26.10.2013 Views

Behavioural-variant frontotemporal dementia: diagnosis, clinical ...

Behavioural-variant frontotemporal dementia: diagnosis, clinical ...

Behavioural-variant frontotemporal dementia: diagnosis, clinical ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Review<br />

Lancet Neurol 2011; 10: 162–72<br />

Published Online<br />

December 13, 2010<br />

DOI:10.1016/S1474-<br />

4422(10)70299-4<br />

Neuroscience Research<br />

Australia, Randwick, NSW,<br />

Australia (O Piguet PhD,<br />

M Hornberger PhD,<br />

E Mioshi PhD,<br />

Prof J R Hodges FMedSci)<br />

Correspondence to:<br />

Prof John Hodges, Neuroscience<br />

Research Australia, Barker St,<br />

Randwick, NSW 2031, Australia<br />

j.hodges@neura.edu.au<br />

<strong>Behavioural</strong>-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>: <strong>diagnosis</strong>,<br />

<strong>clinical</strong> staging, and management<br />

Olivier Piguet, Michael Hornberger, Eneida Mioshi, John R Hodges<br />

Patients with behavioural-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong> (bvFTD) present with insidious changes in personality<br />

and interpersonal conduct that indicate progressive disintegration of the neural circuits involved in social cognition,<br />

emotion regulation, motivation, and decision making. The underlying pathological changes are heterogeneous and<br />

are characterised by various intraneuronal inclusions. Biomarkers to detect these histopathological changes in life are<br />

becoming increasingly important with the development of disease-modifying drugs. Gene mutations have been<br />

found that collectively account for around 10–20% of cases. Recently, criteria proposed for bvFTD defi ne three levels<br />

of diagnostic certainty: possible, probable, and defi nite. Detailed history taking from family members to elicit<br />

behavioural features underpins the diagnostic process, with support from neuropsychological testing designed to<br />

detect impairment in decision making, emotion processing, and social cognition. Brain imaging is important for<br />

increasing the level of diagnostic certainty. A recently developed staging instrument shows much promise for<br />

monitoring patients and evaluating therapies, which at present are aimed at symptom amelioration. Carer education<br />

and support remain of paramount importance.<br />

Introduction<br />

Frontotemporal <strong>dementia</strong> (FTD) is the <strong>clinical</strong> diagnostic<br />

term now preferred to describe patients with a range of<br />

progressive <strong>dementia</strong> syndromes associated with focal<br />

atrophy of the orbitomesial frontal and anterior temporal<br />

lobes. 1 Epidemiological studies suggest that FTD is the<br />

second most common cause of young-onset <strong>dementia</strong><br />

after Alzheimer’s disease (AD). 2,3 Two independent<br />

studies from the UK revealed a prevalence of around 15<br />

cases per 100 000 population aged 45–64 years (95% CI<br />

8–27 per 100 000). 2 Although thought to be a rare cause<br />

of <strong>dementia</strong> after age 65 years, FTD might be more<br />

common than assumed because older adults rarely<br />

undergo the types of investigation needed to establish a<br />

confi dent <strong>diagnosis</strong> in vivo and are not generally followed<br />

to autopsy.<br />

Unlike AD, both the <strong>clinical</strong> profi le and the underlying<br />

pathological changes are heterogeneous in FTD. Two<br />

broad presentations are recognised: progressive<br />

deterioration in social function and personality, known<br />

as behavioural-<strong>variant</strong> FTD (bvFTD), and insidious<br />

decline in language skills, known as primary progressive<br />

aphasia, which can, in turn, be subdivided according the<br />

predominant pattern of language breakdown into<br />

progressive non-fl uent aphasia and semantic <strong>dementia</strong>. 4–6<br />

The syndrome of FTD overlaps with motor neuron<br />

disease (MND) both <strong>clinical</strong>ly and pathologically, and<br />

with some of the extrapyramidal motor disorders. Around<br />

10% of patients with FTD develop <strong>clinical</strong> and<br />

neurophysiological evidence of MND, 7,8 and, similarly,<br />

patients with MND show behavioural and/or language<br />

changes that, in some instances, are severe enough to<br />

qualify for a <strong>diagnosis</strong> of FTD. 9 Of the extrapyramidal<br />

disorders, corticobasal degeneration and progressive<br />

supranuclear palsy show substantial overlap with FTD<br />

and share the fi nding of abnormal tau pathology. 10<br />

This is a broad and rapidly evolving fi eld; therefore, in<br />

this Review, we have focused on the <strong>clinical</strong> aspects of<br />

bvFTD because there have been recent authoritative<br />

reviews of the aphasic syndromes. 4,6 Our aim is to place<br />

advances in the <strong>diagnosis</strong>, staging, and management of<br />

bvFTD within the context of recent pathological and<br />

genetic discoveries. The assessment of any patient with<br />

suspected bvFTD should involve behavioural and<br />

neuropsychiatric tests, assessment of everyday abilities,<br />

cognitive testing, and neuroimaging. Various blood and<br />

CSF biomarkers are under development, but are not yet<br />

available for routine <strong>clinical</strong> application. Genetic testing<br />

is advised for those at high risk of a gene mutation<br />

(fi gure 1). 11<br />

Pathology<br />

The subtypes of underlying pathological changes in<br />

patients with FTD are classifi ed on the basis of the<br />

pattern of protein accumulation, and are referred to<br />

collectively as <strong>frontotemporal</strong> lobar degeneration<br />

(FTLD). 12 At post mortem, cases share, by defi nition, the<br />

fi nding of bilateral <strong>frontotemporal</strong> atrophy with<br />

neuronal loss, microvacuolation, and a variable degree<br />

of astrocytic gliosis. The progression of this atrophy has<br />

been examined by mapping the pattern in patients with<br />

diff erent disease duration. 13 Initially, mesial and orbital<br />

frontal regions bear the brunt of the atrophy, followed<br />

by the temporal pole, hippocampal formation,<br />

dorsolateral frontal cortex, and the basal ganglia. This<br />

pattern of progression of atrophy has been shown to<br />

relate to the volume of cortical and subcortical regions,<br />

and to the underlying neuron loss. 14,15 Furthermore, it<br />

forms the basis of a useful and quick MRI rating scale<br />

described below.<br />

The use of immunohistochemical staining has<br />

revolutionised the fi eld of research in FTD. Inclusions of<br />

the microtubule-binding protein tau are present in<br />

approximately 40% of cases (FTLD-tau). 16 Tau-positive<br />

cases include the subset of patients with mutations in the<br />

microtubule-associated protein tau (MAPT) gene. Further<br />

162 www.thelancet.com/neurology Vol 10 February 2011


Possible phenocopy<br />

Possible bvFTD based on <strong>clinical</strong>, behavioural,<br />

cognitive, and ADL assessments*<br />

MRI<br />

Abnormal<br />

PET Probable bvFTD<br />

Normal<br />

Normal<br />

Abnormal<br />

Strong family history<br />

Figure 1: Possible investigations after the <strong>diagnosis</strong> of suspected bvFTD<br />

based on <strong>clinical</strong> assessment<br />

*Presence of neurodegenerative disease and at least three of six behavioural or<br />

cognitive core diagnostic criteria are required for <strong>diagnosis</strong> of the disease.<br />

bvFTD=behavioural-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>. ADL=activities of<br />

