and diastole, is a valuable tool in the preoperative evaluation of NPH
patients. Higher aqueductal stroke volume in patients with short clinical history
may be associated to a post-shunt improvement of clinical symptoms (Bradley et al.
1996; El Sankari et al. 2012; Scollato et al. 2009). The assessment of the ventricular
size and volume may show very variable changes and may not easily correlate with
242 P. Missori et al.
clinical improvement after CSF withdrawal (Anderson et al. 2002; Lenfeldt et al.
2012; McConnell et al. 2004; Palm et al. 2006). However, if the ventricular and
brain volumes are assessed with a specific software, after prolonged external lumbar
drainage, the brain volume may increase and the ventricular volume may decrease,
more markedly than after a single tap withdrawal (Singer et al. 2012). A study with
functional MRI in NPH patients showed a bilateral increased activation in the sup-
plementary motor area after CSF removal (Lenfeldt et al. 2008b). Moreover, proton
magnetic resonance spectroscopy revealed normalization of N-acetyl-aspartate in
the frontal white matter after a 3-day lumbar drainage (Lenfeldt et al. 2008a).
Clinical evidence supports the value of surgery in NPH patients, despite the lack
of randomized controlled trials comprising surgical treatment versus no surgery
(Esmonde and Cooke 2002; Toma et al. 2011b, 2012). The best surgical technique to
treat NPH should be aimed at obtaining a diversion of flow of CSF and at favoring
the impaired processes involved in CSF reabsorption. There are two surgical options
to enable such events: the endoscopic third ventriculostomy and the placement of
a CSF shunt from a cerebral ventricle into an absorbing cavity or the vascular sys-
tem. The endoscopic third ventriculostomy is performed through a frontal burr hole.
The endoscope is inserted in the brain to gain the Monro’s foramen and the floor of
the third ventricle. After the identification of landmarks (the mammillary bodies and
tuber cinereum), a catheter pierces a chosen place to put CSF flow between the inter-
peduncular cistern and the third ventricle. A low complication rate, a low mortality
rate, and good neurological improvement are reported (Bouras and Sgouros 2012;
Fountas et al. 2012; Gangemi et al. 2004, 2008). Since in the long-term follow-up
closure of the fenestration and recurrent symptoms after initial successful treatment
occur, these patients should be monitored for some years (Amini and Schmidt 2005;
Cage et al. 2011; Fabiano et al. 2010; Longatti et al. 2004). A randomized clinical
trial comparing the endoscopic third ventriculostomy with the ventriculoperitoneal
shunt (shunt into the abdominal cavity) shows that the shunt has a better neurologi-
cal outcome 12 months after surgery (Pinto et al. 2013). Indeed the surgical shunt is
the most widely performed procedure to treat the NPH. Over the past few decades,
the rate of complications after surgical treatment has decreased considerably, due
to the improvement of the techniques and valve systems (Black 1980; Farahmand
et al. 2009; Greenberg et al. 1977; Larsson et al. 1991; Poca et al. 2004; Savitz and
Bobroff 1999; Zemack and Romner 2008). The ventriculoperitoneal shunt is more
widely performed as compared with the ventriculoatrial shunt, probably due to early
reports indicating a less risk of serious complications and neurosurgeon’s confidence
for the abdominal insertion in pediatric patients (Keucher and Mealey 1979; Mazza
et al. 1980; Olsen and Frykberg 1983; Vernet et al. 1993). This conclusion has been
initially transferred to adult patients (Lam and Villemure 1997). More recent surgi-
cal series show no difference between ventriculoperitoneal and ventriculoatrial shunt
in adult hydrocephalus, and in some cases ventriculoatrial shunt is carried out after
a failure of the ventriculoperitoneal shunt (Farahmand et al. 2009; Murakami et al.
2010; Stranjalis et al. 2012; Zhang et al. 2009). An alternative surgical option in
patients with NPH is the lumboperitoneal shunt (Bloch and McDermott 2012; Chang
et al. 1999; Yadav et al. 2010). Since NPH is a communicating hydrocephalus, it is
244 P. Missori et al.
possible to drain the lumbar CSF into the abdominal cavity, through an intrathecal
spinal catheter, with a programmable valve. The technique is apparently simple, but
the rate of complication is not low (Karabatsou et al. 2004; Wang et al. 2007). The
ventriculopleural shunt (into the space between the visceral and parietal pleura of
the lungs) is very rarely performed in patients with NPH and is indicated when other
routes are not available (Megison and Benzel 1988). The introduction of programma-
ble (flow-regulated) versus nonadjustable (differential-pressure) valves has changed
the surgical view: the amount of CSF withdrawal can be varied according to the clini-
cal changes which can occur in the early or late postoperative follow-up (Zemack and
Romner 2000). Readjusting the opening pressure may decrease the incidence of clini-
cal complications of underdrainage or overdrainage (Freimann and Sprung 2012).
A low CSF pressure has been proven to have a better outcome, but it is plagued
by overdrainage complications (Boon et al. 1998). Accordingly, the combination of
programmable valve with a gravitational valve (which eliminates overdrainage by
increasing resistance as patient moves upright) may improve the outcome (Lemcke
and Meier 2010; Meier and Lemcke 2006). A recent trial showed that a gravitational
valve (which switches between a low pressure mode in the supine position and a high
pressure mode in the upright position) reduces the risk of overdrainage complica-
tions, as compared with a standard programmable valve (Lemcke et al. 2013).
Outcome
The most relevant feature of patients who undergo surgical treatment for NPH is the
rapid neurological improvement in the early postoperative period. In the majority of
cases, patients regain the ability to walk and control micturition but also, albeit less
frequently, of cognitive and behavioral symptoms. Accordingly, NPH is included
among the infrequent conditions of reversible dementia. In particular, it has been
observed that deficits of episodic verbal memory (Gallassi et al. 1991), construc-
tional praxic abilities (Goodman and Meyer 2001), and psychomotor speed (Kaye
et al. 1990) may improve after shunt surgery, at least in subgroups of NPH patients.
On the other hand, it has been reported that deficits of executive functions may not
improve after surgery (Iddon et al. 1999). By contrast, in a more recent study (Saito
et al. 2011), 1 year after CSF surgery, the NPH group showed a significant improve-
ment on tasks assessing frontal functions (FAB), including executive functions.
In a recent neuropathological study (Cabral et al. 2011), a group of nine patients
with a clinical diagnosis of NPH was examined, and it was observed that in eight out
of such nine patients, there were neuropathological changes consistent with AD.
This study on a small sample of patients with a clinical diagnosis of NPH suggests
that AD may be a frequent pathological comorbidity in patients with NPH and such
comorbidity may at least partially preclude cognitive improvement after surgery.
A study carried out in 36 patients with NPH (Chang et al. 2006) who underwent
neuropsychological testing before and after ventriculoperitoneal shunt insertion
reported that one third of patients showed good cognitive improvement (defined as
improvement by at least 25 % on at least half of the cognitive tests administered).
14 Normal Pressure Hydrocephalus 245
References
Adams RD, Fisher CM, Hakim S, Ojemann RG, Sweet WT. Symptomatic occult hydrocephalus with
“normal” cerebrospinal fluid pressure: a treatable syndrome. N Engl J Med. 1965;273:117–26.
Aitken RR, Drake CG. Continuous spinal drainage in the treatment of postoperative cerebrospinal-
fluid fistulae. J Neurosurg. 1964;21:275–7.
Albert F. Lumbar puncture as applied to craniocerebral surgery. Ann Surg. 1918;68:549–50.
Alexander GE, DeLong MR, Strick PL. Parallel organization of functionally segregated circuits
linking basal ganglia and cortex. Annu Rev Neurosci. 1986;9:357–81.
Ambarki K, Israelsson H, Wåhlin A, Birgander R, Eklund A, Malm J. Brain ventricular size in
healthy elderly: comparison between Evans index and volume measurement. Neurosurgery.
2010;67:94–9.
Amini A, Schmidt RH. Endoscopic third ventriculostomy in a series of 36 adult patients. Neurosurg
Focus. 2005;19:E9.
Anderson RC, Grant JJ, de la Paz R, Frucht S, Goodman RR. Volumetric measurements in the detec-
tion of reduced ventricular volume in patients with normal-pressure hydrocephalus whose clini-
cal condition improved after ventriculoperitoneal shunt placement. J Neurosurg. 2002;97:73–9.
Aquilina K, Pople IK, Sacree J, Carter MR, Edwards RJ. The constant flow ventricular infusion
test: a simple and useful study in the diagnosis of third ventriculostomy failure. J Neurosurg.
2012;116:445–52.
Aygok G, Marmarou A, Young HF. Three-year outcome of shunted idiopathic NPH patients. Acta
Neurochir Suppl. 2005;95:241–5.
246 P. Missori et al.
Dandy WE. Ventriculography following the injection of air into the cerebral ventricles. Ann Surg.
1918;68:5–11.
