Anda di halaman 1dari 6

Dementia with Lewy Bodies

David Weisman, M.D.,1 and Ian McKeith, M.D.2

ABSTRACT

Synucleinopathies, with and without dementia, encompass a wide range of diseases


including Parkinson’s disease, multiple system atrophy, rapid eye movement (REM) sleep
behavior disorder, and dementia with Lewy bodies (DLB). DLB is a neurodegenerative
disorder resulting in slowly progressive and unrelenting dementia until death. Prevalence
studies suggest that it is the second most common dementing illness in the elderly. The
neuropathologic findings of DLB show a wide anatomic range. Lewy bodies and Lewy-
related pathology are found from the brain stem to the cortex and, in many cases, associated
with concurrent Alzheimer’s disease pathology. A recent international consortium on DLB
has resulted in revised criteria for the clinical and pathological diagnosis of DLB
incorporating new information about the core clinical features and improved methods for
their assessment. The presentation of DLB is typically one of cortical and subcortical
cognitive impairments, with worse visuospatial and executive dysfunction than Alzheimer’s
disease. There may be relative sparing of memory especially in the early stages. Core clinical
features of DLB include fluctuating attention, recurrent visual hallucinations, and parkin-
sonism. Suggestive features include REM sleep behavior disorder, severe neuroleptic
sensitivity, and low dopamine transporter uptake in the basal ganglia on functional neuro-
imaging. Additional supportive features that commonly occur in DLB, but with lower
specificity, include repeated falls and syncope, transient, unexplained loss of consciousness,
severe autonomic dysfunction, hallucinations in other modalities, systematized delusions,
depression, relative preservation of medial temporal lobe structures on structural neuro-
imaging, reduced occipital activity on functional neuroimaging, prominent slow wave
activity on electroencephalogram, and low uptake myocardial scintigraphy. Management
of DLB includes pharmacological and nonpharmacological interventions for its cognitive,
neuropsychiatric, motor, and sleep disturbances.

KEYWORDS: Dementia with Lewy bodies, Lewy body variant, dementia, a-synuclein

D ementia with Lewy bodies (DLB) is the sec- The neuropathologic findings of DLB show a
ond most common cause of dementia in the elderly. wide anatomic range. Lewy bodies and Lewy-related
Lewy bodies are neuronal inclusions classically associ- changes are found from the brain stem to the cortex and,
ated with idiopathic Parkinson’s disease. Over the past in many cases, associated with concurrent Alzheimer’s
two decades, research has suggested that Lewy bodies, disease pathology. Thus, DLB is probably best thought
found in up to 30% of autopsied brains, play a significant of as a spectrum, with a wide range of Lewy body
role in the spectrum of dementing disorders that have involvement in the brains of patients with dementia
come to be known as DLB.1 (Fig. 1). Clinicopathologic studies have identified three

1
Department of Neurosciences and the Alzheimer’s Disease Research search Center, 9500 Gilman Drive, La Jolla, CA 92093.
Center, University of California, San Diego; 2University of Newcastle Dementia; Guest Editor, Jody Corey-Bloom, M.D., Ph.D.
upon Tyne, Wolfson Research Centre, Newcastle General Hospital, Semin Neurol 2007;27:42–47. Copyright # 2007 by Thieme
Newcastle, United Kingdom. Medical Publishers, Inc., 333 Seventh Avenue, New York, NY
Address for correspondence and reprint requests: David Weisman, 10001, USA. Tel: +1(212) 584-4662.
M.D., University of California, San Diego Alzheimer’s Disease Re- DOI 10.1055/s-2006-956754. ISSN 0271-8235.
42
DEMENTIA WITH LEWY BODIES/WEISMAN, McKEITH 43

Figure 1 Relationship of DLB to Alzheimer’s disease and Parkinson’s disease. Diagram is not drawn to epidemiologic scale. AD,
Alzheimer’s disease; LBV, Lewy body variant of AD; DLBD, diffuse Lewy body disease; PDD, Parkinson’s disease with dementia; PD,
Parkinson’s disease; LB, Lewy bodies.