daily living.<br />

subclassifi cation is based on morphological criteria and<br />

the predominance of either tau with three microtubulebinding<br />

repeats (3R tau) or four microtubule-binding<br />

repeats (4R tau). 12 Most of the remaining cases are tau<br />

negative and ubiquitin positive, and have inclusions<br />

comprising the 43 kDa TAR DNA-binding protein (TDP-43;<br />

FTLD-TDP), but a minority (around 5–10%) are negative<br />

for both tau and TDP-43 proteins. 17 Recently, inclusions of<br />

the RNA-binding protein fused in sarcoma (FUS) have<br />

been found in many of these cases (FTLD-FUS). 16,18 The<br />

search for FUS pathology was initiated after FUS gene<br />

mutations were identifi ed in cases of familial MND. 19,20<br />

Both TDP-43 and FUS are RNA-processing proteins,<br />

although the mechanisms leading to TDP-43 and FUS<br />

accumulation and resulting neurodegeneration have not<br />

yet been elucidated. A small proportion of cases have<br />

either no inclusions or show ubiquitin-positive inclusions<br />

that are TDP-43 and FUS negative, suggesting that<br />

additional protein abnormalities will be found in FTLD. 16<br />

A major topic of investigation has been to establish the<br />

association between <strong>clinical</strong> phenotypes and molecular<br />

pathology. Unlike the progressive aphasic syndromes,<br />

which are generally associated more with one histological<br />

form of FTLD than another (progressive non-fl uent<br />

aphasia with FTLD-tau, semantic <strong>dementia</strong> with<br />

FTLD-TDP), in bvFTD any of the histological <strong>variant</strong>s<br />

can be found with an approximately 50:50 split between<br />

FTLD-tau and FTLD-TDP, 21–23 and a small proportion of<br />

FTLD-FUS cases. 24 With the advent of potential diseasemodifying<br />

therapies, ascertainment of a pathological<br />

<strong>diagnosis</strong> in vivo will be increasingly important. As yet,<br />

no reliable method for detecting these pathological<br />

changes in life exists. CSF biomarkers seem to be the<br />

most promising in distinguishing FTLD from AD: the<br />

ratio of tau to amyloid β 1–42 has been found to be lower in<br />

FTLD than in AD. 25 Increased concentrations of<br />

Yes<br />

Genetic screening<br />

CSF TDP-43 have also been reported in patients with<br />

MND and FTD, but without pathological confi rmation of<br />

the <strong>diagnosis</strong>. In addition, the substantial overlap<br />

between cases and controls limits the applicability of the<br />

assay in <strong>clinical</strong> practice. 26 Serum progranulin<br />

concentrations are exceptionally low in patients with<br />

mutations in the granulin gene (GRN), 27–29 and this assay<br />

seems to be a sensitive and specifi c method of screening<br />

for such cases, 30 with suggestions that plasma TDP-43<br />

concentrations could be related to brain pathology. 31<br />

Neuroimaging markers are discussed below.<br />

Genetics<br />

Up to 40% of patients with FTD have a family history<br />

of <strong>dementia</strong>, 3 but the high community prevalence of<br />

non-FTD <strong>dementia</strong> means that many of the elderly family<br />

members included in such estimates almost certainly<br />

have other causes of <strong>dementia</strong>. Patients with an autosomal<br />

dominant pattern (aff ected fi rst-degree relatives across<br />

two generations) account for only 10% of cases. 11 The<br />

strength of family history is highly predictive, in that<br />

mutations can now be shown in most patients with two<br />

or more fi rst-degree relatives with a <strong>dementia</strong> syndrome<br />

compatible with FTD. 11 Overall, patients with mutations<br />

in MAPT and GRN each account for 5–11% of total FTD<br />

cases. 11 Mutations in the gene that encodes TDP-43,<br />

TARDBP, and in FUS, recognised as a cause of familial<br />

amyotrophic lateral sclerosis (ALS), have also been<br />

identifi ed in a small number of cases of FTD-ALS, 32–35 but<br />

seem to be rare in uncomplicated FTD. 11 Rare genetic<br />

mutations causing FTD include those in the genes<br />

encoding valosin-containing protein (VCP) and charged<br />

multivesicular body protein 2B (CHMP2B; also known<br />

as chromatin-modifying protein 2B). Mutations in VCP<br />

cause FTD in association with inclusion body myopathy<br />

and Paget’s disease of bone, 36 whereas the CHMP2B gene<br />

mutation is mostly confi ned to a large Danish cohort<br />

with FTD. 37,38 Familial clusters of FTD and MND have<br />

been reported. A linkage study of FTD-MND clusters<br />

has indicated a common locus in the region of<br />

chromosome 9p13.2–21.3, 39 but the causative gene has<br />

not yet been identifi ed.<br />

From a practical perspective, a detailed family history<br />

should be taken for all patients with suspected FTD,<br />

bearing in mind the overlap between MND and FTD,<br />

that a <strong>diagnosis</strong> of FTD or Pick’s disease was rarely<br />

made in the past, and the phenotypic variability within<br />

families with gene mutations: one member might<br />

present with bvFTD, whereas others have a progressive<br />

aphasic syndrome or corticobasal syndrome. On the<br />

basis of a comprehensive analysis of the frequency of<br />

gene mutations according to strength of family history<br />

and <strong>clinical</strong> syndrome in a large <strong>clinical</strong> cohort, 31 we<br />

recommend that patients with one or more fi rst-degree<br />

relatives with a disease on the FTD spectrum should be<br />

screened for MAPT and GRN gene mutations after<br />

appropriate counselling in a <strong>clinical</strong> genetics setting.<br />

Review<br />

www.thelancet.com/neurology Vol 10 February 2011 163


Review<br />

Those with an informative family history that reveals no<br />

aff ected relatives can be confi dently reassured and need<br />

not undergo gene screening. 11 Of note, the age of onset<br />

in patients with MAPT mutations is almost always<br />

below 65 years, whereas those with GRN mutations are<br />

often older. 11<br />

<strong>Behavioural</strong> features<br />

Insidious changes in personality, interpersonal conduct,<br />

and emotional modulation characterise bvFTD and<br />

indicate progressive disintegration of the neural circuits<br />

involved in social cognition, emotion regulation,<br />

motivation, and decision making. 40–43 Onset is typically<br />

diffi cult to pinpoint. Since insight is limited, or absent, it<br />

is vital that close family members are interviewed alone,<br />

and sensitively, to elicit the nature of the early symptoms<br />

and their progression. Assessment and <strong>diagnosis</strong> have<br />

been greatly assisted by the development of carer-based<br />

questionnaires designed to document the range of<br />

symptoms found in the <strong>dementia</strong>, including the<br />

Neuropsychiatric Inventory, 44 Cambridge <strong>Behavioural</strong><br />

Inventory, 45 and Frontal <strong>Behavioural</strong> Inventory. 46 All of<br />

the features found in bvFTD can occur in other<br />

<strong>dementia</strong>s, but their predominance and early emergence<br />

typify bvFTD.<br />

Apathy is very common and manifests as inertia,<br />

reduced motivation, lack of interest in previous hobbies,<br />

and progressive social isolation. Disinhibition often<br />

coexists with apathy, and produces impulsive actions<br />

leading to overspending, tactless or sexually inappropriate<br />

remarks, and a range of socially embarrassing behaviour.<br />

New-onset pathological gambling or, more rarely, hyperreligiosity,<br />

can be the presenting feature. 47,48 Repetitive or<br />

stereotypic behaviours might be apparent with<br />

perseveration and a tendency to repeat phrases, stories,<br />

or jokes. Some carers describe excessive hoarding, which<br />

results in a state of squalor. Patients often lack empathy<br />

and an inappropriately subdued grief reaction is a<br />

common early symptom. Mental rigidity is common and<br />

patients can have diffi culty adapting to new situations or<br />

routines. A blunting of aff ect and reduction in range of<br />

emotional expression is frequent and some patients show<br />

elevation of mood resembling hypomania. Changes in<br />

eating behaviour, with impaired satiety, change in<br />

preferences towards sweet food, and dysregulation of<br />

food intake are common and seen across cultures. 49<br />

These alterations in eating have recently been found to<br />

be related to pathological changes in the hypothalamus,<br />

which is crucial for coordinating metabolic needs,<br />

including feeding. 50<br />

Psychotic symptoms such as delusions, paranoid<br />

ideation, and hallucinations are relatively rare in FTD,<br />

except in patients with combined MND and in patients<br />

with young-onset FTLD-FUS in whom such features are<br />

present in up to 50%. 7,51,52 Clinically, these patients seem<br />

to present with fl orid behavioural symptoms. In addition,<br />

their age of onset is often exceptionally young (≤40 years), 24<br />

and a positive family history seems rare in keeping with<br />

the absence of FUS gene mutations in this group. 24,52<br />

Patients destined to develop <strong>clinical</strong> MND who have<br />

FTLD-TDP also have a high rate of psychotic features<br />

and progress rapidly even before the onset of typically<br />

bulbar MND. 7<br />

Of the features listed above, social disinhibition,<br />

euphoria, stereotypical and aberrant motor behaviour,<br />

and changes in eating preference most clearly<br />

discriminate bvFTD from AD. 45,53 Variability in the<br />

symptom profi les reported across studies might have<br />

arisen from the aggregation of patients at diff erent stages<br />

of the disease. Increased behavioural changes have been<br />

associated with disease severity. 54 Agitation, disinhibition,<br />

and irritability also seem to be more frequent in the later<br />

stages, 55 whereas restlessness and hyperorality are<br />

present throughout the disease. 56 Another important<br />

variable is age of disease onset, with apathy being more<br />

prominent in late-onset bvFTD, 57 although this fi nding is<br />

not universal. 58<br />

In summary, behavioural assessment is at the core of<br />

assessment in patients with potential bvFTD and seems<br />

to be more sensitive in distinguishing bvFTD from AD<br />

than standard cognitive testing. Despite a substantial<br />

increase in our knowledge of the behavioural changes in<br />

bvFTD, which are at the root of so much carer distress<br />

(see below), much remains uncertain concerning their<br />

specifi city, neural basis, and their relation to the<br />

underlying pathology.<br />

bvFTD phenocopy syndrome: implications for<br />

diagnostic criteria<br />

The <strong>diagnosis</strong> of bvFTD is by no means an easy task in<br />

the early stages, and many of the symptoms overlap with<br />

those seen in psychiatric disorders and other <strong>dementia</strong>s. 51<br />

It is also increasingly apparent that a subset of patients<br />

who present with the <strong>clinical</strong> features of bvFTD do not<br />

progress to frank incapacitating <strong>dementia</strong>. 59 Such patients<br />

are almost always men and they either remain stable over<br />

many years or improve. 60,61 The symptom profi le as<br />

reported by family members is identical, except that<br />

activities of daily living (ADL) are less disrupted. 60,62<br />

Several features distinguish these non-progressor or<br />

phenocopy cases from those with true FTD, notably<br />

normal or marginal impairment on neuropsychological<br />

tests of executive function, preserved memory and social<br />

cognition, a lack of overt atrophy on MRI, and normal<br />

metabolic (PET) brain imaging. 59–61,63 The aetiology of the<br />

phenocopy syndrome is a matter of debate. A proportion<br />

of patients seem to have a developmental personality<br />

disorder on the Asperger’s spectrum with decompensation<br />

due to altered life circumstances (Hodges J, unpublished).<br />

Some might have a chronic low-grade mood disorder, but<br />

others remain a mystery.<br />

The phenocopy syndrome has major implications for<br />

the current <strong>clinical</strong> diagnostic criteria for FTD, 5 which<br />

require a profi le of symptoms compatible with the<br />

164 www.thelancet.com/neurology Vol 10 February 2011


<strong>diagnosis</strong> without imaging or other confi rmatory test<br />

results. These criteria also present diffi culties in their<br />

application due to under-specifi cation of some features<br />

and were derived by <strong>clinical</strong> consensus prior to the<br />

publication of quantitative studies comparing cohorts<br />

with pathologically verifi ed diagnoses. Recently proposed<br />

criteria developed by an international FTD research<br />

group (panel) 64 build on recent work with operationalised<br />

defi nitions that have three levels of diagnostic certainty:<br />

possible, probable, and defi nite bvFTD. Patients qualify<br />

for possible bvFTD on the basis of three core behavioural<br />

or cognitive features (including social disinhibition,<br />

apathy, loss of empathy, stereotypic behaviours or<br />

alterations in eating pattern, and neuropsychological<br />

defi cits indicative of frontal executive dysfunction). A<br />

probable <strong>diagnosis</strong> requires the same <strong>clinical</strong> features<br />