Delavallée M, Raftopoulos C. Normal pressure hydrocephalus in a patient with myotonic dystro-
phy: case report with a 10-year follow-up. Neurosurgery. 2006;58:E796.
Devito EE, Pickard JD, Salmond CH, Iddon JL, Loveday C, Sahakian BJ. The neuropsychology of
normal pressure hydrocephalus (NPH). Br J Neurosurg. 2005;19:217–24.
Di Lauro L, Mearini M, Bollati A. The predictive value of 5 days CSF diversion for shunting in
normal pressure hydrocephalus. J Neurol Neurosurg Psychiatry. 1986;49:842–3.
Edsbagge M, Tisell M, Jacobsson L, Wikkelso C. Spinal CSF absorption in healthy individuals.
Am J Physiol Regul Integr Comp Physiol. 2004;287:R1450–5.
El Sankari S, Fichten A, Gondry-Jouet C, Czosnyka M, Legars D, Deramond H, Balédent O.
Correlation between tap test and CSF aqueductal stroke volume in idiopathic normal pressure
hydrocephalus. Acta Neurochir Suppl. 2012;113:43–6.
Esmonde T, Cooke S. Shunting for normal pressure hydrocephalus (NPH). Cochrane Database
Syst Rev. 2002;(3):CD003157.
Evans WA. An encephalographic ratio for estimating ventricular enlargement and cerebral atrophy.
Arch Neurol Psychiatry. 1942;47:931–7.
Fabiano AJ, Doyle K, Grand W. Delayed stoma failure in adult communicating hydrocephalus
after initial successful treatment by endoscopic third ventriculostomy: case report. Neurosurgery.
2010;66:E1210–1.
Farahmand D, Hilmarsson H, Högfeldt M, Tisell M. Perioperative risk factors for short term shunt
revisions in adult hydrocephalus patients. J Neurol Neurosurg Psychiatry. 2009;80:1248–53.
Fisher CM. Communicating hydrocephalus. Lancet. 1978;1(8054):37.
Fisher CM. Hydrocephalus as a cause of disturbances of gait in the elderly. Neurology.
1982;32:1358–63.
Fountas KN, Kapsalaki EZ, Paterakis KN, Lee GP, Hadjigeorgiou GM. Role of endoscopic third
ventriculostomy in treatment of selected patients with normal pressure hydrocephalus. Acta
Neurochir Suppl. 2012;113:129–33.
Freimann FB, Sprung C. Shunting with gravitational valves–can adjustments end the era of revi-
sions for overdrainage-related events?: clinical article. J Neurosurg. 2012;117:1197–204.
Gallassi R, Morreale A, Montagna P, Sacquegna T, Di Sarro R, Lugaresi E. Binswanger’s disease
and normal-pressure hydrocephalus. Clinical and neuropsychological comparison. Arch
Neurol. 1991;48:1156–9.
Gangemi M, Maiuri F, Buonamassa S, Colella G, de Divitiis E. Endoscopic third ventriculostomy
in idiopathic normal pressure hydrocephalus. Neurosurgery. 2004;55:129–34.
Gangemi M, Maiuri F, Naddeo M, Godano U, Mascari C, Broggi G, Ferroli P. Endoscopic third
ventriculostomy in idiopathic normal pressure hydrocephalus: an Italian multicenter study.
Neurosurgery. 2008;63:62–7.
Gawler J, Du Boulay GH, Bull JW, Marshall J. Computerized tomography (the EMI Scanner): a
comparison with pneumoencephalography and ventriculography. J Neurol Neurosurg
Psychiatry. 1976;39:203–11.
Goodman M, Meyer WJ. Dementia reversal in post-shunt normal pressure hydrocephalus pre-
dicted by neuropsychological assessment. J Am Geriatr Soc. 2001;49:685–6.
Greenberg JO, Shenkin HA, Adam R. Idiopathic normal pressure hydrocephalus– a report of 73
patients. J Neurol Neurosurg Psychiatry. 1977;40:336–41.
Greenfield JG. Lumbar puncture in diagnosis. Br Med J. 1935;2:1265–7.
Gunasekera L, Richardson AE. Computerized axial tomography in idiopathic hydrocephalus.
Brain. 1977;100:749–54.
Hashimoto M, Ishikawa M, Mori E, Kuwana N, Study of INPH on neurological improvement
(SINPHONI). Diagnosis of idiopathic normal pressure hydrocephalus is supported by MRI-
based scheme: a prospective cohort study. Cerebrospinal Fluid Res. 2010;7:18.
Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US popula-
tion: prevalence estimates using the 2000 census. Arch Neurol. 2003;60:1119–22.
Hellström P, Klinge P, Tans J, Wikkelsø C. A new scale for assessment of severity and outcome in
iNPH. Acta Neurol Scand. 2012;126:229–37.
248 P. Missori et al.
Lam CH, Villemure JG. Comparison between ventriculoatrial and ventriculoperitoneal shunting in
the adult population. Br J Neurosurg. 1997;11:43–8.
Larsson A, Wikkelsö C, Bilting M, Stephensen H. Clinical parameters in 74 consecutive patients
shunt operated for normal pressure hydrocephalus. Acta Neurol Scand. 1991;84:475–82.
Lemcke J, Meier U. Improved outcome in shunted iNPH with a combination of a Codman Hakim
programmable valve and an Aesculap-Miethke Shunt Assistant. Cent Eur Neurosurg. 2010;71:
113–6.
Lemcke J, Meier U. Idiopathic normal pressure hydrocephalus (iNPH) and co-morbidity: an out-
come analysis of 134 patients. Acta Neurochir Suppl. 2012;114:255–9.
Lemcke J, Meier U, Müller C, Fritsch MJ, Kehler U, Langer N, Kiefer M, Eymann R, Schuhmann
MU, Speil A, Weber F, Remenez V, Rohde V, Ludwig HC, Stengel D. Safety and efficacy of
gravitational shunt valves in patients with idiopathic normal pressure hydrocephalus: a prag-
matic, randomised, open label, multicentre trial (SVASONA). J Neurol Neurosurg Psychiatry.
2013;84:850–7.
Lenfeldt N, Hauksson J, Birgander R, Eklund A, Malm J. Improvement after cerebrospinal fluid
drainage is related to levels of N-acetyl-aspartate in idiopathic normal pressure hydrocephalus.
Neurosurgery. 2008a;62:135–41.
Lenfeldt N, Larsson A, Nyberg L, Andersson M, Birgander R, Eklund A, Malm J. Idiopathic nor-
mal pressure hydrocephalus: increased supplementary motor activity accounts for improve-
ment after CSF drainage. Brain. 2008b;131:2904–12.
Lenfeldt N, Hansson W, Larsson A, Birgander R, Eklund A, Malm J. Three-day CSF drainage
barely reduces ventricular size in normal pressure hydrocephalus. Neurology. 2012;79:237–42.
Longatti PL, Fiorindi A, Martinuzzi A. Failure of endoscopic third ventriculostomy in the treatment
of idiopathic normal pressure hydrocephalus. Minim Invasive Neurosurg. 2004;47:342–5.
Luedemann W, Kondziella D, Tienken K, Klinge P, Brinker T, Berens von Rautenfeld D. Spinal
cerebrospinal fluid pathways and their significance for the compensation of kaolin-
hydrocephalus. Acta Neurochir Suppl. 2002;81:271–3.
MacCordick AH. Observations on the value of lumbar puncture. Can Med Assoc
J. 1922;12:168–70.
Malm J, Lundkvist B, Eklund A, Koskinen LO, Kristensen B. CSF outflow resistance as predictor
of shunt function. A long-term study. Acta Neurol Scand. 2004;110:154–60.
Malm J, Graff-Radford NR, Ishikawa M, Kristensen B, Leinonen V, Mori E, Owler BK, Tullberg
M, Williams MA, Relkin NR. Influence of comorbidities in idiopathic normal pressure hydro-
cephalus – research and clinical care. A report of the ISHCSF task force on comorbidities in
INPH. Fluids Barriers CNS. 2013;10:22.
Marmarou A, Bergsneider M, Klinge P, Relkin N, Black PM. The value of supplemental prognos-
tic tests for the preoperative assessment of idiopathic normal-pressure hydrocephalus.
Neurosurgery. 2005a;57(3 Suppl):S17–28.
Marmarou A, Bergsneider M, Relkin N, Klinge P, Black PM. Development of guidelines for idio-
pathic normal-pressure hydrocephalus: introduction. Neurosurgery. 2005b;57(3 Suppl):S1–3.
Marmarou A, Young HF, Aygok GA. Estimated incidence of normal pressure hydrocephalus and
shunt outcome in patients residing in assisted-living and extended-care facilities. Neurosurg
Focus. 2007;22:E1.
Martin BJ, Roberts MA, Turner JW. Normal pressure hydrocephalus and Paget’s disease of bone.
Gerontology. 1985;31:397–402.
Matera R, Althabe BM. Continuous lumbar drainage of the cerebrospinal fluid in neurosurgery.
Sem Med. 1962;121:1083–7.