dementia types in which clinical phenomenology may be Parkinson’s disease,4 Spillantini et al showed that both
related to Lewy bodies. These are: Parkinson’s disease and cortical Lewy bodies contained
a-synuclein.5 Alpha-synuclein is a 140 amino acid
1. Parkinson’s disease with dementia: Patients with protein of unknown function. It is currently believed
Parkinson’s disease who become demented at least that abnormal protein processing gives rise to the cyto-
1 year after the onset of motor symptoms. plasmic collections of a-synuclein, which then coalesce
2. Diffuse Lewy body disease: Patients with dementia to form Lewy bodies.6 Using immunohistochemistry,
and diffuse cortical Lewy bodies pathology without antibodies to a-synuclein reliably identify Lewy body
other significant pathological abnormalities. pathology.
3. Lewy body variant of Alzheimer’s disease: Patients Thus, despite uncertainties in clinical phenotypes,
with clinical dementia whose brains display both pathogenesis, and pathophysiology, the spectrum of
cortical Lewy bodies and Alzheimer-type neuro- DLB and Parkinson’s disease is now recognized as
pathologic changes. This is the most common form
of Lewy body dementia, occurring in 70% of
patients with DLB.1

PATHOPHYSIOLOGY
The discovery of Lewy bodies within substantia nigra
neurons in patients with Parkinson’s disease was made by
Lewy in 1912.2 These Lewy bodies consist of cytoplas-
mic inclusions within monoaminergic and cholinergic
neurons of the substantia nigra, locus ceruleus, and basal
nucleus of Meynert. They have a dense eosinophilic core
surrounded by a lucent halo and are easily seen with
routine staining. In contrast to Parkinson’s disease, the
neocortical Lewy bodies seen in DLB are smaller, lack a
halo, and are difficult to see under routine staining
conditions (Fig. 2). Because of this, it has been only
20 years since Kosaka et al identified widely distributed
Lewy bodies in the cortex and subcortex of patients with
parkinsonism and dementia.3
On the heels of Polymeropoulos et al’s 1997 Figure 2 Neocortical Lewy body (at tip of arrow) abutting the
discovery that a mutation on the a-synuclein gene nucleus of a neuron (60  ). The stain is anti-a-synuclein with a
(SNCA) on chromosome 4 was associated with familial hematoxylin counterstain.
44 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 1 2007

Table 1 Probability That the Pathological Findings Are Associated with a DLB Clinical Syndrome
Alzheimer-Type Pathology
Lewy Body-Type Pathology Braak Stage 0–II Braak Stage III–IV Braak Stage V–VI

Brain stem-predominant Low Low Low


Limbic (transitional) High Intermediate Low
Diffuse neocortical High High Intermediate
12
Data modified from McKeith et al.

belonging to a class of disorders characterized by patho- pathological findings are associated with a DLB clinical
logic a-synuclein processing at a cellular level, termed syndrome.
synucleinopathies.7 Abnormal synuclein processing is Dementia eventually occurs in 30 to 50% of all
also implicated in multiple system atrophy, which shows Parkinson’s disease cases.13 Typically, the dementia
glial a-synuclein-positive cytoplasmic inclusions. occurs 10 years after motor onset. It is characterized by
Although 35 to 90% (depending on pathological impairment in executive functions, visuospatial skills,
definition) of DLB cases prove to have concurrent free-recall memory, and verbal fluency, consistent with
Alzheimer-type pathology,1 the molecular relationship a pattern of frontal/subcortical dementia. The neuro-
between Lewy bodies and Alzheimer changes is un- pathology of Parkinson’s disease with dementia is con-
known. The pathological Alzheimer’s disease changes troversial. Cortical Lewy bodies have been reported in up
that accompany DLB are plaque predominant (i.e., there to 90%, with many having Alzheimer-type pathological
is usually significantly less tangle pathology than that changes.14 There is also evidence that cortical Lewy body
seen in typical Alzheimer’s disease brains).8 involvement, not Alzheimer pathology, determines clin-
Whether Lewy bodies have direct or indirect ical dementia.15 For the purposes of this review, how-
effects on local neurons remains unclear. Brains of ever, we will focus our remarks on neocortical Lewy body
patients with Lewy bodies demonstrate severe depletion disease (i.e., Lewy body variant and diffuse Lewy body
of both cholinergic and dopaminergic markers.9 Cortical disease).
Lewy bodies, however, do not correlate well with the
clinical severity of dementia, particularly when there is
significant superimposed Alzheimer’s disease pathol- CLINICAL PRESENTATION
ogy.10,11 A recent international consortium on DLB Dementia with Lewy bodies usually presents in late
has proposed a new scheme for the pathological assess- adulthood, between the ages of 60 and 90, with no
ment of Lewy bodies and Lewy neurites using a-synu- significant gender or ethnic differences in prevalence.16
clein immunohistochemistry and semiquantitative The disease is a dementing disorder; thus, progressive
grading of lesion density, with the pattern of regional and disabling cognitive impairment (of enough severity
involvement being more important than total Lewy body to interfere with day-to-day function) is the central
count.12 The new scheme takes into account both Lewy- feature.17 Patients show a combination of cortical and
related and Alzheimer’s disease-type pathology to allo- subcortical cognitive impairments with significant atten-
cate a probability that these are associated with the tional deficits in addition to executive and visuospatial
clinical DLB syndrome (Table 1). It predicts, for exam- dysfunction (Table 2). Compared with Alzheimer’s dis-
ple, that patients with Lewy bodies found throughout ease, there may be relative sparing of memory especially
the neocortex (diffuse neocortical) who have minimal to in the early stages.18,19 However, because the cognitive
moderate Alzheimer’s disease-type pathology (Braak differences are quantitative and not qualitative, the
stage 0 to IV) will have a high probability that their distinction between cognitive profiles may be difficult.