with evidence of progression and unequivocal neuroimaging<br />

abnormalities. The term “defi nite” is reserved<br />

for those with neuropathology or a pathogenic gene<br />

mutation. These new criteria (panel) are currently<br />

undergoing validation against neuropathological changes<br />

by an international consortium of researchers. 64<br />

Neuropsychology<br />

Cognition<br />

Early in the disease process, patients with bvFTD can<br />

perform relatively well on formal neuropsychological<br />

tests despite the presence of signifi cant personality and<br />

behavioural changes. 65 The mini mental state examination<br />

is insensitive, but the Addenbrooke’s cognitive<br />

examination seems to detect at least 90% of cases at<br />

presentation. 66 The prototypical cognitive profi le is one of<br />

relatively preserved language and visuospatial/<br />

constructive abilities. Whether patients with bvFTD show<br />

executive dysfunctions remains contentious, 67,68 and has<br />

been complicated by the inclusion of phenocopy cases.<br />

However, such defi cits constitute a central diagnostic<br />

feature of the newly proposed <strong>clinical</strong> diagnostic criteria. 64<br />

Recent evidence suggests that the combination of specifi c<br />

tests (eg, digit span backward task, Hayling test of<br />

response inhibition, and the short version of the executive<br />

and social cognition battery) might help diff erentiate<br />

these cases, because results are typically abnormal in<br />

patients with true bvFTD and normal in phenocopy<br />

cases. 68,69<br />

The presence of severe defi cits of episodic memory has<br />

been used as an exclusion criterion for a <strong>clinical</strong> <strong>diagnosis</strong><br />

of bvFTD, 5 although a proportion (10–15%) of patients<br />

with pathologically confi rmed FTLD present with severe<br />

amnesia. 21,70 A recent report has indicated that defi cits in<br />

episodic memory are more common than previously<br />

reported. 71 Carefully selected patients with bvFTD (ie,<br />

after excluding phenocopy cases) had similar memory<br />

impairments as seen in AD on tests of episodic memory,<br />

even after accounting for disease severity. 71 The criterion<br />

of relative sparing of episodic memory compared with<br />

executive functions proposed in the recent international<br />

consensus criteria for bvFTD might need to be revisited<br />

in the light of current research. 62<br />

The evidence reviewed thus far indicates that no specifi c<br />

cognitive profi le seems to be associated with bvFTD early<br />

in the disease, although careful cognitive assessment will<br />

reveal defi cits, generally in the domains of executive<br />

function and episodic memory. With disease progression,<br />

the atrophy evolves to involve the anterior temporal<br />

regions, and the pattern of defi cits becomes less distinct<br />

from other FTD subtypes, notably semantic <strong>dementia</strong>. 10<br />

Panel: International consensus criteria for bvFTD<br />

Review<br />

Neurodegenerative disease<br />

Must be present for any FTD <strong>clinical</strong> syndrome<br />

Shows progressive deterioration of behaviour and/or cognition by observation or history<br />

Possible bvFTD<br />

Three of the features (A–F) must be present; symptoms should occur repeatedly, not just<br />

as a single instance:<br />

A Early (3 years) behavioural disinhibition<br />

B Early (3 years) apathy or inertia<br />

C Early (3 years) loss of sympathy or empathy<br />

D Early (3 years) perseverative, stereotyped, or compulsive/ritualistic behaviour<br />

E Hyperorality and dietary changes<br />

F Neuropsychological profi le: executive function defi cits with relative sparing of<br />

memory and visuospatial functions<br />

Probable bvFTD<br />

All the following criteria must be present to meet <strong>diagnosis</strong>:<br />

A Meets criteria for possible bvFTD<br />

B Signifi cant functional decline<br />

C Imaging results consistent with bvFTD (frontal and/or anterior temporal atrophy on<br />

CT or MRI or frontal hypoperfusion or hypometabolism on SPECT or PET)<br />

Defi nite bvFTD<br />

Criteria A and either B or C must be present to meet <strong>diagnosis</strong>:<br />

A Meets criteria for possible or probable bvFTD<br />

B Histopathological evidence of FTLD on biopsy at post mortem<br />

C Presence of a known pathogenic mutation<br />

Exclusion criteria for bvFTD<br />

Criteria A and B must both be answered negatively; criterion C can be positive for possible<br />

bvFTD but must be negative for probable bvFTD:<br />

A Pattern of defi cits is better accounted for by other non-degenerative nervous system<br />

or medical disorders<br />

B <strong>Behavioural</strong> disturbance is better accounted for by a psychiatric <strong>diagnosis</strong><br />

C Biomarkers strongly indicative of Alzheimer’s disease or other neurodegenerative process<br />

Additional features<br />

A Presence of motor neuron fi ndings suggestive of motor neuron disease<br />

B Motor symptoms and signs similar to corticobasal degeneration and progressive<br />

supranuclear palsy<br />

C Impaired word and object knowledge<br />

D Motor speech defi cits<br />

E Substantial grammatical defi cits<br />

Criteria from Rascovsky et al. 64 bvFTD=behavioural-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>. FTD=<strong>frontotemporal</strong><br />

<strong>dementia</strong>. SPECT=single-photon emission computed tomography. FTLD=<strong>frontotemporal</strong> lobar degeneration.<br />

www.thelancet.com/neurology Vol 10 February 2011 165


Review<br />

Mild Moderate Severe<br />

Dorsolateral prefrontal and<br />

medial prefrontal cortices<br />

Orbitofrontal cortex<br />

Figure 2: Visual rating scale to estimate the severity of cortical atrophy in the dorsolateral, medial, and<br />

orbitofrontal cortices in suspected cases of bvFTD<br />

Modifi ed from Hornberger et al, 87 by permission of Karger AG, Basel. bvFTD=behavioural-<strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong>.<br />

The diffi culty in identifying profi les of cognitive defi cits<br />

specifi c to bvFTD has led to an interest in aspects of social<br />

cognition (theory of mind), emotion recognition and<br />

complex problem solving, and the use of naturalistic tasks<br />

(ie, tasks indicating cognitive and non-cognitive demands<br />

more akin to daily activities). The orbitomesial frontal<br />

cortices are crucial for performance in these domains,<br />

and lesions within these brain regions have been shown<br />

to negatively aff ect tests measuring these cognitive<br />

constructs. 72 The Iowa gambling task of complex decision<br />

making requires individuals to inhibit responses that<br />

result in short-term high fi nancial gains but long-term<br />

losses in favour of responses resulting in small gains in<br />

the short term and reduced long-term losses. 73 This task<br />

was found to diff erentiate between patients with bvFTD,<br />

who consistently failed to inhibit the prepotent responses<br />

favouring short-term gains leading to long-term loss, and<br />

healthy controls. Interestingly, this defi cit is not related to<br />

performance on other cognitive tasks. 74<br />

Emotion recognition and social cognition<br />

A marked impairment in emotion detection and<br />

recognition is evident early in the course of bvFTD, 75,76<br />

and seems to be most pronounced for negative emotions<br />

(eg, fear, sadness, anger, and disgust). 76,77 Disorders of<br />

emotion detection and regulation are part of the <strong>clinical</strong><br />

diagnostic criteria for the disease (ie, early emotional<br />

blunting, early decline in social interpersonal conduct).<br />

However, such defi cits are not limited to this subtype of<br />

FTD and are also present in semantic <strong>dementia</strong>. 78<br />

Diffi culties in detecting and understanding emotions are<br />

observed with static (photos) or dynamic (fi lms) visual<br />

stimuli, voices, or even music. Importantly, physiological<br />

responses (eg, skin conductance) to some emotional<br />

stimuli are preserved. 79 Defi cits have also been observed<br />

in detecting more complex emotions, such as<br />

embarrassment. 80 These defi cits might be amenable to<br />

retraining to enhance their recognition and improve<br />

interpersonal relationships.<br />

Patients with bvFTD are also impaired on many aspects<br />

of social cognition. For example, the often reported<br />

feature of lack of empathy and coldness is confi rmed on<br />

formal testing. 81 Theory of mind is impaired in bvFTD,<br />

as exemplifi ed by defective ability to infer intention and<br />

mental states in others or to take someone else’s point of<br />

view, 74,75,82 and impaired detection of social faux pas, 82<br />

discrimination of sincere from sarcastic exchanges, 63<br />

and understanding of situations requiring moral<br />

judgment. 83 Whereas most of the tasks developed remain<br />

in the research arena, well validated tests of emotion and<br />

sarcasm detection exist, 84,85 which will hopefully become<br />

part of the standard cognitive assessment in suspected<br />

bvFTD.<br />

Neuroimaging<br />

By use of structural MRI, atrophy of the mesial frontal,<br />

orbitofrontal, and anterior insula cortices can be reliably<br />

observed on images acquired in the coronal plane<br />

(fi gure 2). 42,86,87 A combination of frontal and anterior<br />

temporal cortical and basal ganglia atrophy might also be<br />

seen at presentation in some patients. 88 This atrophy can<br />

be quickly and reliably estimated using relatively simple<br />

visual rating scales developed specifi cally for FTD. 89<br />

However, an apparently normal MRI on visual inspection<br />

does not completely exclude a <strong>diagnosis</strong> of bvFTD,<br />

because the changes can be subtle in the early stages.<br />

The development of automated quantitative methods,<br />

including voxel-based morphometry and cortical<br />

thickness mapping, has been important in revealing<br />

selective atrophy of the anterior cingulate and frontal<br />

insular cortices early in the course of bvFTD, 42,90,91 which<br />

is distinct from the pattern seen in other <strong>variant</strong>s of FTD<br />