Matousek M, Wikkelsö C, Blomsterwall E, Johnels B, Steg G. Motor performance in normal pres-
sure hydrocephalus assessed with an optoelectronic measurement technique. Acta Neurol
Scand. 1995;91:500–5.
Matson DD. A new operation for the treatment of communicating hydrocephalus: report of a case
secondary to generalized meningitis. J Neurosurg. 1949;6:238–47.
Mazza C, Pasqualin A, Da Pian R. Results of treatment with ventriculoatrial and ventriculoperitoneal
shunt in infantile nontumoral hydrocephalus. Childs Brain. 1980;7:1–14.
250 P. Missori et al.
McConnell KA, Zou KH, Chabrerie AV, Bailey NO, Black PM. Decreases in ventricular volume
correlate with decreases in ventricular pressure in idiopathic normal pressure hydrocephalus
patients who experienced clinical improvement after implantation with adjustable valve shunts.
Neurosurgery. 2004;55:582–92.
McGirr A, Cusimano MD. Familial aggregation of idiopathic normal pressure hydrocephalus:
novel familial case and a family study of the NPH triad in an iNPH patient cohort. J Neurol Sci.
2012;321:82–8.
McHugh PR. Occult hydrocephalus. Q J Med. 1964;33:297–308.
Megison DP, Benzel EC. Ventriculo-pleural shunting for adult hydrocephalus. Br J Neurosurg.
1988;2:503–5.
Meier U, Lemcke J. Clinical outcome of patients with idiopathic normal pressure hydrocephalus
three years after shunt implantation. Acta Neurochir Suppl. 2006;96:377–80.
Meier U, Lemcke J. The influence of co-morbidity on the postoperative outcomes of patients with
idiopathic normal pressure hydrocephalus (iNPH). Acta Neurochir Suppl. 2008;102:141–4.
Meier U, Lemcke J. Co-morbidity as a predictor of outcome in patients with idiopathic normal-
pressure hydrocephalus. Acta Neurochir Suppl. 2010;106:127–30.
Meier U, Paris S, Gräwe A, Stockheim D, Hajdukova A, Mutze S. Is there a correlation between
operative results and change in ventricular volume after shunt placement? A study of 60 cases
of idiopathic normal-pressure hydrocephalus. Neuroradiology. 2003;45:377–80.
Mirzayan MJ, Luetjens G, Borremans JJ, Regel JP, Krauss JK. Extended long-term (> 5 years)
outcome of cerebrospinal fluid shunting in idiopathic normal pressure hydrocephalus.
Neurosurgery. 2010;67:295–301.
Missori P, Paolini S, Currà A. From congenital to idiopathic adult hydrocephalus: a historical
research. Brain. 2010;133:1836–49.
Missori P, Paolini S, Domenicucci M. The origin of the cannula for ventriculostomy in pediatric
hydrocephalus. J Neurosurg Pediatr. 2011;7:290–4.
Moiyadi AV, Praharaj SS, Pillai VS, Chandramouli BA. Hydrocephalus in Paget’s disease. Acta
Neurochir (Wien). 2006;148:1297–300.
Mollanji R, Bozanovic-Sosic R, Zakharov A, Makarian L, Johnston MG. Blocking cerebrospinal
fluid absorption through the cribriform plate increases resting intracranial pressure. Am
J Physiol Regul Integr Comp Physiol. 2002;282:R1593–9.
Murakami M, Morine M, Iwasa T, Takahashi Y, Miyamoto T, Hon PK, Nakagawa Y. Management
of maternal hydrocephalus requires replacement of ventriculoperitoneal shunt with ventricu-
loatrial shunt: a case report. Arch Gynecol Obstet. 2010;282:339–42.
Naruse H, Matsuoka Y. Post-operative improvement of 14 cases who were considered iNPH
despite Evans’ index of 0.3 or less. No Shinkei Geka. 2013;41:25–30.
Nulsen F, Spitz EB. Treatment of hydrocephalus by direct shunt from ventricle to jugular vein.
Surg Forum. 1951;2:399–409.
Olsen L, Frykberg T. Complications in the treatment of hydrocephalus in children. A comparison
of ventriculoatrial and ventriculoperitoneal shunts in a 20-year material. Acta Paediatr Scand.
1983;72:385–90.
Palm WM, Walchenbach R, Bruinsma B, Admiraal-Behloul F, Middelkoop HA, Launer LJ, van
der Grond J, van Buchem MA. Intracranial compartment volumes in normal pressure hydro-
cephalus: volumetric assessment versus outcome. AJNR Am J Neuroradiol. 2006;27:76–9.
Patwardhan RV, Nanda A. Implanted ventricular shunts in the United States: the billion-dollar-a-
year cost of hydrocephalus treatment. Neurosurgery. 2005;56:139–44.
Pinto FC, Saad F, Oliveira MF, Pereira RM, Miranda FL, Tornai JB, Lopes MI, Ribas ES, Valinetti
EA, Teixeira MJ. Role of endoscopic third ventriculostomy and ventriculoperitoneal shunt in
idiopathic normal pressure hydrocephalus: preliminary results of a randomized clinical trial.
Neurosurgery. 2013;72:845–54.
Poca MA, Mataró M, Del Mar Matarín M, Arikan F, Junqué C, Sahuquillo J. Is the placement of
shunts in patients with idiopathic normal-pressure hydrocephalus worth the risk? Results of a
study based on continuous monitoring of intracranial pressure. J Neurosurg. 2004;
100:855–66.
14 Normal Pressure Hydrocephalus 251
Taylor AE, Saint-Cyr JA, Lang AE. Frontal lobe dysfunction in Parkinson’s disease. The cortical
focus of neostriatal outflow. Brain. 1986;109:845–83.
Tisell M, Hoglund M, Wikkelso C. National and regional incidence of surgery for adult hydro-
cephalus in Sweden. Acta Neurol Scand. 2005;112:72–5.
Toma AK, Holl E, Kitchen ND, Watkins LD. Evans’ index revisited: the need for an alternative in
normal pressure hydrocephalus. Neurosurgery. 2011a;68:939–44.
Toma AK, Stapleton S, Papadopoulos MC, Kitchen ND, Watkins LD. Natural history of idiopathic
normal-pressure hydrocephalus. Neurosurg Rev. 2011b;34:433–9.
Toma AK, Papadopoulos MC, Stapleton S, Kitchen ND, Watkins LD. Conservative versus surgical
management of idiopathic normal pressure hydrocephalus: a prospective double-blind random-
ized controlled trial: study protocol. Acta Neurochir Suppl. 2012;113:21–3.
Trenkwalder C, Schwarz J, Gebhard J, Ruland D, Trenkwalder P, Hense HW, Oertel WH. Starnberg
trial on epidemiology of Parkinsonism and hypertension in the elderly. Prevalence of
Parkinson’s disease and related disorders assessed by a door-to-door survey of inhabitants older
than 65 years. Arch Neurol. 1995;52:1017–22.
Vanneste J, Augustijn P, Tan WF, Dirven C. Shunting normal pressure hydrocephalus: the predic-
tive value of combined clinical and CT data. J Neurol Neurosurg Psychiatry. 1993;56:251–6.
Vernet O, Campiche R, de Tribolet N. Long-term results after ventriculoatrial shunting in children.
Childs Nerv Syst. 1993;9:253–5.
Virhammar J, Cesarini KG, Laurell K. The CSF tap test in normal pressure hydrocephalus: evalu-
ation time, reliability and the influence of pain. Eur J Neurol. 2012;19:271–6.
von Ziemssen HW. Paracentesis of the spinal dura. Epitome of current medical literature. Br Med
J. 1893;1:81.
Vourc’h G. Continuous cerebrospinal fluid drainage by indwelling spinal catheter. Br J Anaesth.
1963;35:118–20.
Vourc’h G, Rougerie J. Continuous drainage of the cerebrospinal fluid. New method by the instal-
lation of a permanent spinal catheter. Its value in neurosurgery. Presse Med. 1960;68:1491–2.
Wang VY, Barbaro NM, Lawton MT, Pitts L, Kunwar S, Parsa AT, Gupta N, McDermott MW.
Complications of lumboperitoneal shunts. Neurosurgery. 2007;60:1045–8.
Westhout FD, Linskey ME. Obstructive hydrocephalus and progressive psychosis: rare
presentations of neurosarcoidosis. Surg Neurol. 2008;69:288–92.
Wikkelsø C, Andersson H, Blomstrand C, Lindqvist G. The clinical effect of lumbar puncture in
normal pressure hydrocephalus. J Neurol Neurosurg Psychiatry. 1982;45:64–9.
Wikkelsø C, Hellström P, Klinge PM, Tans JT. The European iNPH Multicentre Study on the pre-
dictive values of resistance to CSF outflow and the CSF Tap Test in patients with idiopathic
normal pressure hydrocephalus. J Neurol Neurosurg Psychiatry. 2013;84:562–8.
Wright EM. Transport processes in the formation of the cerebrospinal fluid. Rev Physiol Biochem
Pharmacol. 1978;83:3–34.