Table 2 Case Presentation


A 70-year-old man noted that his coordination was ‘‘off’’ and that he was having problems balancing his checkbook for about a year.
On a recent trip to Italy he had became quite confused with difficulty driving. Increasingly, his sleep was disturbed by vivid,
and sometimes violent dreams in which his wife reported that he vocalized and thrashed around in the bed.
His examination showed full orientation and normal calculations. Language was normal. The patient remembered three of five items
on a short-term verbal memory task. He had difficulty drawing a clock and intersecting pentagons. There was mild rigidity but no
postural instability.
Comment: This case is suggestive of DLB because of the prominent visuospatial difficulties, parkinsonism (a core feature), and REM
sleep behavior disorder (a suggestive feature). This patient would meet revised consortium criteria for probable DLB.
DEMENTIA WITH LEWY BODIES/WEISMAN, McKEITH 45

The recent international consortium on DLB has difficult to ascertain, even by the most experienced
resulted in revised criteria for the clinical diagnosis of clinician, but can be very helpful in identifying DLB.
DLB incorporating new information about the core clini- When asked explicitly, caregivers may affirm daytime
cal features and improved methods for their assessment.12 drowsiness and sleep, periods of confusion, and episodes
of staring into space.21
Visual hallucinations in DLB are recurrent and
Core Features well formed. The hallucinations are a useful symptom
The core features of DLB include fluctuating cognition, when diagnosing DLB because they are generally
recurrent visual hallucinations, and spontaneous parkin- present early in the course of the illness and do not
sonism (Table 320). diminish in later periods. Functional brain imaging
Fluctuations in cognitive function due to waxing demonstrates altered patterns of activation and blood
and waning attention over minutes, hours, or days are flow in the visual cortex. Harding et al showed slightly
increased numbers of Lewy bodies in the anterior and
medial temporal lobe in those with hallucinations.22
Table 3 Consensus Criteria for DLB
Visual hallucinations may predict a good response to
1. Central feature (essential for a diagnosis of possible or cholinesterase inhibitors.23
probable DLB) Extrapyramidal signs, including bradykinesia, fa-
Dementia defined as progressive cognitive decline of cial masking, and rigidity, are the most frequent signs of
sufficient magnitude to interfere with normal social or parkinsonism in DLB patients. Resting tremor is dis-
occupational function; prominent or persistent memory tinctly uncommon. Parkinsonism is usually bilateral and
impairment may not necessarily occur in the early stages occurs with the onset of dementia, although the onset
but is usually evident with progression; deficits on tests and the severity of the parkinsonism are highly variable.
of attention, executive function, and visuospatial ability There is often more axial rigidity and facial masking in
may be especially prominent DLB than in idiopathic Parkinson’s disease. Although
2. Core features (two core features are sufficient for a diagnosis conventional teaching suggests that DLB may be poorly
of probable DLB, one for possible DLB) responsive to levodopa, small trials have shown that
Fluctuating cognition with pronounced variations in attention DLB patients tolerate this medication and, in some
and alertness cases, show a beneficial response.24
Recurrent visual hallucinations that are typically well formed
and detailed
Spontaneous features of parkinsonism Suggestive Features
3. Suggestive features (one or more of these in the presence of In addition to these core items, the recent DLB
one or more core features is sufficient for a diagnosis of consortium identified additional clinical features sug-
probable DLB; in the absence of any core features, one or gestive of DLB, including rapid eye movement
more suggestive features is sufficient for a diagnosis of (REM) sleep disorder, severe neuroleptic sensitivity,
possible DLB; probable DLB should not be diagnosed on and low dopamine transporter uptake in basal ganglia
the basis of suggestive features alone) demonstrated by single photon-emission computed
REM sleep behavior disorder tomography (SPECT) or positron-emission tomogra-
Severe neuroleptic sensitivity phy (PET) imaging.
Low dopamine transporter uptake in basal ganglia REM sleep behavior disorder consists of partic-
demonstrated by SPECT or PET imaging ularly vivid (and sometimes violent) dreams during
4. Supportive features (commonly present but proven to REM sleep without the normal muscle atonia that
have diagnostic specificity) prevents movements (other than eye movements) during
Repeated falls and syncope dreams. Patients literally act out their dreams, mani-
Transient, unexplained loss of consciousness fested by talking and moving, sometimes with goal-
Severe autonomic dysfunction oriented violence. Because the patients have little mem-
Hallucinations in other modalities ory of these dreams, the history is dependent on the
Systematized delusions patient’s bed partner. Interestingly, the sleep disorder
Depression may be present many years before the onset of demen-
Relative preservation of mesial temporal lobe structures on tia.12
computed tomography/magnetic resonance imaging Neuroleptic sensitivity to D2 receptor blockers
Reduced occipital activity on SPECT/PET occurs in about half of DLB patients and, when present,
Low uptake MIBG myocardial scintigraphy is considered suggestive of DLB. Because a neuroleptic
Prominent slow wave activity on EEG with temporal lobe ‘‘stress test’’ to aid diagnosis is dangerous in this pop-
transient sharp waves20 ulation and may prove irreversibly damaging, this is not
Data modified from McKeith et al.12 recommended.25
46 SEMINARS IN NEUROLOGY/VOLUME 27, NUMBER 1 2007