and in AD. 92 The anterior cingulate-frontal insular<br />

complex contains a unique population of neurons, the<br />

von Economo cells, which are thought to be crucial in the<br />

development and maintenance of social cognition and<br />

have been shown to be depleted in patients with bvFTD<br />

at autopsy. 43,93 More recently, sophisticated MRI methods<br />

of exploring network connectivity have shown signifi cant<br />

changes in connectivity among the regions most sensitive<br />

to atrophy in bvFTD compared with healthy controls or<br />

patients with other <strong>dementia</strong> syndromes. 94,95<br />

Patterns of grey matter atrophy might be predictive of<br />

the underlying pathological process in bvFTD, with<br />

bilateral dorsolateral prefrontal atrophy being suggestive<br />

of Pick body inclusions (ie, Pick’s disease), and<br />

166 www.thelancet.com/neurology Vol 10 February 2011


asymmetric left and right temporal lobe atrophy being<br />

associated with FTLD-TDP and FTLD-tau, respectively. 96<br />

Marked atrophy of the caudate nucleus might also be<br />

predictive of FTLD-FUS. 24,97 However, patterns of atrophy<br />

seem to relate more closely to <strong>clinical</strong> features than to<br />

specifi c pathological changes. 98<br />

Brain changes in bvFTD are not limited to the cortex.<br />

Atrophy is also present in many subcortical brain regions,<br />

including the amygdala, hippocampus, caudate, striatum,<br />

putamen, thalamus, and hypothalamus, 50,99 accompanied<br />

by reduction in connectivity among subcortical structures. 95<br />

In addition, atrophy of the amygdala has been proven to be<br />

an effi cient discriminator between bvFTD and AD. 99<br />

By contrast with the well documented cortical grey<br />

matter changes, presence and severity of white matter<br />

changes in bvFTD have only recently been investigated. 100<br />

Frontal lobe white matter volume reduction largely<br />

parallels the atrophy in the adjacent grey matter in bvFTD,<br />

with diff erent subtypes of FTD showing specifi c patterns<br />

of white matter atrophy. 101 Using diff usion tensor imaging,<br />

an index of changes in the microstructural organisation of<br />

white matter, a study has successfully diff erentiated<br />

bvFTD from AD and other FTD subtypes. 102 Patients with<br />

bvFTD seem to show a selective reduction in some white<br />

matter tracts (superior longitudinal fasciculus, uncinate<br />

fasciculus, cingulum tracts, and genu and splenium of the<br />

corpus callosum), particularly those within frontal lobe<br />

(eg, genu of the corpus callosum) or those connecting<br />

frontal and temporal brain regions (eg, uncinate<br />

fasciculus). 102 Whether these white matter changes are<br />

reliable diagnostic markers for bvFTD remains unclear.<br />

Patients with the phenocopy syndrome who present with<br />

the <strong>clinical</strong> features of bvFTD show normal or marginal<br />

MRI changes. 61,89<br />

Serial MRIs show signifi cant atrophy over time in<br />

bvFTD. Annual rate of overall brain atrophy can reach 8%,<br />

almost twice as severe as that documented in AD, although<br />

some patients with bvFTD show atrophy rates similar to<br />

those seen in healthy controls (0·3% per year), which<br />

might be due to the inclusion of phenocopy cases. 103<br />

Functional neuroimaging techniques, such as [ 99m Tc]hexamethylpropyleneamine<br />

oxime single-photon emission<br />

computed tomography (SPECT) or [ 18 F]-fl uorodeoxyglucose<br />

(FDG)-PET are increasingly being used to help with the<br />

<strong>diagnosis</strong> of bvFTD. Frontal hypoperfusion is present on<br />

SPECT in bvFTD and diff ers from the pattern of<br />

hypoperfusion observed in AD, which is predominant in<br />

the temporoparietal and posterior cingulate cortices. 104<br />

Although SPECT seems to be more sensitive than<br />

structural MRI in detecting early pathological changes in<br />

bvFTD, quantifi cation and specifi city of these changes<br />

have not been established. Hypometabolism on FDG-PET<br />

is detected consistently and reliably in frontal brain regions<br />

in patients with bvFTD compared with those with AD, who<br />

show posterior cingulate hypometabolism early in the<br />

disease process. 105 These changes are detected before any<br />

changes are visible on structural MRI, making FDG-PET<br />

the most sensitive diagnostic tool currently available.<br />

FDG-PET is also particularly useful in helping to identify<br />

phenocopy cases as these will show preserved frontal<br />

metabolism. However, in patients showing clear brain<br />

atrophy on structural MRI, little additional diagnostic<br />

benefi t is gained by doing a PET scan, because focal<br />

atrophy is a positive predictive marker of FTD.<br />

A novel PET technique, which uses the amyloid-βdetecting<br />

[ 11 C]-Pittsburgh compound B, has shown<br />

promising results in discriminating AD and FTD<br />

cases, 106,107 particularly those presenting with language<br />

defi cits rather than behavioural changes. Its use as a<br />

routine test remains to be established, but its <strong>clinical</strong><br />

applicability is evident as therapeutic interventions are<br />

being developed that are likely to be pathology specifi c.<br />

In summary, neuroimaging investigations in the<br />

<strong>diagnosis</strong> of bvFTD are powerful tools, which can reliably<br />

diff erentiate bvFTD from other FTD subtypes and from<br />

other <strong>dementia</strong> syndromes, and can corroborate <strong>clinical</strong><br />

diagnostics based on neuropsychiatric symptoms.<br />

Functional abilities<br />

Disability in everyday life is more pronounced in bvFTD<br />

than in AD or in the language <strong>variant</strong>s of FTD, 65,108,109 even<br />

after controlling for length of symptoms or cognitive<br />

performance. 65,109 Compared with AD, a large proportion<br />

of patients with bvFTD are impaired in ADL, and show<br />

an early impairment in basic ADL at initial assessment. 110<br />

Marked changes in driving abilities occur and are<br />

associated with the degree of behavioural change, 111 which<br />

has clear practical implications. A 12-month follow-up<br />

study also identifi ed signifi cant changes in ADL, which<br />

were associated with general cognitive decline. 112 However,<br />

these changes were not present in phenocopy cases.<br />

Disease progression, functional staging, and<br />

survival<br />

Most studies, to date, have used the <strong>clinical</strong> <strong>dementia</strong><br />

rating (CDR) scale to measure <strong>dementia</strong> severity. 113 This<br />

instrument, which was originally developed for AD, is<br />

biased towards memory impairment and most likely<br />

underestimates the level of <strong>dementia</strong> severity in bvFTD. A<br />

version adapted for FTD includes language and behavioural<br />

domains (FTLD-CDR), 114 and the sum-of-boxes score seems<br />

to be sensitive to change in most patients with FTD.<br />

Recently, the <strong>frontotemporal</strong> <strong>dementia</strong> rating<br />