Wynter WE. Four cases of tubercular meningitis in which paracentesis was performed for the relief
of fluid pressure. Lancet. 1891;1:981–2.
Yadav YR, Parihar V, Sinha M. Lumbar peritoneal shunt. Neurol India. 2010;58:179–84.
Yusim A, Anbarasan D, Bernstein C, Boksay I, Dulchin M, Lindenmayer JP, Saavedra-Velez C,
Shapiro M, Sadock B. Normal pressure hydrocephalus presenting as Othello syndrome: case
presentation and review of the literature. Am J Psychiatry. 2008;165:1119–25.
Zemack G, Romner B. Seven years of clinical experience with the programmable Codman Hakim
valve: a retrospective study of 583 patients. J Neurosurg. 2000;92:941–8.
Zemack G, Romner B. Adjustable valves in normal-pressure hydrocephalus: a retrospective study
of 218 patients. Neurosurgery. 2008;62 Suppl 2:677–87.
Zhang J, Qu C, Wang Z, Wang C, Ding X, Pan S, Ji Y. Improved ventriculoatrial shunt for cerebro-
spinal fluid diversion after multiple ventriculoperitoneal shunt failures. Surg Neurol. 2009;72
Suppl 1:S29–33.
Chapter 15
Movement Disorders in Infectious Dementias
Introduction
The aim of this chapter is to describe the clinical features of movement disorders in
conditions where dementia is caused by infectious agents. From a historical point of
view, neurosyphilis is the prototypical infection that combines cognitive and move-
ment disorders. Also with a historical interest is postencephalitic parkinsonism, a con-
sequence of the encephalitis lethargica that plagued Europe in the first decades of the
twentieth century. In the late twentieth century, many patients of the epidemics of HIV
infection also displayed movement disorders in combination with dementia. The aim
of this chapter is to provide a description of the clinical features, epidemiology, and
guidelines of management of movement disorders present in the context of infectious
dementing illnesses. Of note, although outside of the scope of this chapter, there is
evidence suggesting that Parkinson’s disease (PD) may be caused by an infectious
proteinaceous particle that has a prion-like behavior (Olanow and Brundin 2013).
Neurosyphilis
Cognition
Epidemiology
In the past century, there has been a significant decline of the prevalence of neuro-
syphilis. However, in the last years there is a resurgence of this condition, with clini-
cians seeing more patients (Kent and Romanelli 2008). Although coinfection with
HIV is well recognized, this is not the main factor responsible for increase of the
incidence of neurosyphilis (Zetola and Klausner 2007). Movement disorders are
rarely seen in patients with the latter: in a recent series from China, they were seen
in no more than seven of 169 subjects (Tong et al. 2013).
Treatment
Postencephalitic Parkinsonism
Cognition
Despite intensive effort, the etiology of the pandemic encephalitis lethargica that
swept Europe in the beginning of the twentieth century was never determined. The
possibility of influenza virus was ruled out, and there is a recent suggestion that it
was caused by an enterovirus (Dourmashkin et al. 2012). There are occasional reports
of patients who develop parkinsonism following other encephalitic illnesses: western
equine encephalitis, Coxsackie B, measles, Epstein-Barr virus (Hsieh et al. 2002;
Dimova et al. 2006; Roselli et al. 2006), chicken pox encephalitis, and Japanese
encephalitis. The importance of the latter as cause of parkinsonism is shown by a
256 F. Cardoso and P. Caramelli
Epidemiology
Treatment
HIV Infection
Cognition
Cognitive impairment associated with HIV infection can be classified in three dif-
ferent categories, namely, asymptomatic neurocognitive impairment, mild neuro-
cognitive disorder, and HIV-associated dementia (Antinori et al. 2007). With the
advent of highly active antiretroviral therapy almost two decades ago, a dramatic
decline in the number of cases of moderate to severe dementia occurred. However,
mild forms of cognitive impairment are still very frequent and cause significant
morbidity for the patients (Kranick and Nath 2012).
Asymptomatic neurocognitive impairment is characterized by deficits on objec-
tive neuropsychological testing in two or more cognitive domains in patients with
no cognitive complaints. Subsequent symptomatic decline is observed in many
cases, although it is not predictable. Mild neurocognitive disorder is diagnosed in
patients presenting mild functional decline, which is not explained by another con-
dition, and performing at least one standard deviation below normative values on
neuropsychological testing in two or more cognitive domains. HIV-associated
dementia features cognitive deficits in at least two cognitive domains, falling below
two standard deviations on neuropsychological tests and also by significant func-
tional impairment (Kranick and Nath 2012).
The cognitive profile found in HIV infection includes deficits in information
processing, working memory, prospective memory, decision making, and verbal
fluency. This clinical picture resembles to the one found in subcortical dementias,
such as Parkinson disease and Huntington disease, and reflects damage to frontos-
triatal structures (Ances and Ellis 2007).
clinical practice, tremor is the most common movement disorder identified in HIV+
patients (Cardoso 2002b). However, hemichorea-hemiballismus is the most fre-
quently reported dyskinesia in HIV+ subjects (Piccolo et al. 1999; Cardoso 2002a).
Characteristically, this hyperkinesia is an acute complication of patients with estab-
lished AIDS, but in a few instances, hemichorea-hemiballismus may be the present-
ing symptom of HIV infection (Pardo et al. 1998; Cardoso 2002b). Chorea as direct
manifestation of HIV encephalopathy has been reported by some (Passarin et al.
2005; Sevigny et al. 2005). A French group described remission of chorea related to
HIV encephalopathy after introduction of highly active antiretroviral therapy
(Trocello et al. 2006).
Tremor is the second most commonly reported movement disorder in AIDS
patients. As it is often caused by Toxoplasma abscesses located in the mesencepha-
lon or thalamus, patients present with tremor of subacute onset with rest, postural,
and kinetic components, known as Holmes tremor (Koppel and Daras 1990; Micheli
et al. 1997). “Wing beating” tremor, related to middle cerebellar peduncle lesions,
mild postural tremor associated with HIV encephalopathy, and isolated rest tremor
induced by dopamine receptor blockers or trimethoprim-sulfamethoxazole have
also been reported (Swenson et al. 1989; Singer et al. 1990; Manji et al. 1995;
Cardoso 2002a). There are reports on the association of tremor with progressive
multifocal leukoencephalopathy, a common opportunistic infection in HIV+
patients (Rieder and Ziomkowski 2005; Sporer et al. 2005).
Less often, HIV+ patients may develop other hyperkinesias, such as generalized
or focal dystonia, Meige syndrome, myoclonus, painful legs and moving toes syn-
drome, akathisia, tics, stiff person syndrome, oculomasticatory myorhythmia asso-
ciated with Whipple disease, and opsoclonus-myoclonus (Jankovic 1986; Nath
et al. 1987; Tolge and Factor 1991; McDaniel and Summerville 1994; Lannuzel
et al. 2002; Canafoglia et al. 2003; Factor et al. 2003; Shah and Chudgar 2005;
Wicki et al. 2008; Wiersinga et al. 2012). Recently, there is a report of one patient
with acute onset of myoclonus and other neurologic clinical features who was found
to have West Nile virus infection in association with HIV (Josekutty et al. 2013).
There is also one study describing the occurrence of paroxysmal hyperkinesia in six
patients with advanced AIDS without underlying opportunistic infections (Mirsattari
et al. 1999).
Parkinsonism as part of HIV encephalopathy and, less frequently, resulting from
toxoplasmosis abscesses, parenchymatous tuberculosis granuloma, cryptococcal
abscesses, and other opportunistic infections has also been reported in patients with
AIDS (Carrazana et al. 1989; de la Fuente Aguado et al. 1996; Mirsattari et al. 1998;
Bouffard et al. 2003). In one of these studies, the authors describe six patients with
parkinsonism and ten others with “parkinsonian features.” In one half of the sub-
jects, the akinetic rigid syndrome was related to exposure to neuroleptics, whereas
no cause was identified in the remaining ones. These patients, who accounted for
5.2 % of all subjects seen in a Neuro-AIDS clinic, did not present with rest tremor,
and the majority of them also had additional clinical features such as dementia,
seizures, vacuolar myelopathy, and peripheral neuropathy. The CD4 values were
consistently low (mean of 14 cells/mm3). There is also a description of
15 Movement Disorders in Infectious Dementias 259
Epidemiology
Treatment
not clearly established, initial evidence suggests that highly active antiretroviral
therapy has a positive impact on these problems (Maschke et al. 2000; Sacktor et al.
2000; Brew 2001; Hersh et al. 2001; Cardoso 2002b).
Prions
Cognition
Typically, the onset of sporadic CJD is in the sixth and seventh decades of life with
a clinical picture characterized by a rapidly progressive dementia and other behav-
ioral disorders. In most cases, death ensues in less than a year (Brown et al. 1994;
Paterson et al. 2012; Puoti et al. 2012). The age at onset of iatrogenic forms is more
variable, with the latency ranging from a few months to 10–15 years. Shorter laten-
cies are usually observed in surgical cases, whereas patients exposed to contami-
nated hormones developed CJD after a much longer period (Cardoso 2002a).