Functional imaging of the dopamine transporter several have examined Parkinson’s disease with demen-
(DAT) defines the integrity of the nigrostriatal dopa- tia). A 5-month study of rivastigmine in DLB showed
minergic system and, using specific ligands, provides a a significant benefit of treatment on behavioral symp-
marker for presynaptic neuronal degeneration. Low toms as assessed by the Neuropsychiatric Inventory and
striatal DAT activity occurs in DLB but not in Alz- on a cognitive speed score. It is not clear if there was an
heimer’s disease and, thus, may be helpful in distinguish- actual change in cognition.
ing between the two disorders.26 Presently AChEs are the mainstay of DLB treat-
ment. These medications, including donepezil (Aricept),
rivastigmine (Exelon), and galantamine (Razadyne ER
Supportive Features or Reminyl), block the breakdown of acetylcholine
Commonly present but not specific (‘‘supportive’’) within the synapse, thereby prolonging its effect on
features of DLB include repeated falls and syncope, postsynaptic receptors. The AChE inhibitors are usually
transient unexplained loss of consciousness, severe auto- well tolerated at their standard dosing. The primary side
nomic dysfunction, hallucinations in other modalities, effects associated with treatment are gastrointestinal
systematized delusions, depression, relative preservation (e.g., nausea, vomiting, diarrhea, anorexia, weight loss);
of medial temporal lobe structures on structural insomnia, vivid dreams, leg cramps, and urinary fre-
neuroimaging, reduced occipital activity on functional quency have also been reported.
neuroimaging, prominent slow wave activity on electro- It is common to find a patient on an AChE
encephalogram (EEG), and low uptake iodine-123 inhibitor while also maintained on a centrally acting
metaiodobenzylguanidine (MIBG) myocardial scintig- anticholinergic agent, such as diphenhydramine for sleep,
raphy. Scintigraphy with [I-123] MIBG, which enables other antihistamines for allergy, meclizine for dizziness or
quantification of postganglionic sympathetic cardiac in- vertigo, oxybutynin for bladder control, or a highly
nervation, is reduced in DLB and may have high anticholinergic tricyclic (such as amitriptyline) for de-
sensitivity and specificity in differentiating it from Alz- pression or chronic pain. A careful review of medications,
heimer’s disease.27 According to the revised DLB con- including over-the-counter agents, is essential to avoid
sortium criteria, two core features (or one core feature pharmacological contradictions.
and one suggestive feature) are sufficient for a diagnosis No controlled clinical trials have evaluated the
of probable DLB; one core feature or one or more treatment of parkinsonism in DLB, although there are
suggestive features are sufficient for a diagnosis of reports of small series of DLB patients whose motor
possible DLB.12 impairments were successfully treated with levodopa.31
Clinical experience suggests that levodopa is less effec-
tive in treating motor impairments in DLB than in
DIAGNOSIS idiopathic Parkinson’s disease. Because the parkinson-
As with Alzheimer’s disease, definitive diagnosis of DLB ism may be mild and the dopaminergic agents may
rests on brain autopsy following death. Generally speak- produce side effects, such as a worsening of attention
ing, the accuracy of the clinical diagnosis of DLB is and visual hallucinations, one should only treat the
relatively low based on the consortium for DLB diag- movement disorder if the symptoms interfere with
nostic criteria prior to the recent revision. Using the function. Antiparkinsonism drugs should be started at
older criteria against neuropathology, several studies the lowest possible dose and increased with caution.
have reported acceptable specificities (71 to 95%) but Visual hallucinations and delusions may be ex-
suboptimal sensitivities (18 to 83%).17,28 There are acerbated by dopaminergic agents, so tapering of these
currently no cerebrospinal fluid or genotypic biomarkers agents is typically the first step in managing psychosis.
available to support a diagnosis of DLB in life.29 The visual hallucinations may respond to AChE inhib-
itors32 or antipsychotics. Traditional neuroleptics are
best avoided. If necessary, the newer atypical antipsy-
TREATMENT chotic medications can be used in lowest effective dose
The treatment of DLB generally parallels that for and cautiously. We often begin with quetiapine (Sero-
Alzheimer’s disease. There are no preventatives or cura- quel) 12.5 to 25 mg and titrate to an effective dose. It is
tive agents for DLB. worth mentioning that hallucinations that are non-
It has been suggested that acetylcholinesterase threatening to the patient and do not disturb function
(AChE) inhibitors may be more effective in treating may not require any pharmacological intervention. Per-
DLB due to early and prominent central nervous sistent postural hypotension may respond to midodrine
system cholinergic dysfunction in these patients.30 (an a agonist) and fludrocortisone (a mineralocorticoid)
However, only one double-blind, placebo-controlled, or a reduction in blood pressure medications. REM sleep
multicenter trial has evaluated the safety and efficacy of behavior disorder typically responds to low doses of
AChE inhibitors in the treatment of DLB (although clonazepam prior to sleep.
DEMENTIA WITH LEWY BODIES/WEISMAN, McKEITH 47