scale (FRS) was developed specifi cally for FTD. 115 This<br />

staging tool incorporates changes in behaviour and ADL<br />

(table). On the FRS, patients with bvFTD tend to show<br />

greater disease severity and a faster progression through<br />

the <strong>clinical</strong> stages than patients with the language<br />

<strong>variant</strong>s of FTD, even after adjusting for length of<br />

symptoms. 115 Importantly, the FRS confi rmed that the<br />

CDR tends to underestimate <strong>dementia</strong> severity in bvFTD,<br />

with a great proportion of moderate and severe cases on<br />

the FRS being rated as “questionable <strong>dementia</strong>” or “mild”<br />

on CDR.<br />

Review<br />

www.thelancet.com/neurology Vol 10 February 2011 167


Review<br />

For the Mental Capacity Act<br />

see http://www.publicguardian.<br />

gov.uk<br />

Patients with bvFTD seem to progress faster than<br />

patients with AD, with median survival (from fi rst<br />

assessment) of about 3·0–4·5 years, 110,116,117 although this<br />

fi nding has not been universal. 118 Factors determining<br />

reduced survival are associated with MND or ALS and<br />

language impairment at <strong>diagnosis</strong>. 117 The eff ect of<br />

neuropathological subtype on survival remains<br />

contentious. 116,119<br />

Therapeutic intervention and caregiver stress<br />

Currently, no disease-specifi c treatment interventions for<br />

FTD exist. Consequently, treatment largely remains<br />

supportive and involves a combination of nonpharmacological<br />

and pharmacological measures aimed at<br />

reducing the eff ect of distressing symptoms. 120 The role of<br />

pharmacological interventions in FTD remains uncertain,<br />

and only small and often confl icting treatment trials have<br />

been done so far; these studies have not considered the<br />

eff ect on carer stress as a major outcome variable.<br />

Selective serotonin reuptake inhibitors have been used to<br />

treat disinhibition and challenging behaviours, but<br />

evidence for their use remains contradictory. 121,122 Atypical<br />

antipsychotics such as olanzapine have been used for<br />

patients with prominent agitation, aggressive behaviour,<br />

or psychosis. 123 Anticholinesterase inhibitors, the mainstay<br />

of AD therapy, do not have an established role in the<br />

treatment of FTD. One study reported improvement in<br />

measures of behavioural disturbance and carer stress<br />

with rivastigmine, 124 although deterioration in neuropsychiatric<br />

symptoms without cognitive improve ment<br />

was shown with donepezil. 125 Several drugs under<br />

development attempt to reduce aggregation of tau or<br />

TDP-43 and hence slow the fundamental pathological<br />

process in FTD. 120,126<br />

Behaviour Activities of daily living<br />

Caregiver intervention, which should be the most<br />

eff ective treatment within the context of <strong>dementia</strong>, has<br />

also not been trialled in FTD caregivers. A model for<br />

behavioural management has been proposed, whereby a<br />

focus on the antecedent–behaviour–consequence model<br />

could be applied to diff erent patient situations with<br />

recommendations for specifi c behaviours exhibited in<br />

bvFTD, 127 but research studies are needed to verify their<br />

benefi ts. This would be timely, because caregivers of<br />

patients with FTD present with high rates of burden and<br />

stress. Recent studies have shown that caregiver burden<br />

in FTD is much greater than in AD. 128–130 <strong>Behavioural</strong><br />

changes rather than level of disability seem to be<br />

correlated with caregiver distress and burden in bvFTD, 129<br />

although very few studies have been done. Evidence<br />

indicates that caregiver health is a major contributor to<br />

carer stress, with depression accounting for 58% of the<br />

variance of stress scores in FTD caregivers. 130 The key to<br />

reducing caregiver stress seems to lie in increasing their<br />

understanding of the symptoms and ways of dealing with<br />

challenging behaviours.<br />

Driving and other aspects of capacity in bvFTD are<br />

clearly of great practical importance, but have been<br />

subject to little systematic research. A single study of<br />

driving competence in bvFTD suggested signifi cant<br />

problems, even at an early stage of the disease. 111 On that<br />

basis, a driving assessment would seem to be<br />

recommended in all cases. In Australia, the UK, and<br />

many other countries, an established code of practice<br />

exists governing the issue of legal capacity associated<br />

with the relevant Mental Capacity Act. Its application is<br />

relatively straightforward in patients with advanced<br />

bvFTD, but establishing a lack of capacity could be<br />

complex in the early stages of the disease. Of note,<br />

Basic Instrumental<br />

Mild Lack of aff ection No changes Diffi culties in managing correspondence<br />

Diffi culties in planning of fi nances<br />

Moderate Confused in unusual places<br />

Agitated, restless<br />

“Sweet tooth”<br />

Uncooperative<br />

Confused with the date<br />

Eats the same foods<br />

Impulsive<br />

Inability to choose clothes adequate to the<br />

occasion<br />

Severe .. Lack of appropriate manners at mealtimes<br />

Lack of initiation to eat if not served<br />

Very severe .. Cannot stay at home alone<br />

Not using cutlery adequately<br />

Does not go to toilet in time (urine)<br />

Profound .. Bedridden ..<br />

Modifi ed from Mioshi et al, 115 by permission of Wolters Kluwer Health. bvFTD=behavioural-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Table: Characteristics of each severity stage in the disease progression of bvFTD<br />

Changes in driving ability<br />

Decline in standard of execution of household chores<br />

Diffi culties dialling telephone<br />

Lacks interest in fi nances<br />

Cannot plan meals as well as before<br />

Problems with shopping<br />

Needs supervision when preparing a meal<br />

Needs prompting to do household chores<br />

Needs prompting to prepare meal<br />

Needs prompting to take medications<br />

Needs prompting to do leisure activities<br />

Needs help with dosage of medications<br />

Diffi culties managing cash<br />

..<br />

168 www.thelancet.com/neurology Vol 10 February 2011


Search strategy and selection criteria<br />

References for this Review were identifi ed through searches<br />

of PubMed from 2000, until November, 2010, with the terms<br />

“<strong>frontotemporal</strong> <strong>dementia</strong>”, “<strong>frontotemporal</strong> lobar<br />

degeneration”, “behavioural-<strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong>”, and “frontal-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>”.<br />

Articles were also identifi ed through searches of the authors’<br />

own fi les. Only papers published in English were reviewed.<br />

capacity is not a global phenomenon but is situation<br />

specifi c. Moreover, unlike in AD, in which capacity can<br />

be impaired by amnesia (which is easier to assess), in<br />

bvFTD, the key cognitive variables are insight and<br />

judgment, which are much harder to assess and quantify.<br />

Finally, many useful publications and DVDs are available<br />

for caregivers, which can be accessed through the<br />

Frontotemporal Dementia Research Group website or<br />

the Association for FTDs.<br />

Conclusions and future directions<br />

Knowledge of the <strong>clinical</strong> presentation of bvFTD and its<br />

pathological processes has improved substantially over<br />

the past 20 years. Clinicians have become more aware of<br />

this disabling neurodegenerative condition, which aff ects<br />

individuals who are often still in the workforce or have<br />

young children. Careful medical history and information<br />

from family members, combined with <strong>clinical</strong><br />

investigations, neuro psychological testing, and<br />

investigations of social cognition, have increased case<br />

identifi cation. Sensitivity has also improved with the use<br />

of advanced structural and functional neuroimaging<br />

techniques. However, the major challenge that remains is<br />

to improve the prediction of the underlying neuropathology<br />

in patients with bvFTD during life. Eff orts to identify<br />

potential disease biomarkers for the disease are promising<br />

but will require further investigations. This line of<br />

research will become particularly relevant as diseasemodifying<br />

agents are developed.<br />

Contributors<br />

All authors contributed equally to the conception, literature review, and<br />

writing of this Review.<br />

Confl icts of interest<br />

The authors declare that they have no confl icts of interest.<br />

Acknowledgments<br />

This project was supported in part by a National Health and Medical<br />

Research Council (NHMRC) of Australia project grant (#510106). OP is<br />

supported by an NHMRC <strong>clinical</strong> career development award<br />

fellowship (#510184). JRH is supported by an Australian Research<br />

Council Federation fellowship (#FF0776229) and was previously<br />

supported by the UK Medical Research Council.<br />

References<br />

1 Hodges JR. Overview of <strong>frontotemporal</strong> <strong>dementia</strong>. In: Hodges JR,<br />

ed. Frontotemporal <strong>dementia</strong> syndromes. Cambridge: Cambridge<br />

University Press, 2007: 1–24.<br />

2 Ratnavalli E, Brayne C, Dawson K, Hodges JR. The prevalence of<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2002; 58: 1615–21.<br />

3 Rosso SM, Donker Kaat L, Baks T, et al. Frontotemporal <strong>dementia</strong><br />

in The Netherlands: patient characteristics and prevalence<br />

estimates from a population-based study. Brain 2003;<br />

126: 2016–22.<br />

4 Hodges JR, Patterson K. Semantic <strong>dementia</strong>: a unique<br />

clinicopathological syndrome. Lancet Neurol 2007; 6: 1004–14.<br />

5 Neary D, Snowden JS, Gustafson L, et al. Frontotemporal lobar<br />

degeneration: a consensus on <strong>clinical</strong> diagnostic criteria. Neurology<br />

1998; 51: 1546–54.<br />

6 Grossman M. Primary progressive aphasia: clinicopathological<br />

correlations. Nat Rev Neurol 2010; 6: 88–97.<br />

7 Lillo P, Garcin B, Hornberger M, Bak TH, Hodges JR.<br />

Neurobehavioral features in <strong>frontotemporal</strong> <strong>dementia</strong> with<br />

amyotrophic lateral sclerosis. Arch Neurol 2010; 67: 826–30.<br />

8 Lomen-Hoerth C, Anderson T, Miller B. The overlap of amyotrophic<br />

lateral sclerosis and <strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2002;<br />

59: 1077–79.<br />

9 Lillo P, Mioshi E, Zoing MC, Kiernan MC, Hodges JR. How<br />

common are behavioural changes in amyotrophic lateral sclerosis?<br />

Amyotroph Lateral Scler 2010; published online Sept 19.<br />

DOI: 10.3109/17482968.2010.520718.<br />

10 Kertesz A, McMonagle P, Blair M, Davidson W, Munoz DG. The<br />

evolution and pathology of <strong>frontotemporal</strong> <strong>dementia</strong>. Brain 2005;<br />

128: 1996–2005.<br />

11 Rohrer JD, Guerreiro R, Vandrovcova J, et al. The heritability and<br />

genetics of <strong>frontotemporal</strong> lobar degeneration. Neurology 2009;<br />

73: 1451–56.<br />

12 Cairns NJ, Bigio EH, Mackenzie IR, et al. Neuropathologic<br />

diagnostic and nosologic criteria for <strong>frontotemporal</strong> lobar<br />

degeneration: consensus of the Consortium for Frontotemporal<br />

Lobar Degeneration. Acta Neuropathol (Berl) 2007; 114: 5–22.<br />

13 Broe M, Hodges JR, Schofi eld E, Shepherd CE, Kril JJ,<br />

Halliday GM. Staging disease severity in pathologically confi rmed<br />

cases of <strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2003; 60: 1005–11.<br />

14 Kril JJ, Macdonald V, Patel S, Png F, Halliday GM. Distribution of<br />

brain atrophy in behavioral <strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

J Neurol Sci 2005; 232: 83–90.<br />

15 Kersaitis C, Halliday GM, Kril JJ. Regional and cellular pathology in<br />

<strong>frontotemporal</strong> <strong>dementia</strong>: relationship to stage of disease in cases<br />

with and without Pick bodies. Acta Neuropathol 2004; 108: 515–23.<br />

16 Mackenzie IR, Neumann M, Bigio EH, et al. Nomenclature and<br />

nosology for neuropathologic subtypes of <strong>frontotemporal</strong> lobar<br />

degeneration: an update. Acta Neuropathol 2010; 119: 1–4.<br />

17 Mackenzie IR, Foti D, Woulfe J, Hurwitz TA. Atypical<br />

<strong>frontotemporal</strong> lobar degeneration with ubiquitin-positive,<br />