Movement disorders were identified in 91 % of patients, of whom 81 % had
multifocal cortical myoclonus, of a series of 300 subjects with pathologically veri-
fied sporadic CJD (Brown et al. 1994). Although not always clearly described in the
literature, other movement disorders found in these patients are tremor, rigidity,
parkinsonism, dystonia, and chorea (Brown et al. 1994; Cardoso 2002a, b). Ataxia
is the clinical hallmark of kuru and Gerstmann-Sträussler-Scheinker disease
(Cardoso 2002a; Liberski et al. 2012). Variant Creutzfeldt-Jakob disease (vCJD) is
a novel human prion disease caused by the bovine spongiform encephalopathy
agent (Ironside 2012). In addition to myoclonus, these patients often have ataxia
and chorea (Will et al. 1996; Bowen et al. 2000; Ironside 2012).
15 Movement Disorders in Infectious Dementias 261
Epidemiology
Sporadic prion diseases are rare with the incidence of CJD estimated to be one case
per million population per year (Brown et al. 1987; Puoti et al. 2012). Infectious
prion diseases are even rarer. In the past kuru was common among subjects of the
Fore tribe of the highlands of New Guinea, but it has been eliminated with the disap-
pearance of individuals at genetic risk (those homozygous for the 129 Met allele of
the PRNP gene encoding for prion protein) and cannibalism (Gajdusek 1977;
Liberski et al. 2012).
There are less than 100 published cases of iatrogenic CJD related to corneal and
dura mater transplants, inadequately sterilized neurosurgical instruments, and con-
taminated growth hormone or gonadotrophins prepared from cadaveric pituitary
tissue (Brown et al. 1992).
Most cases of vCJD have occurred in the United Kingdom, with smaller numbers
in 11 other countries (Ironside 2012). There are also recent reports describing indi-
viduals who acquired vCJD by transfusion of blood-derived products of asymptom-
atic patients (Gregori et al. 2011; Ironside 2012; Millar and Makris 2012).
Treatment
Movement disorders are a relatively minor finding in the clinical picture of patients
with prion diseases. Nevertheless, if they cause disability, symptomatic treatment
for movement disorders can be used following guidelines similar to those used for
patients without prion diseases. Unfortunately, the prognosis of these conditions is
ominous not existing any measure effective in halting their relentless and fatal
progression.
Neurocysticercosis
Cognition
being identified in 12.5 % of all cases (Rodrigues et al. 2012). Importantly, cogni-
tive impairment or dementia could not be explained by the use of antiepileptic drugs
use, seizures, or depression (Ciampi de Andrade et al. 2010).
Hence, NC leads to a spectrum of cognitive changes from mild impairment in a
single domain to multiple cognitive deficits without functional decline and, some-
times, to dementia. These features are more prominent during the active phase of the
disease, with attenuation in the calcified stage.
Epidemiology
NC remains as a major public health problem in Latin American and India (Del
Brutto 2013). It is, however, becoming increasingly common in regions with high
rates of immigration from endemic areas, such as the South as well as the East Coast
of the United States (Scharf 1988; Croker et al. 2012).
Treatment
Patients with NC and parkinsonism may improve with L-dopa therapy, but the
majority of reported cases just respond to treatment of the intraparenchymal lesions
or the causative underlying hydrocephalus (Cardoso 2002a; Sá et al. 2005;
Prashantha et al. 2008). Subjects who present with hemichorea-hemiballismus are
usually responsive to neuroleptics or tetrabenazine.
15 Movement Disorders in Infectious Dementias 263
Sydenham’s Chorea
Cognition
Cognitive functioning has been object of only few studies in Sydenham’s chorea (SC).
Executive dysfunction has been reported in adult patients even when chorea was in
remission (Beato et al. 2010). Simple and brief cognitive tests, sensitive to evaluation
of prefrontal functions, such as phonemic verbal fluency, are able to detect significant
changes; thus, they may be used in clinical practice (Cunningham et al. 2006).
in a few patients with SC result from choreic contractions of the upper respiratory
tract muscles rather than true tics (Teixeira et al. 2009).
Dysarthria is common, and patients with more severe forms of this condition
may present a remarkably decreased verbal output. There is evidence that many
patients with active chorea have hypometric saccades, and a few also show oculo-
gyric crisis. In the older literature, there are also references to papilledema, central
retinal artery occlusion, and seizures in some patients with SC. Migraine is more
common in children with SC than in controls. In a cohort of 55 patients with this
movement disorder, 21.8 % were found to have migraine, whereas this type of head-
ache was seen in no more than 8 % of 110 matched controls (Teixeira et al. 2005a).
Attention has been drawn to behavioral abnormalities associated with this con-
dition. We investigated the behavior of 56 patients with SC, 50 subjects with
rheumatic fever without chorea, and 50 healthy matched controls. Obsessive-
compulsive disorder was diagnosed in 23 % of the patients with chorea, whereas
just 6 and 4 % of the rheumatic fever group and healthy controls met criteria for
this condition (Maia et al. 2005). Other studies have confirmed that obsessions
and compulsions are commonly seen in patients with SC (Asbahr et al. 1998;
Mercadante et al. 2000; Hounie et al. 2004). We saw one patient with paranoid
psychosis with onset in parallel with SC and also found that another patient with
SC developed trichotillomania (Kummer et al. 2007; Teixeira et al. 2007). These
findings and the observation that hyperactivity, learning disorders, and other
behavioral problems are common in patients with rheumatic fever and chorea
contributed to establish the notion that SC is a model for childhood autoimmune
neuropsychiatric disorders (Swedo 1994).
It must be kept in mind that SC is a major manifestation of rheumatic fever,
although in approximately 20 % of patients, chorea is the sole finding.
Nevertheless, up to 80 % of patients display cardiac involvement in SC, whereas
the association with arthritis is less common, seen in 30 % of subjects (Cardoso
et al. 1997). The current diagnostic criteria of SC are a modification of the Jones
criteria: chorea with acute or subacute onset and lack of clinical and laboratory
evidence of alternative cause are mandatory findings, and the diagnosis is further
supported by the presence of additional major or minor manifestations of rheu-
matic fever (Special Writing Group of the Committee of Rheumatic Fever 1992;
Cardoso et al. 1997, 1999).
Epidemiology
SC is the most common cause of acute chorea in children; however, its prevalence
has decreased in conjunction with the reduction of rheumatic fever in North
America and Western Europe. For instance, in Fairfax County, Virginia, the
annual age-adjusted incidence rate of initial attacks of rheumatic fever per 100,000
children declined from 3.0 in 1970 to 0.5 in 1980 (Schwartz et al. 1983).
Furthermore, Nausieda and colleagues demonstrated that SC accounted for 0.9 %
15 Movement Disorders in Infectious Dementias 265
Treatment
Conclusions
In this chapter we reviewed a number of infectious diseases that course with a com-
bination of movement disorders and cognitive decline. Historically, postencephalitic
parkinsonism and neurosyphilis were the first infectious conditions combining these
features to be studied. The former has vanished, but syphilis remains an important
cause of dementia although movement disorders are rare. Prion diseases are uncom-
mon and their infectious forms are even rarer. Nevertheless, they should always be
ruled out in patients with rapidly progressive dementia and myoclonus. The majority
of HIV + patients develop neurologic complications despite the advent of highly
active antiretroviral therapy. A substantial proportion of these patients display the
combination of movement disorder, particularly parkinsonism, and cognitive impair-
ment. Neurocysticercosis, the most common parasitic disease worldwide, is increas-
ingly recognized as a cause of cognitive disorder although movement disorders are
exceedingly rare in these subjects. Finally, although the incidence of Sydenham’s
chorea is in decline, it remains as the most common cause of acute chorea in children
and not infrequently it is associated with a dysexecutive syndrome.
References
Ances BM, Ellis RJ. Dementia and neurocognitive disorders due to HIV-1 infection. Semin Neurol.
2007;27:86–92.
Antinori A, Arendt G, Becker JT, et al. Updated research nosology for HIV-associated neurocogni-
tive disorders. Neurology. 2007;69:1789–99.
Asbahr FR, Negrao AB, Gentil V, et al. Obsessive-compulsive and related symptoms in children
and adolescents with rheumatic fever with and without chorea: a prospective 6-month study.
Am J Psychiatry. 1998;155:1122–4.
Ayoub EM. Resurgence of rheumatic fever in the United States. The changing picture of a prevent-
able illness. Postgrad Med. 1992;92:133–6.
Baird RA, Wiebe S, Zunt JR, Halperin JJ, Gronseth G, Roos KL. Evidence-based guideline: treat-
ment of parenchymal neurocysticercosis: report of the Guideline Development Subcommittee
of the American Academy of Neurology. Neurology. 2013;80(15):1424–9.
Barash J, Margalith D, Matitiau A. Corticosteroid treatment in patients with Sydenham’s chorea.