REFERENCES dementia with Lewy bodies. Neurology 2000;54:1050–


1058
1. Hansen LA, Samuel W. Criteria for Alzheimer’s disease and 18. Collerton D, Burn D, McKeith I, O’Brien J. Systematic review
the nosology of dementia with Lewy bodies. Neurology and meta-analysis show that dementia with Lewy bodies is a
1997;48:126–132 visual-perceptual and attentional-executive dementia. Dement
2. Lewy FH. Handbuch der Neurologie. Berlin: Springer; 1912 Geriatr Cogn Disord 2003;16:229–237
3. Kosaka K, Yoshimura M, Ikeda K, Budka H. Diffuse type of 19. Tiraboschi P, Salmon DP, Hansen LA, Hofstetter RC, Thal
Lewy body disease: progressive dementia with abundant LJ, Corey-Bloom J. What best differentiates Lewy body from
cortical Lewy bodies and senile changes of varying degree—a Alzheimer’s disease in early-stage dementia? Brain 2006;129:
new disease? Clin Neuropathol 1984;3:185–192 729–735
4. Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation 20. Briel RC, McKeith IG, Barker WA, et al. EEG findings in
in the alpha-synuclein gene identified in families with dementia with Lewy bodies and Alzheimer’s disease. J Neurol
Parkinson’s disease. Science 1997;276:2045–2047 Neurosurg Psychiatry 1999;66:401–403
5. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, 21. Ferman TJ, Smith GE, Boeve BF, et al. DLB fluctuations:
Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. specific features that reliably differentiate DLB from AD and
Nature 1997;388:839–840 normal aging. Neurology 2004;62:181–187
6. Dawson TM, Dawson VL. Molecular pathways of neuro- 22. Harding AJ, Broe GA, Halliday GM. Visual hallucinations
degeneration in Parkinson’s disease. Science 2003;302:819– in Lewy body disease relate to Lewy bodies in the temporal
822 lobe. Brain 2002;125:391–403
7. Galvin JE, Lee VM, Trojanowski JQ. Synucleinopathies: 23. McKeith IG, Wesnes KA, Perry E, Ferrara R. Hallucinations
clinical and pathological implications. Arch Neurol 2001;58: predict attentional improvements with rivastigmine in demen-
186–190 tia with Lewy bodies. Dement Geriatr Cogn Disord 2004;18:
8. Kotzbauer PT, Trojanowsk JQ, Lee VM. Lewy body 94–100
pathology in Alzheimer’s disease. J Mol Neurosci 2001;17: 24. Molloy S, McKeith IG, O’Brien JT, Burn DJ. The role of
225–232 levodopa in the management of dementia with Lewy bodies.
9. Minoshima S, Foster NL, Petrie EC, Albin RL, Frey KA, J Neurol Neurosurg Psychiatry 2005;76:1200–1203
Kuhl DE. Neuroimaging in dementia with Lewy bodies: 25. McKeith IG, Perry RH, Fairbairn AF, Jabeen S, Perry EK.
metabolism, neurochemistry, and morphology. J Geriatr Operational criteria for senile dementia of Lewy body type