TDP-43-negative neuronal inclusions. Brain 2008; 131: 1282–93.<br />

18 Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA,<br />

Mackenzie IR. A new subtype of <strong>frontotemporal</strong> lobar degeneration<br />

with FUS pathology. Brain 2009; 132: 2922–31.<br />

19 Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, et al. Mutations in the<br />

FUS/TLS gene on chromosome 16 cause familial amyotrophic<br />

lateral sclerosis. Science 2009; 323: 1205–08.<br />

20 Vance C, Rogelj B, Hortobagyi T, et al. Mutations in FUS, an RNA<br />

processing protein, cause familial amyotrophic lateral sclerosis<br />

type 6. Science 2009; 323: 1208–11.<br />

21 Hodges JR, Davies RR, Xuereb JH, et al. Clinicopathological<br />

correlates in <strong>frontotemporal</strong> <strong>dementia</strong>. Ann Neurol 2004; 56: 399–406.<br />

22 Shi J, Shaw CL, Du Plessis D, et al. Histopathological changes<br />

underlying <strong>frontotemporal</strong> lobar degeneration with<br />

clinicopathological correlation. Acta Neuropathol 2005; 110: 501–12.<br />

23 Snowden J, Neary D, Mann D. Frontotemporal lobar degeneration:<br />

<strong>clinical</strong> and pathological relationships. Acta Neuropathol 2007;<br />

114: 31–38.<br />

24 Seelaar H, Klijnsma KY, de Koning I, et al. Frequency of ubiquitin<br />

and FUS-positive, TDP-43-negative <strong>frontotemporal</strong> lobar<br />

degeneration. J Neurol 2010; 257: 747–53.<br />

25 Bian H, Van Swieten JC, Leight S, et al. CSF biomarkers in<br />

<strong>frontotemporal</strong> lobar degeneration with known pathology. Neurology<br />

2008; 70: 1827–35.<br />

26 Steinacker P, Hendrich C, Sperfeld AD, et al. TDP-43 in<br />

cerebrospinal fl uid of patients with <strong>frontotemporal</strong> lobar<br />

degeneration and amyotrophic lateral sclerosis. Arch Neurol 2008;<br />

65: 1481–87.<br />

Review<br />

For the Frontotemporal<br />

Dementia Research Group see<br />

http://www.ftdrg.org<br />

For the Association for FTDs see<br />

http://www.ftd-picks.org<br />

www.thelancet.com/neurology Vol 10 February 2011 169


Review<br />

27 Sleegers K, Brouwers N, Van Damme P, et al. Serum biomarker for<br />

progranulin-associated <strong>frontotemporal</strong> lobar degeneration.<br />

Ann Neurol 2009; 65: 603–09.<br />

28 Finch N, Baker M, Crook R, et al. Plasma progranulin levels predict<br />

progranulin mutation status in <strong>frontotemporal</strong> <strong>dementia</strong> patients<br />

and asymptomatic family members. Brain 2009; 132: 583–91.<br />

29 Ghidoni R, Benussi L, Glionna M, Franzoni M, Binetti G. Low<br />

plasma progranulin levels predict progranulin mutations in<br />

<strong>frontotemporal</strong> lobar degeneration. Neurology 2008; 71: 1235–39.<br />

30 Schofi eld E, Halliday GM, Kwok JB, Loy C, Hodges JR. Low serum<br />

progranulin is diagnostic for progranulin mutations: a prospective<br />

study. J Alzheimers Dis 2010; published online Sept 21.<br />

DOI: 10.3233/JAD-2010-101032.<br />

31 Foulds PG, Davidson Y, Mishra M, et al. Plasma phosphorylated-<br />

TDP-43 protein levels correlate with brain pathology in<br />

<strong>frontotemporal</strong> lobar degeneration. Acta Neuropathol 2009;<br />

118: 647–58.<br />

32 Benajiba L, Le Ber I, Camuzat A, et al. TARDBP mutations in<br />

motoneuron disease with <strong>frontotemporal</strong> lobar degeneration.<br />

Ann Neurol 2009; 65: 470–73.<br />

33 Ticozzi N, Silani V, LeClerc AL, et al. Analysis of FUS gene<br />

mutation in familial amyotrophic lateral sclerosis within an Italian<br />

cohort. Neurology 2009; 73: 1180–85.<br />

34 Blair IP, Williams KL, Warraich ST, et al. FUS mutations in<br />

amyotrophic lateral sclerosis: <strong>clinical</strong>, pathological,<br />

neurophysiological and genetic analysis.<br />

J Neurol Neurosurg Psychiatry 2010; 81: 639–45.<br />

35 Kovacs GG, Murrell JR, Horvath S, et al. TARDBP variation<br />

associated with <strong>frontotemporal</strong> <strong>dementia</strong>, supranuclear gaze palsy,<br />

and chorea. Mov Disord 2009; 24: 1843–47.<br />

36 Watts GD, Wymer J, Kovach MJ, et al. Inclusion body myopathy<br />

associated with Paget disease of bone and <strong>frontotemporal</strong> <strong>dementia</strong><br />

is caused by mutant valosin-containing protein. Nat Genet 2004;<br />

36: 377–81.<br />

37 Parkinson N, Ince PG, Smith MO, et al. ALS phenotypes with<br />

mutations in CHMP2B (charged multivesicular body protein 2B).<br />

Neurology 2006; 67: 1074–77.<br />

38 van der Zee J, Urwin H, Engelborghs S, et al.<br />

CHMP2B C-truncating mutations in <strong>frontotemporal</strong> lobar<br />

degeneration are associated with an aberrant endosomal phenotype<br />

in vitro. Hum Mol Genet 2008; 17: 313–22.<br />

39 Vance C, Al-Chalabi A, Ruddy D, et al. Familial amyotrophic lateral<br />

sclerosis with <strong>frontotemporal</strong> <strong>dementia</strong> is linked to a locus on<br />

chromosome 9p13.2-21.3. Brain 2006; 129: 868–76.<br />

40 Kipps CM, Mioshi E, Hodges JR. Emotion, social functioning and<br />

activities of daily living in <strong>frontotemporal</strong> <strong>dementia</strong>. Neurocase 2009;<br />

15: 182–89.<br />

41 Huey ED, Goveia EN, Paviol S, et al. Executive dysfunction in<br />

<strong>frontotemporal</strong> <strong>dementia</strong> and corticobasal syndrome. Neurology<br />

2009; 72: 453–59.<br />

42 Seeley WW, Crawford R, Rascovsky K, et al. Frontal paralimbic<br />

network atrophy in very mild behavioral <strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong>. Arch Neurol 2008; 65: 249–55.<br />

43 Seeley WW. Selective functional, regional, and neuronal vulnerability<br />

in <strong>frontotemporal</strong> <strong>dementia</strong>. Curr Opin Neurol 2008; 21: 701–07.<br />

44 Cummings JL, Mega M, Gray K, Rosenberg-Thompson S,<br />

Carusi DA, Gornbein J. The Neuropsychiatric Inventory:<br />

comprehensive assessment of psychopathology in <strong>dementia</strong>.<br />

Neurology 1994; 44: 2308–14.<br />

45 Bozeat S, Gregory CA, Ralph MA, Hodges JR. Which<br />

neuropsychiatric and behavioural features distinguish frontal and<br />

temporal <strong>variant</strong>s of <strong>frontotemporal</strong> <strong>dementia</strong> from Alzheimer’s<br />

disease? J Neurol Neurosurg Psychiatry 2000; 69: 178–86.<br />

46 Kertesz A, Davidson W, Fox H. Frontal behavioral inventory:<br />

diagnostic criteria for frontal lobe <strong>dementia</strong>. Can J Neurol Sci 1997;<br />

24: 29–36.<br />

47 Manes FF, Torralva T, Roca M, Gleichgerrcht E, Bekinschtein TA,<br />

Hodges JR. Frontotemporal <strong>dementia</strong> presenting as pathological<br />

gambling. Nat Rev Neurol 2010; 6: 347–52.<br />

48 Postiglione A, Milan G, Pappata S, et al. Fronto-temporal <strong>dementia</strong><br />

presenting as Geschwind’s syndrome. Neurocase 2008; 14: 264–70.<br />

49 Shinagawa S, Ikeda M, Nestor PJ, et al. Characteristics of abnormal<br />

eating behaviours in <strong>frontotemporal</strong> lobar degeneration: a crosscultural<br />

survey. J Neurol Neurosurg Psychiatry 2009; 80: 1413–14.<br />

50 Piguet O, Petersén Å, Lam BYK, et al. Eating and hypothalamus<br />

changes in behavioural-<strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Ann Neurol 2010; published online Nov 12.<br />

DOI: 10.1002/ana.22244.<br />

51 Loy CT, Kril JJ, Trollor JN, et al. The case of a 48 year-old woman<br />

with bizarre and complex delusions. Nat Rev Neurol 2010; 6: 175–79.<br />

52 Urwin H, Josephs KA, Rohrer JD, et al. FUS pathology defi nes the<br />

majority of tau- and TDP-43-negative <strong>frontotemporal</strong> lobar<br />

degeneration. Acta Neuropathol 2010; 120: 33–41.<br />

53 Liu W, Miller BL, Kramer JH, et al. Behavioral disorders in the<br />

frontal and temporal <strong>variant</strong>s of <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Neurology 2004; 62: 742–48.<br />

54 Diehl-Schmid J, Pohl C, Perneczky R, Forstl H, Kurz A. Behavioral<br />

disturbances in the course of <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Dement Geriatr Cogn Disord 2006; 22: 352–57.<br />

55 Srikanth S, Nagaraja AV, Ratnavalli E. Neuropsychiatric symptoms<br />

in <strong>dementia</strong>-frequency, relationship to <strong>dementia</strong> severity and<br />

comparison in Alzheimer’s disease, vascular <strong>dementia</strong> and<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. J Neurol Sci 2005; 236: 43–48.<br />