Pediatr Neurol. 2005;32:205–7.
Beato R, Maia DP, Teixeira Jr AL, Cardoso F. Executive functioning in adult patients with
Sydenham’s chorea. Mov Disord. 2010;25:853–7.
Benito-León J, Alvarez-Linera J, Louis ED. Neurosyphilis masquerading as corticobasal degen-
eration. Mov Disord. 2004;19(11):1367–70.
Bianchin MM, Velasco TR, Santos AC, Sakamoto AC. On the relationship between neurocysticer-
cosis and mesial temporal lobe epilepsy associated with hippocampal sclerosis: coincidence or
a pathogenic relationship? Pathog Glob Health. 2012;106(5):280–5.
Bouffard JP, Mena H, Ripple M, Troncoso J. Mesencephalic cryptococcal abscesses presenting
with parkinsonism as an initial manifestation of AIDS. Mov Disord. 2003;18:1354–7.
Bowen J, Mitchell T, Pearce R, Quinn N. Chorea in new variant Creutzfeldt-Jacob disease. Mov
Disord. 2000;15(6):1284–5.
Bremell D, Säll C, Gisslén M, Hagberg L. Lyme neuroborreliosis in HIV-1 positive men success-
fully treated with oral doxycycline: a case series and literature review. J Med Case Reports.
2011;5:465.
15 Movement Disorders in Infectious Dementias 267
Brew BJ. HIV neurology. New York: Oxford University Press; 2001.
Brown P, Cathala F, Raubertas RF, Gajdusek DC, Castaigne P. The epidemiology of Creutzfeldt-
Jakob disease: conclusion of a 15-year investigation in France and review of the world litera-
ture. Neurology. 1987;37(6):895–904.
Brown P, Gibbs Jr CJ, Rodgers-Johnson P, Asher DM, Sulima MP, Bacote A, Goldfarb LG,
Gajdusek DC. Human spongiform encephalopathy: the National Institutes of Health series of
300 cases of experimentally transmitted disease. Ann Neurol. 1994;35(5):513–29.
Brown P, Preece MA, Will RG. “Friendly fire” in medicine: hormones,homografts, and Creutzfeldt-
Jakob disease. Lancet. 1992;340(8810):24–7.
Canafoglia L, Panzica F, Franceschetti S, et al. Rhythmic cortical myoclonus in a case of HIV-
related encephalopathy. Mov Disord. 2003;18:1533–8.
Caramelli P, Teixeira AL, Buchpiguel CA, Lee HW, Livramento JA, Fernandez LL, Anghinah R. Diagnosis
of Alzheimer’s disease in Brazil: supplementary exams. Dement Neuropsychol. 2011;5:1–10.
Carapetis JR, Wolff DR, Currie BJ. Acute rheumatic fever and rheumatic heart disease in the top
end of Australia’s Northern Territory. Med J Aust. 1996;164:146–9.
Cardoso F. Infectious and transmissible movement disorders. In: Jankovic J, Tolosa E, editors.
Parkinson’s disease and movement disorders. 4th ed. Baltimore: Lippincott Williams &
Wilkins; 2002a. p. 945–66.
Cardoso F. HIV-related movement disorders: epidemiology, pathogenesis and management. CNS
Drugs. 2002b;16(10):663–8.
Cardoso F. Sydenham’s chorea. Curr Treat Options Neurol. 2008;10(3):230–5.
Cardoso F, Silva CE, Mota CC. Sydenham’s chorea in 50 consecutive patients with rheumatic
fever. Mov Disord. 1997;12:701–3.
Cardoso F, Vargas AP, Oliveira LD, Guerra AA, Amaral SV. Persistent Sydenham’s chorea. Mov
Disord. 1999;14:805–7.
Cardoso F, Maia D, Cunningham MC, Valenca G. Treatment of Sydenham chorea with corticoste-
roids. Mov Disord. 2003;18(11):1374–7.
Carr J. Neurosyphilis. Pract Neurol. 2003;3:328–41.
Carrazana E, Rossitch E, Samuels MA. Parkinsonian symptoms in a patient with AIDS and cere-
bral toxoplasmosis. J Neurol Neurosurg Psychiatry. 1989;52:1445–6.
Chauhan VV, Pickens 4th AT. Opisthotonus in neurosyphilis: a case report. J Emerg Med. 2011;41:
613–5.
Ciampi de Andrade D, Rodrigues CL, Abraham R, et al. Cognitive impairment and dementia in
neurocysticercosis: a cross-sectional controlled study. Neurology. 2010;74:1288–95.
Clay PG, Adams MM. Pseudo-Parkinson disease secondary to ritonavir-buspirone interaction.
Ann Pharmacother. 2003;37:202–5.
Cosentino C, Velez M, Torres L, Garcia HH, Cysticercosis Working Group in Perú. Cysticercosis
lesions in basal ganglia are common but clinically silent. Clin Neurol Neurosurg. 2002;104(1):
57–60.
Cosentino C, Vélez M, Torres L, Garcia HH. Neurocysticercosis-induced hemichorea. Mov
Disord. 2006;21(2):286–7.
Croker C, Redelings M, Reporter R, Sorvillo F, Mascola L, Wilkins P. The impact of neurocysti-
cercosis in California: a review of hospitalized cases. PLoS Negl Trop Dis. 2012;6(1):e1480.
Cunningham MC, Maia DP, Teixeira Jr AL, Cardoso F. Sydenham’s chorea is associated with
decreased verbal fluency. Parkinsonism Relat Disord. 2006;12:165–7.
Davis LE, Kornfeld M. Neurocysticercosis: neurologic, pathogenic, diagnostic and therapeutic
aspects. Eur Neurol. 1991;31(4):229–40.
de la Fuente-Aguado J, Bordon J, Moreno JA, Sopena B, Rodriguez A, Martinez-Vasquez C.
Parkinsonism in an HIV-infected patient with hypodense cerebral lesion. Tuber Lung Dis.
1996;77(2):191–2.
Del Brutto OH. Neurocysticercosis: new thoughts on controversial issues. Curr Opin Neurol.
2013;26(3):289–94.
Del Brutto OH, Wadia NH, Dumas M, Cruz M, Tsang VC, Schantz PM. Proposal of diagnostic
criteria for human cysticercosis and neurocysticercosis. J Neurol Sci. 1996;142(1–2):1–6.
268 F. Cardoso and P. Caramelli
McDaniel JS, Summerville MB. Tic disorder associated with encephalopathy in advanced HIV
disease. Gen Hosp Psychiatry. 1994;16:298–300.
Mercadante MT, Busatto GF, Lombroso PJ, et al. The psychiatric symptoms of rheumatic fever.
Am J Psychiatry. 2000;157:2036–8.
Micheli F, Granana N, Scorticati MC, et al. Unilateral postural and action tremor resulting from
thalamic toxoplasmosis in a patient with acquired immunodeficiency syndrome. Mov Disord.
1997;12:1096–8.
Millar CM, Makris M. Dealing with the uncertain risk of variant Creutzfeldt-Jakob disease trans-
mission by coagulation replacement products. Br J Haematol. 2012;158(4):442–52.
Mirsattari SM, Berry ME, Holden JK, Ni W, Nath A, Power C. Paroxysmal dyskinesias in patients
with HIV infection. Neurology. 1999;52:109–14.
Mirsattari SM, Power C, Nath A. Parkinsonism with HIV infection. Mov Disord. 1998;13:684–9.
Nath A, Jankovic J, Pettigrew LC. Movement disorders and AIDS. Neurology. 1987;37:37–41.
Nausieda PA, Grossman BJ, Koller WC, et al. Sydenham chorea: an update. Neurology. 1980;30:
331–4.
Nitrini R. The history of tabes dorsalis and the impact of observational studies in neurology. Arch
Neurol. 2000;57:605–6.
Nitrini R. Clinical and therapeutic aspects of dementia in syphilis and Lyme disease. Handb Clin
Neurol. 2008;89:819–23.
Nitrini R, Teixeira da Silva LS, Rosemberg S, et al. Prion disease resembling frontotemporal
dementia and parkinsonism linked to chromosome 17. Arq Neuropsiquiatr. 2001;59:161–4.
Nitrini R, Paiva ARB, Takada LT, Brucki SMD. Did you rule out neurosyphilis? Dement
Neuropsychol. 2010;4:338–45.
Olanow CW, Brundin P. Parkinson’s disease and alpha synuclein: is Parkinson’s disease a prion-
like disorder? Mov Disord. 2013;28(1):31–40.
Ozben S, Erol C, Ozer F, Tiras R. Chorea as the presenting feature of neurosyphilis. Neurol India.
2009;57(3):347–9.
Pardo J, Marcos A, Bhathal H, Castro M, Varea de Seijas E. Chorea as a form of presentation of
human immunodeficiency virus-associated dementia complex. Neurology. 1998;50:568–9.
Passarin MG, Alessandrini F, Nicolini GG, et al. Reversible choreoathetosis as the early onset of
HIV-encephalopathy. Neurol Sci. 2005;26:55–6.