Psychiatry Neurol 2002;15:200–209 (SDLT). Psychol Med 1992;22:911–922
10. Colosimo C, Hughes AJ, Kilford L, Lees AJ. Lewy body 26. Walker Z, Costa DC, Walker RW, et al. Striatal dopamine
cortical involvement may not always predict dementia in transporter in dementia with Lewy bodies and Parkinson
Parkinson’s disease. J Neurol Neurosurg Psychiatry 2003;74: disease: a comparison. Neurology 2004;62:1568–1572
852–856 27. Taki J, Yoshita M, Yamada M, Tonami N. Significance of
11. Merdes AR, Hansen LA, Jeste DV, et al. Influence of 123I-MIBG scintigraphy as a pathophysiological indicator in
Alzheimer pathology on clinical diagnostic accuracy in the assessment of Parkinson’s disease and related disorders: it
dementia with Lewy bodies. Neurology 2003;60:1586–1590 can be a specific marker for Lewy body disease. Ann Nucl
12. McKeith IG, Dickson DW, Lowe J, et al. Diagnosis and Med 2004;18:453–461
management of dementia with Lewy bodies: third report of 28. Litvan I, MacIntyre A, Goetz CG, et al. Accuracy of the
the DLB Consortium. Neurology 2005;65:1863–1872 clinical diagnoses of Lewy body disease, Parkinson disease,
13. Lauterbach EC. The neuropsychiatry of Parkinson’s disease. and dementia with Lewy bodies: a clinicopathologic study.
Minerva Med 2005;96:155–173 Arch Neurol 1998;55:969–978
14. Hurtig HI, Trojanowski JQ, Galvin J, et al. Alpha-synuclein 29. Galasko D. Cerebrospinal fluid biomarkers in Alzheimer
cortical Lewy bodies correlate with dementia in Parkinson’s disease: a fractional improvement? Arch Neurol 2003;60:
disease. Neurology 2000;54:1916–1921 1195–1196
15. Aarsland D, Perry R, Brown A, Larsen JP, Ballard C. 30. Touchon J, Bergman H, Bullock R, Rapatz G, Nagel J, Lane
Neuropathology of dementia in Parkinson’s disease: a R. Response to rivastigmine or donepezil in Alzheimer’s
prospective, community-based study. Ann Neurol 2005;58: patients with symptoms suggestive of concomitant Lewy
773–776 body pathology. Curr Med Res Opin 2006;22:49–59
16. McKeith IG, Galasko D, Kosaka K, et al. Consensus 31. Molloy S, McKeith IG, O’Brien JT, Burn DJ. The role of
guidelines for the clinical and pathologic diagnosis of levodopa in the management of dementia with Lewy bodies.
dementia with Lewy bodies (DLB): report of the consortium J Neurol Neurosurg Psychiatry 2005;76:1200–1203
on DLB international workshop. Neurology 1996;47:1113– 32. Aarsland D, Mosimann UP, McKeith IG. Role of chol-
1124 inesterase inhibitors in Parkinson’s disease and dementia with
17. McKeith IG, Ballard CG, Perry RH, et al. Prospective Lewy bodies. J Geriatr Psychiatry Neurol 2004;17:164–171
validation of consensus criteria for the diagnosis of

Anda mungkin juga menyukai