56 Pasquier F, Lebert F, Lavenu I, Guillaume B. The <strong>clinical</strong> picture of<br />

<strong>frontotemporal</strong> <strong>dementia</strong>: <strong>diagnosis</strong> and follow-up.<br />

Dement Geriatr Cogn Disord 1999; 10 (suppl 1): 10–14.<br />

57 Shinagawa S, Toyota Y, Ishikawa T, et al. Cognitive function and<br />

psychiatric symptoms in early- and late-onset <strong>frontotemporal</strong><br />

<strong>dementia</strong>. Dement Geriatr Cogn Disord 2008; 25: 439–44.<br />

58 Borroni B, Agosti C, Bellelli G, Padovani A. Is early-onset <strong>clinical</strong>ly<br />

diff erent from late-onset <strong>frontotemporal</strong> <strong>dementia</strong>? Eur J Neurol<br />

2008; 15: 1412–15.<br />

59 Kipps CM, Nestor PJ, Fryer TD, Hodges JR. <strong>Behavioural</strong> <strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong>: not all it seems? Neurocase 2007;<br />

13: 237–47.<br />

60 Hornberger M, Shelley BP, Kipps CM, Piguet O, Hodges JR. Can<br />

progressive and non-progressive behavioral <strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong> be distinguished at presentation?<br />

J Neurol Neurosurg Psychiatry 2009; 80: 591–93.<br />

61 Davies RR, Kipps CM, Mitchell J, Kril JJ, Halliday GM, Hodges JR.<br />

Progression in <strong>frontotemporal</strong> <strong>dementia</strong>: identifying a benign<br />

behavioral <strong>variant</strong> by magnetic resonance imaging. Arch Neurol<br />

2006; 63: 1627–31.<br />

62 Piguet O, Hornberger M, Shelley BP, Kipps CM, Hodges JR.<br />

Sensitivity of current criteria for the <strong>diagnosis</strong> of behavioral <strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2009; 72: 732–37.<br />

63 Kipps CM, Hodges JR, Fryer TD, Nestor PJ. Combined magnetic<br />

resonance imaging and positron emission tomography brain<br />

imaging in behavioural <strong>variant</strong> <strong>frontotemporal</strong> degeneration:<br />

refi ning the <strong>clinical</strong> phenotype. Brain 2009; 132: 2566–78.<br />

64 Rascovsky K, Hodges JR, Kipps CM, et al. Diagnostic criteria for the<br />

behavioral <strong>variant</strong> of <strong>frontotemporal</strong> <strong>dementia</strong> (bvFTD): current<br />

limitations and future directions. Alzheimer Dis Assoc Disord 2007;<br />

21: S14–18.<br />

65 Mioshi E, Kipps CM, Dawson K, Mitchell J, Graham A, Hodges JR.<br />

Activities of daily living in <strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer<br />

disease. Neurology 2007; 68: 2077–84.<br />

66 Kipps CM, Nestor PJ, Dawson CE, Mitchell J, Hodges JR.<br />

Measuring progression in <strong>frontotemporal</strong> <strong>dementia</strong>: implications<br />

for therapeutic interventions. Neurology 2008; 70: 2046–52.<br />

67 Giovagnoli AR, Erbetta A, Reati F, Bugiani O. Diff erential<br />

neuropsychological patterns of frontal <strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong> and Alzheimer’s disease in a study of diagnostic<br />

concordance. Neuropsychologia 2008; 46: 1495–504.<br />

68 Hornberger M, Piguet O, Kipps C, Hodges JR. Executive function<br />

in progressive and nonprogressive behavioral <strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2008; 71: 1481–88.<br />

69 Gleichgerrcht E, Torralva T, Roca M, Manes F. Utility of an<br />

abbreviated version of the executive and social cognition battery in<br />

the detection of executive defi cits in early behavioral <strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong> patients. J Int Neuropsychol Soc 2010;<br />

19: 1–8.<br />

70 Graham A, Davies R, Xuereb J, et al. Pathologically proven<br />

<strong>frontotemporal</strong> <strong>dementia</strong> presenting with severe amnesia. Brain<br />

2005; 128: 597–605.<br />

71 Hornberger M, Piguet O, Graham AJ, Nestor PJ, Hodges JR. How<br />

preserved is episodic memory in behavioral <strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong>? Neurology 2010; 74: 472–79.<br />

170 www.thelancet.com/neurology Vol 10 February 2011


72 Fellows LK, Farah MJ. Ventromedial frontal cortex mediates<br />

aff ective shifting in humans: evidence from a reversal learning<br />

paradigm. Brain 2003; 126: 1830–37.<br />

73 Bechara A, Damasio AR, Damasio H, Anderson SW. Insensitivity to<br />

future consequences following damage to human prefrontal cortex.<br />

Cognition 1994; 50: 7–15.<br />

74 Torralva T, Roca M, Gleichgerrcht E, Bekinschtein T, Manes F. A<br />

neuropsychological battery to detect specifi c executive and social<br />

cognitive impairments in early <strong>frontotemporal</strong> <strong>dementia</strong>. Brain<br />

2009; 132: 1299–309.<br />

75 Lough S, Kipps CM, Treise C, Watson P, Blair JR, Hodges JR. Social<br />

reasoning, emotion and empathy in <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Neuropsychologia 2006; 44: 950–58.<br />

76 Snowden JS, Austin NA, Sembi S, Thompson JC, Craufurd D,<br />

Neary D. Emotion recognition in Huntington’s disease and<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neuropsychologia 2008; 46: 2638–49.<br />

77 Fernandez-Duque D, Black SE. Impaired recognition of negative<br />

facial emotions in patients with <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Neuropsychologia 2005; 43: 1673–87.<br />

78 Rankin KP, Gorno-Tempini ML, Allison SC, et al. Structural<br />

anatomy of empathy in neurodegenerative disease. Brain 2006;<br />

129: 2945–56.<br />

79 Werner KH, Roberts NA, Rosen HJ, et al. Emotional reactivity and<br />

emotion recognition in <strong>frontotemporal</strong> lobar degeneration.<br />

Neurology 2007; 69: 148–55.<br />

80 Sturm VE, Rosen HJ, Allison S, Miller BL, Levenson RW. Selfconscious<br />

emotion defi cits in <strong>frontotemporal</strong> lobar degeneration.<br />

Brain 2006; 129: 2508–16.<br />

81 Fernandez-Duque D, Hodges SD, Baird JA, Black SE. Empathy in<br />

<strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer’s disease.<br />

J Clin Exp Neuropsychol 2010; 32: 289–98.<br />

82 Gregory C, Lough S, Stone V, et al. Theory of mind in patients<br />

with frontal <strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer’s<br />

disease: theoretical and practical implications. Brain 2002;<br />

125: 752–64.<br />

83 Mendez MF, Shapira JS. Altered emotional morality in<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Cogn Neuropsychiatry 2009; 14: 165–79.<br />

84 Kipps CM, Nestor PJ, Acosta-Cabronero J, Arnold R, Hodges JR.<br />

Understanding social dysfunction in the behavioural <strong>variant</strong> of<br />

<strong>frontotemporal</strong> <strong>dementia</strong>: the role of emotion and sarcasm<br />

processing. Brain 2009; 132: 592–603.<br />

85 McDonald S, Flanagan S, Rollins J, Kinch J. TASIT: a new <strong>clinical</strong><br />

tool for assessing social perception after traumatic brain injury.<br />

J Head Trauma Rehabil 2003; 18: 219–38.<br />

86 Davatzikos C, Resnick SM, Wu X, Parmpi P, Clark CM. Individual<br />

patient <strong>diagnosis</strong> of AD and FTD via high-dimensional pattern<br />

classifi cation of MRI. Neuroimage 2008; 41: 1220–27.<br />

87 Hornberger M, Savage S, Hsieh S, Mioshi E, Piguet O, Hodges JR.<br />

Orbitofrontal dysfunction discriminates behavioural <strong>variant</strong><br />

<strong>frontotemporal</strong> <strong>dementia</strong> from Alzheimer’s disease.<br />

Dement Geriatr Cogn Disord 2011 (in press).<br />

88 Whitwell JL, Przybelski SA, Weigand SD, et al. Distinct anatomical<br />

subtypes of the behavioural <strong>variant</strong> of <strong>frontotemporal</strong> <strong>dementia</strong>: a<br />

cluster analysis study. Brain 2009; 132: 2932–46.<br />

89 Kipps CM, Davies RR, Mitchell J, Kril JJ, Halliday GM, Hodges JR.<br />

Clinical signifi cance of lobar atrophy in <strong>frontotemporal</strong> <strong>dementia</strong>:<br />

application of an MRI visual rating scale.<br />

Dement Geriatr Cogn Disord 2007; 23: 334–42.<br />

90 Boccardi M, Sabattoli F, Laakso MP, et al. Frontotemporal<br />

<strong>dementia</strong> as a neural system disease. Neurobiol Aging 2005;<br />

26: 37–44.<br />

91 Schroeter ML, Raczka K, Neumann J, Yves von Cramon D. Towards<br />

a nosology for <strong>frontotemporal</strong> lobar degenerations—a meta-analysis<br />

involving 267 subjects. NeuroImage 2007; 36: 497–510.<br />

92 Davies RR, Scahill VL, Graham A, Williams GB, Graham KS,<br />

Hodges JR. Development of an MRI rating scale for multiple brain<br />

regions: comparison with volumetrics and with voxel-based<br />

morphometry. Neuroradiology 2009; 51: 491–503.<br />

93 Seeley WW, Carlin DA, Allman JM, et al. Early <strong>frontotemporal</strong><br />

<strong>dementia</strong> targets neurons unique to apes and humans. Ann Neurol<br />