Paterson RW, Torres-Chae CC, Kuo AL, Ando T, Nguyen EA, Wong K, Dearmond SJ, Haman A,
Garcia P, Johnson DY, Miller BL, Geschwind MD. Differential diagnosis of Jakob-Creutzfeldt
disease. Arch Neurol. 2012;24:1–5.
Paz JA, Silva CA, Marques-Dias MJ. Randomized double-blind study with prednisone in
Sydenham’s chorea. Pediatr Neurol. 2006;34:264–9.
Piccolo I, Causarano R, Sterzi R, et al. Chorea in patients with AIDS. Acta Neurol Scand. 1999;100:
332–6.
Pradhan S, Pandey N, Shashank S, Gupta RK, Mathur A. Parkinsonism due to predominant
involvement of substantia nigra in Japanese encephalitis. Neurology. 1999;53:1781–6.
Prashantha DK, Netravathi M, Ravishankar S, Panda S, Pal PK. Reversible parkinsonism follow-
ing ventriculoperitoneal shunt in a patient with obstructive hydrocephalus secondary to intra-
ventricular neurocysticercosis. Clin Neurol Neurosurg. 2008;110(7):718–21.
Puoti G, Bizzi A, Forloni G, Safar JG, Tagliavini F, Gambetti P. Sporadic human prion diseases:
molecular insights and diagnosis. Lancet Neurol. 2012;11(7):618–28.
Rail D, Scholtz C, Swash M. Post-encephalitic parkinsonism: current experience. J Neurol
Neurosurg Psychiatry. 1981;44:670–6.
Rieder CR, Ziomkowski SC. Head tremor and progressive multifocal leukoencephalopathy in
AIDS patients: report of two cases. Arq Neuropsiquiatr. 2005;63:150–3.
Rodrigues CL, de Andrade DC, Livramento JA, et al. Spectrum of cognitive impairment in neuro-
cysticercosis: differences according to disease phase. Neurology. 2012;78:861–6.
Roselli F, Russo I, Fraddosio A, et al. Reversible Parkinsonian syndrome associated with anti-
neuronal antibodies in acute EBV encephalitis: a case report. Parkinsonism Relat Disord. 2006;
12:257–60.
270 F. Cardoso and P. Caramelli
Sá DS, Teive HA, Troiano AR, Werneck LC. Parkinsonism associated with neurocysticercosis.
Parkinsonism Relat Disord. 2005;11(1):69–72.
Sacktor NC, Skolasky RL, Lyles RH, Esposito D, Selnes OA, McArthur JC. Improvement in HIV-
associated motor slowing after antiretroviral therapy including protease inhibitors. J Neurovirol.
2000;6(1):84–8.
Scharf D. Neurocysticercosis. Two hundred thirty-eight cases from a California hospital. Arch
Neurol. 1988;45(7):777–80.
Schwartz RH, Hepner SI, Ziai M. Incidence of acute rheumatic fever. A suburban community
hospital experience during the 1970s. Clin Pediatr. 1983;22:798–801.
Sevigny JJ, Chin SS, Milewski Y, et al. HIV encephalitis simulating Huntington’s disease. Mov
Disord. 2005;20:610–3.
Shah I, Chudgar P. Progressive multifocal leukoencephalopathy (PML) presenting as intractable
dystonia in an HIV-infected child. J Trop Pediatr. 2005;51(6):380–2.
Singer C, Sanchez-Ramos J, Rey G, et al. AIDS dementia complex presenting as an unusual uni-
lateral hand tremor, clumsiness and posturing. Ann Neurol. 1990;28:301.
Singh G, Burneo JG, Sander JW. From seizures to epilepsy and its substrates: neurocysticercosis.
Epilepsia. 2013;54(5):783–92.
Sorbi S, Hort J, Erkinjuntti T, et al. EFNS-ENS Guidelines on the diagnosis and management of
disorders associated with dementia. Eur J Neurol. 2012;19:1159–79.
Special Writing Group of the Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of
the Council on Cardiovascular Disease in the Young of the American Heart Association. Guidelines
for the diagnosis of rheumatic fever. Jones Criteria, 1992 update. JAMA. 1992;268(15):2069–73.
Spitz M, Maia FM, Gomes HR, Scaff M, Barbosa ER. Parkinsonism secondary to neurosyphilis.
Mov Disord. 2008;23(13):1948–9.
Sporer B, Seelos K, Asmus F, et al. Posterior fossa tremor induced by HIV-associated progressive
multifocal leukoencephalopathy. Eur Neurol. 2005;53:96–7.
Swedo SE. Sydenham’s chorea. A model for childhood autoimmune neuropsychiatric disorders
[clinical conference]. JAMA. 1994;272:1788–91.
Swenson JR, Erman M, Labelle J, Dimsdale JE. Extrapyramidal reactions. Neuropsychiatric mim-
ics in patients with AIDS. Gen Hosp Psychiatry. 1989;11:248–53.
Takada LT, Caramelli P, Radanovic M, Anghinah R, Hartmann AP, Guariglia CC, Bahia VS,
Nitrini R. Prevalence of potentially reversible dementias in a dementia outpatient clinic of a
tertiary university-affiliated hospital in Brazil. Arq Neuropsiquiatr. 2003;61:925–9.
Takayanagui OM, Odashima NS, Bonato PS, Lima JE, Lanchote VL. Medical management of
neurocysticercosis. Expert Opin Pharmacother. 2011;12(18):2845–56.
Teixeira AL, Cardoso F, Maia DP, Cunningham MC. Sydenham’s chorea may be a risk factor for
drug induced parkinsonism. J Neurol Neurosurg Psychiatry. 2003;74(9):1350–1.
Teixeira Jr AL, Cardoso F, Maia DP, et al. Frequency and significance of vocalizations in
Sydenham’s chorea. Parkinsonism Relat Disord. 2009;15:62–3.
Teixeira Jr AL, Maia DP, Cardoso F. Psychosis following acute Sydenham’s chorea. Eur Child
Adolesc Psychiatry. 2007;16(1):67–9.
Teixeira Jr AL, Meira FC, Maia DP, Cunningham MC, Cardoso F. Migraine headache in patients
with Sydenham’s chorea. Cephalalgia. 2005a;25:542–4.
Teixeira Jr AL, Maia DP, Cardoso F. Treatment of acute Sydenham’s chorea with methyl-
prednisolone pulse-therapy. Parkinsonism Relat Disord. 2005b;11(5):327–30.
Tolge CF, Factor SA. Focal dystonia secondary to cerebral toxoplasmosis in a patient with acquired
immune deficiency syndrome. Mov Disord. 1991;6:69–72.
Tong ML, Lin LR, Zhang HL, Huang SJ, Liu GL, Zheng WH, Su YH, Wu JY, Dong J, Liu LL,
Yang TC. Spectrum and characterization of movement disorders secondary to neurosyphilis.
Parkinsonism Relat Disord. 2013;19(4):441–5.
Toovey S, Jick SS, Meier CR. Parkinson’s disease or Parkinson symptoms following seasonal
influenza. Influenza Other Respi Viruses. 2011;5:328–33.
Trocello JM, Blanchet A, Bourdain F, et al. Resolution of choreic movements associated with HIV
encephalitis with anti-retroviral therapy. Rev Neurol (Paris). 2006;162:89–91.
15 Movement Disorders in Infectious Dementias 271
von Economo C. Encephalitis lethargica: its sequelae and treatment (trans: Newman KO). New
York: Oxford University Press; 1931.
Wenning GK, Jellinger K, Litvan I. Supranuclear gaze palsy and eyelid apraxia in postencephalitic
parkinsonism. J Neural Transm. 1997;104(8–9):845–65.
Wicki J, Germanier Y, Sztajzel R, Burkhard PR. Brueghel syndrome as a new manifestation of
HIV encephalopathy. Eur Neurol. 2008;60:107–8.
Wiersinga WJ, Prins JM, van de Beek D. Therapy-resistant opsoclonus-myoclonus syndrome sec-
ondary to HIV-1 infection. Clin Infect Dis. 2012;54:447–8.
Will RG, Ironside JW, Zeidler M, Cousens SN, Estibeiro K, Alperovitch A, Poser S, Pocchiari M,
Hofman A, Smith PG. A new variant of Creutzfeldt-Jakob disease in the UK. Lancet.
1996;347(9006):921–5.
Zetola NM, Klausner JD. Syphilis and HIV infection: an update. Clin Infect Dis. 2007;44(9):
1222–8.
Zomorrodi A, Wald ER. Sydenham’s chorea in western Pennsylvania. Pediatrics. 2006;117:e675–9.
Index
CBD. See Corticobasal degeneration (CBD) Dementia associated with Parkinson’s disease,
CBS. See Corticobasal syndrome (CBS) 4, 46–50, 52, 55, 62–64, 68, 118, 119, 122,
Center for Epidemiologic Studies Depression 157, 160, 161, 163, 164, 166, 167,
Scale (CES-D), 76 180–188, 191
Cerebrospinal fluid (CSF), 130, 159, 166, 232, Dementia with Lewy bodies (DLB), 7, 8, 20,
233, 238, 240–245 26, 46–49, 62–64, 68, 69, 118, 119, 122,
Cerebrovascular disease (CVD), 27, 49–52, 133–135, 145, 148, 155–172, 181–184,
55, 200–205, 207, 208, 239 186–188, 192, 233
CES-D. See Center for Epidemiologic Studies Depression, 6, 23, 73–84, 132, 137, 148, 162,
Depression Scale (CES-D) 165, 171, 181–182, 184, 189, 190, 209,
ChEIs. See Cholinesterase 237, 238, 262
inhibitors (ChEIs) Diagnostic and Statistical Manual of Mental
Cholinergic dysfunction, 49, 50 Disorders, Fourth Edition (DSM-IV), 74,
Cholinesterase inhibitors (ChEIs), 55, 79, 122, 75, 81, 82
137, 171, 184, 209 DIMDs. See Drug-induced movement
Chorea, 3, 4, 88, 89, 91–93, 109, 254, 255, disorders (DIMDs)
258–260, 263–266 DIP. See Drug-induced parkinsonism (DIP)
Chromosome 17, 68, 145–147, 225 DIPD. See Drug-induced parkinsonism and
CJD. See Creutzfeldt–Jakob disease (CJD) dementia (DIPD)
Clinical Global Impression of Change (CGIC) DLBD, 8, 133–135, 137
scale, 83 Drug-induced movement disorders (DIMDs),
Cognitive fluctuations, 158 87–111, 124
Cognitive impairment, 17–38, 46–56, 62, 66, Drug-induced parkinsonism (DIP), 61, 67–69,
68, 90, 91, 123, 130, 159–161, 164, 166, 88, 91, 93, 94, 107, 109, 144
168, 171, 180, 183, 184, 188–190, 192, Drug-induced parkinsonism and dementia
204, 207, 237, 238, 241, 245, 254, 257, (DIPD), 63, 67
261, 262, 266 Dual energy x-ray absorptiometry (DXA), 56
Cognitive manifestations, 159, 161 DXA. See Dual energy x-ray absorptiometry
Columbia University Parkinson’s Disease (DXA)
Rating Scale, 124 Dystonia, 4, 5, 11, 62, 88–90, 92, 93,
C9ORF72, 142–144, 147–148, 150, 226 107–110, 124, 135–137, 146–149, 188,
Cornell Scale for Depression in Dementia 217, 219, 254, 258–260
(CSDD), 76
Corticobasal degeneration (CBD), 3, 5, 8, 9,
68, 187–192, 224, 225 E
Corticobasal syndrome (CBS), 7, 62, 63, 66, ECT. See Electroconvulsive therapy (ECT)
69, 136, 137, 142, 143, 145, 149, 188–191, EDS. See Excessive daytime sleepiness (EDS)
214–223, 254 Electroconvulsive therapy (ECT), 82, 92
Creutzfeldt–Jakob disease (CJD), 3–5, 11, Encephalitis subcorticalis chronica
133–135, 260 progressive, 201
CSDD. See Cornell Scale for Depression in ESRS. See Extrapyramidal Symptom Rating
Dementia (CSDD) Scale (ESRS)
CSF. See Cerebrospinal fluid (CSF) Excessive daytime sleepiness (EDS), 170, 172,
CVD. See Cerebrovascular disease (CVD) 184
Extrapyramidal Symptom Rating Scale
(ESRS), 124
D
Dementia, 1–12, 17–38, 45–56, 61–69, 74,
76–79, 82, 83, 99, 118–120, 122–125, F
129–131, 133, 134, 137, 141–150, Falls, 3, 18–26, 29–33, 38, 45–56, 76,
157–159, 161, 163–165, 179–192, 147–148, 162, 187, 188, 207, 214, 218,
199–209, 224, 233–235, 237, 244, 236, 239
253–266 Filament inclusion disorders, 9–10
Index 275
Frontotemporal dementia (FTD), 3, 4, 6, 7, 9, Lewy body disease (LBD), 48, 49, 135, 157,
10, 12, 68, 131, 141–150, 157, 182, 188, 158, 164, 224
224, 233, 260 Lewy body inclusions, 157
Frontotemporal lobe dementia (FTLD), 62, 63, Logopenic variant of PPA (lvPPA), 217–219,
67–69, 142, 143, 147 221, 222, 224–226
FTD. See Frontotemporal dementia (FTD) Lower-body parkinsonism, 236
FTDP-17, 8, 9, 145–147, 149, 224, 226 lvPPA. See Logopenic variant
FTLD. See Frontotemporal lobe dementia of PPA (lvPPA)
(FTLD)
Functional Assessment Staging (FAST)
scale, 125 M
FUSpathies, 9 Magnetic gait, 66
MAP. See Microtubule-associated
protein (MAP)
G MCI. See Mild cognitive impairment (MCI)
Gait, 17–38, 46–56, 62, 64, 66, 92, 118, 119, MDS-UPDRS. See Movement Disorder
123–125, 132, 133, 144, 188, 202, Society-sponsored version of the UPDRS
205–207, 214, 219, 236, 237, 239–242 (MDS-UPDRS)
Gait abnormalities, 27, 38, 53–54, 124 Methylphenidate (MPH), 38, 79
Gait disorders, 17–38, 48, 132 Microtubule-associated protein (MAP), 9
Gegenhalten, 81, 137, 217 Microtubule-associated protein tau
Genetics, 130, 147, 148, 150, 163, 183, (MAPT) gene, 9, 142, 145–148,
225–226 150, 183, 226
Mild cognitive impairment (MCI), 18, 21–27,
37, 46, 49, 54–56, 123, 159, 161, 180, 181,
H 184, 186, 187, 240
Hallucinations, 62–65, 68–69, 83, 91, 144, MND. See Motor neuron disease (MND)
147, 159, 161, 165, 171, 181, 184, Monoamine oxidase B (MAO-B)
189–190, 254, 256 inhibitors, 64
High-level gait disorders (HLGD), 50 Motor, 4–6, 18–21, 23, 27, 33, 38, 50, 52, 53,
HIV infection, 254, 257–260 55, 63–68, 73–74, 76, 77, 79–82, 89, 93,
HLGD. See High-level gait disorders (HLGD) 107, 108, 118–125, 131–134, 143, 147,
Huntington’s disease, 11, 88, 257 156, 159, 161, 162, 164, 165, 171, 180,
Hydrocephalus, 5, 61–63, 165–166, 232–235, 181, 183, 184, 189–192, 200, 201,
239, 243–244, 262 214–216, 218–220, 226, 234, 236,
242, 256, 257, 259–260, 263
Motor manifestations, 74, 159
I Motor neuron disease (MND), 5, 218, 225
ICARS. See International Cerebellar Ataxia Movement disorders, 2–5, 7, 9–10, 49, 82–84,
Rating Scale (ICARS) 87–111, 124, 129–137, 141–150, 158,
Idiopathic, 7, 61, 62, 67, 68, 88, 90, 91, 218–219, 253–266
110, 157, 159, 163, 171, 200, 203, Movement Disorder Society-sponsored
233–235, 238 version of the UPDRS (MDS-UPDRS),
Infectious dementias, 253–266 121, 122
International Cerebellar Ataxia Rating Scale MPH. See Methylphenidate (MPH)
(ICARS), 122 MSA. See Multiple system
atrophy (MSA)
Multiple system atrophy (MSA), 3, 5, 8, 10,
L 62, 91, 122–125, 136, 163, 167, 187–188,
LBs. See Lewy bodies (LBs) 191–192, 206, 224
Lewy bodies (LBs), 3, 10–11, 20, 46, 48, 134, Myoclonus, 62, 89, 91–93, 108, 134, 136,
145, 157, 162–164, 166–167, 180, 182, 146–149, 159, 188, 217, 219, 258,
183, 224 260, 266
276 Index
UPDRS. See Unified Parkinson’s disease vCJD. See Variant Creutzfeldt–Jakob disease
rating scale (UPDRS) (vCJD)
Urinary incontinence, 66, 162, 172, 207, 236, Ventricle, 201–202, 232, 233, 243
240, 241 VH. See Visual hallucinations (VH)
Visual hallucinations (VH), 158, 161, 171,
181, 184, 256
V Voxel-based morphometry (VBM), 147, 219
VAD. See Vascular dementia (VAD) VP. See Vascular parkinsonism (VP)
Variant Creutzfeldt–Jakob disease (vCJD),
260, 261
Vascular dementia (VAD), 27, 28, 31, 32, 38, W
46, 67, 131, 137, 157, 199–209, 233 Webster scale, 124
Vascular parkinsonism (VP), 61, 63, 67, White matter hyperintensities (WMH),
199–209 19, 32, 33
VBM. See Voxel-based morphometry WMH. See White matter hyperintensities
(VBM) (WMH)