2006; 60: 660–67.<br />

94 Seeley WW, Crawford RK, Zhou J, Miller BL, Greicius MD.<br />

Neurodegenerative diseases target large-scale human brain<br />

networks. Neuron 2009; 62: 42–52.<br />

95 Zhou J, Greicius MD, Gennatas ED, et al. Divergent network<br />

connectivity changes in behavioural <strong>variant</strong> <strong>frontotemporal</strong><br />

<strong>dementia</strong> and Alzheimer’s disease. Brain 2010; 133: 1352–67.<br />

96 Whitwell JL, Josephs KA, Rossor MN, et al. Magnetic resonance<br />

imaging signatures of tissue pathology in <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Arch Neurol 2005; 62: 1402–08.<br />

97 Josephs KA, Whitwell JL, Parisi JE, et al. Caudate atrophy on MRI is<br />

a characteristic feature of FTLD-FUS. Eur J Neurol 2010; 17: 969–75.<br />

98 Pereira JM, Williams GB, Acosta-Cabronero J, et al. Atrophy<br />

patterns in histologic vs <strong>clinical</strong> groupings of <strong>frontotemporal</strong> lobar<br />

degeneration. Neurology 2009; 72: 1653–60.<br />

99 Barnes J, Whitwell JL, Frost C, Josephs KA, Rossor M, Fox NC.<br />

Measurements of the amygdala and hippocampus in pathologically<br />

confi rmed Alzheimer disease and <strong>frontotemporal</strong> lobar<br />

degeneration. Arch Neurol 2006; 63: 1434–39.<br />

100 Cardenas VA, Boxer AL, Chao LL, et al. Deformation-based<br />

morphometry reveals brain atrophy in <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Arch Neurol 2007; 64: 873–77.<br />

101 Chao LL, Schuff N, Clevenger EM, et al. Patterns of white matter<br />

atrophy in <strong>frontotemporal</strong> lobar degeneration. Arch Neurol 2007;<br />

64: 1619–24.<br />

102 Zhang Y, Schuff N, Du AT, et al. White matter damage in<br />

<strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer’s disease measured by<br />

diff usion MRI. Brain 2009; 132: 2579–92.<br />

103 Chan D, Fox NC, Jenkins R, Scahill RI, Crum WR, Rossor MN.<br />

Rates of global and regional cerebral atrophy in AD and<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2001; 57: 1756–63.<br />

104 Varma AR, Adams W, Lloyd JJ, et al. Diagnostic patterns of regional<br />

atrophy on MRI and regional cerebral blood fl ow change on SPECT<br />

in young onset patients with Alzheimer’s disease, <strong>frontotemporal</strong><br />

<strong>dementia</strong> and vascular <strong>dementia</strong>. Acta Neurol Scand 2002;<br />

105: 261–69.<br />

105 Kanda T, Ishii K, Uemura T, et al. Comparison of grey matter and<br />

metabolic reductions in <strong>frontotemporal</strong> <strong>dementia</strong> using FDG-PET<br />

and voxel-based morphometric MR studies.<br />

Eur J Nucl Med Mol Imaging 2008; 35: 2227–34.<br />

106 Rowe CC, Ng S, Ackermann U, et al. Imaging beta-amyloid burden<br />

in aging and <strong>dementia</strong>. Neurology 2007; 68: 1718–25.<br />

107 Engler H, Santillo AF, Wang SX, et al. In vivo amyloid imaging with<br />

PET in <strong>frontotemporal</strong> <strong>dementia</strong>. Eur J Nucl Med Mol Imaging 2008;<br />

35: 100–06.<br />

108 Wicklund AH, Johnson N, Rademaker A, Weitner BB, Weintraub S.<br />

Profi les of decline in activities of daily living in non-Alzheimer<br />

<strong>dementia</strong>. Alzheimer Dis Assoc Disord 2007; 21: 8–13.<br />

109 Rosen HJ, Allison SC, Schauer GF, Gorno-Tempini ML,<br />

Weiner MW, Miller BL. Neuroanatomical correlates of behavioural<br />

disorders in <strong>dementia</strong>. Brain 2005; 128: 2612–25.<br />

110 Rascovsky K, Salmon DP, Lipton AM, et al. Rate of progression<br />

diff ers in <strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer disease.<br />

Neurology 2005; 65: 397–403.<br />

111 de Simone V, Kaplan L, Patronas N, Wassermann EM, Grafman J.<br />

Driving abilities in <strong>frontotemporal</strong> <strong>dementia</strong> patients.<br />

Dement Geriatr Cogn Disord 2007; 23: 1–7.<br />

112 Mioshi E, Hodges JR. Rate of change of functional abilities in<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Dement Geriatr Cogn Disord 2009;<br />

28: 419–26.<br />

113 Morris JC. Clinical <strong>dementia</strong> rating: a reliable and valid diagnostic<br />

and staging measure for <strong>dementia</strong> of the Alzheimer type.<br />

Int Psychogeriatr 1997; 9 (suppl 1): 173–78.<br />

114 Knopman DS, Kramer JH, Boeve BF, et al. Development of<br />

methodology for conducting <strong>clinical</strong> trials in <strong>frontotemporal</strong> lobar<br />

degeneration. Brain 2008; 131: 2957–68.<br />

115 Mioshi E, Hsieh S, Savage S, Hornberger M, Hodges JR. Clinical<br />

staging and disease progression in <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Neurology 2010; 74: 1591–97.<br />

116 Roberson ED, Hesse JH, Rose KD, et al. Frontotemporal <strong>dementia</strong><br />

progresses to death faster than Alzheimer disease. Neurology 2005;<br />

65: 719–25.<br />

117 Garcin B, Lillo P, Hornberger M, et al. Determinants of survival in<br />

behavioral <strong>variant</strong> <strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2009;<br />

73: 1656–61.<br />

118 Pasquier F, Richard F, Lebert F. Natural history of <strong>frontotemporal</strong><br />

<strong>dementia</strong>: comparison with Alzheimer’s disease.<br />

Dement Geriatr Cogn Disord 2004; 17: 253–57.<br />

Review<br />

www.thelancet.com/neurology Vol 10 February 2011 171


Review<br />

119 Hodges JR, Davies R, Xuereb J, Kril J, Halliday G. Survival in<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Neurology 2003; 61: 349–54.<br />

120 Boxer AL, Boeve BF. Frontotemporal <strong>dementia</strong> treatment: current<br />

symptomatic therapies and implications of recent genetic,<br />

biochemical, and neuroimaging studies. Alzheimer Dis Assoc Disord<br />

2007; 21: S79–87.<br />

121 Moretti R, Torre P, Antonello RM, Cazzato G, Bava A.<br />

Frontotemporal <strong>dementia</strong>: paroxetine as a possible treatment of<br />

behavior symptoms. A randomized, controlled, open 14-month<br />

study. Eur Neurol 2003; 49: 13–19.<br />

122 Deakin JB, Rahman S, Nestor PJ, Hodges JR, Sahakian BJ.<br />

Paroxetine does not improve symptoms and impairs cognition in<br />

<strong>frontotemporal</strong> <strong>dementia</strong>: a double-blind randomized controlled<br />

trial. Psychopharmacology (Berl) 2004; 172: 400–08.<br />

123 Moretti R, Torre P, Antonello RM, Cazzato G, Griggio S, Bava A.<br />

Olanzapine as a treatment of neuropsychiatric disorders of<br />

Alzheimer’s disease and other <strong>dementia</strong>s: a 24-month follow-up of<br />

68 patients. Am J Alzheimers Dis Other Demen 2003; 18: 205–14.<br />

124 Moretti R, Torre P, Antonello RM, Cattaruzza T, Cazzato G, Bava A.<br />

Rivastigmine in <strong>frontotemporal</strong> <strong>dementia</strong>: an open-label study.<br />

Drugs Aging 2004; 21: 931–97.<br />

125 Mendez MF, Shapira JS, McMurtray A, Licht E. Preliminary<br />

fi ndings: behavioral worsening on donepezil in patients with<br />

<strong>frontotemporal</strong> <strong>dementia</strong>. Am J Geriatr Psychiatry 2007; 15: 84–87.<br />

126 Mendez MF. Frontotemporal <strong>dementia</strong>: therapeutic interventions.<br />

Front Neurol Neurosci 2009; 24: 168–78.<br />

127 Merrilees J, Klapper J, Murphy J, Lomen-Hoerth C, Miller BL.<br />

Cognitive and behavioral challenges in caring for patients with<br />

<strong>frontotemporal</strong> <strong>dementia</strong> and amyotrophic lateral sclerosis.<br />

Amyotroph Lateral Scler 2010; 11: 298–302.<br />

128 Riedijk SR, De Vugt ME, Duivenvoorden HJ, et al. Caregiver<br />

burden, health-related quality of life and coping in <strong>dementia</strong><br />

caregivers: a comparison of <strong>frontotemporal</strong> <strong>dementia</strong> and<br />

Alzheimer’s disease. Dement Geriatr Cogn Disord 2006; 22: 405–12.<br />

129 Boutoleau-Bretonniere C, Vercelletto M, Volteau C, Renou P,<br />

Lamy E. Zarit burden inventory and activities of daily living in the<br />

behavioral <strong>variant</strong> of <strong>frontotemporal</strong> <strong>dementia</strong>.<br />

Dement Geriatr Cogn Disord 2008; 25: 272–77.<br />

130 Mioshi E, Bristow M, Cook R, Hodges JR. Factors underlying<br />

caregiver stress in <strong>frontotemporal</strong> <strong>dementia</strong> and Alzheimer’s<br />

disease. Dement Geriatr Cogn Disord 2009; 27: 76–81.<br />

172 www.thelancet.com/neurology Vol 10 February 2011

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!