10.1016@B978 0 444 64012 3.00003 4

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Handbook of Clinical Neurology, Vol.

165 (3rd series)


Psychopharmacology of Neurologic Disease
V.I. Reus and D. Lindqvist, Editors
https://doi.org/10.1016/B978-0-444-64012-3.00003-4
Copyright © 2019 Elsevier B.V. All rights reserved

Chapter 3

Frontotemporal dementia
BRUCE MILLER1* AND JORGE J. LLIBRE GUERRA2,3
1
Memory and Aging Center and Global Brain Health Institute, University of California, San Francisco, CA, United States
2
Cognitive and Behavior Research Unit, National Institute of Neurology and Neurosurgery, Havana, Cuba
3
Global Brain Health Institute, University of California, San Francisco, CA, United States

Abstract
Recent research reveals an overlap between frontotemporal dementia (FTD) and a variety of primary psy-
chiatric disorders, challenging the artificial divisions between psychiatry and neurology. This chapter
offers an overview of the clinical syndromes associated with FTD while describing links between these
syndromes and neuroimaging. This is followed by a review of the neuropathology and genetic changes in
the brain. We will illustrate the syndromic overlap that exists between FTD and several primary psychiatric
disorders including bipolar affective disorder and schizophrenia. Emphasis will be placed on the behav-
ioral variant of FTD (bvFTD), which is the common clinical syndrome seen with degeneration of the fron-
tal lobes and is the most likely to be encountered in psychiatric settings.

The frontal lobes constitute about 40% of the human (svPPA); and nonfluent variant primary progressive
cerebral cortex and are divided into three main anatomic aphasia (nfvPPA) (Lanata and Miller, 2016).
regions: primary motor cortex, premotor cortex, and The most common FTD syndrome is bvFTD, which is
prefrontal cortex. Prefrontal cortex functions remain elu- characterized by early and often severely disabling
sive, but this cortical region plays a central role in the changes in personality and behavior that carry a huge
control of personality, judgment, planning and multitask- impact on the patient, family, and society. The full-blown
ing, drive, motivation, eating and other reward-related syndrome may be preceded by a long phase of subclinical
behaviors, and social regulation. Hence, disorders that behavioral changes and social disruption, often occur-
affect this area usually overlap with primary psychiatric ring in the absence of cognitive impairment (Pressman
conditions. Studies that probe clinical disorders of the and Miller, 2014). bvFTD shares many symptoms with
prefrontal cortex using MRI, EEG, transcranial magnetic primary psychiatric disorders including schizophrenia,
stimulation, and neuropathology offer a clearer under- obsessive–compulsive disorder, borderline personality
standing of frontal lobe functions and are transforming disorder and bipolar disorder (Manoochehri and Huey,
clinical practice. 2012; Pressman and Miller, 2014).
The term frontotemporal lobar degeneration (FTLD) Prevalence studies on bvFTD and the other FTD syn-
is a neuropathologic designation used to identify a group dromes are challenging because many cases are misclas-
of neurodegenerative diseases of the frontal and anterior sified, as the disease is largely unrecognized in the
temporal lobes, associated with specific pathologies communities to which these patients present, including
(Rabinovici and Miller, 2010). The principal clinical syn- the courts, marriage counselors, addiction specialists,
dromes associated with FTLD are classified under the and human resource departments. Furthermore, the med-
rubric of FTD, an umbrella term for three clinical variants ical community is still unable to diagnose FTD in a
distinguished by early and predominant symptoms: reliable fashion. Additionally, the related neurologic con-
bvFTD; semantic variant primary progressive aphasia ditions that involve movement, progressive supranuclear

*Correspondence to: Bruce Miller, M.D., Memory and Aging Center, University of California, San Francisco, CA, United States.
Tel: +1-415-476-5591, Fax: +1-415-353-8292, E-mail: bruce.miller@ucsf.edu
34 B. MILLER AND J.J. LLIBRE GUERRA
syndrome, corticobasal syndrome, and motor neuron dis- Early apathy
ease (MND) often show FTLD pathology, yet are rarely
In bvFTD, approximately 85% of patients show early apa-
considered in epidemiologic studies (Knopman and
thy and inertia, and patients usually present with lack of
Roberts, 2011; Onyike and Diehl-Schmid, 2013). Knop-
spontaneity and motivation. This may progress to mutism
man and Roberts report that the total number of cases
and immobility in the end stages of the disease (Rascovsky
with FTLD syndromes ranges from 15 to 22 per
et al., 2011; Manoochehri and Huey, 2012; Pressman and
100,000 person and incidence rates are between 2.7
Miller, 2014). Rosen and Eslinger have related this symp-
and 4.0 per 100,000 person/years. In the United States,
tom anatomically with dysfunction of brain circuits that
at the minimum, 20,000–30,000 persons live with FTD
involve the right anterior cingulate and caudate regions
(Knopman and Roberts, 2011; Onyike and Diehl-
(Rosen et al., 2005; Eslinger et al., 2012). Sometimes, it
Schmid, 2013).
is challenging to differentiate between apathy and depres-
bvFTD cases have been reported as early as the second
sion, but the absence of sadness, hopelessness, and suicidal
decade and as late as the tenth decade, although the mean
ideas suggest that apathy is more likely than depression.
age at presentation for bvFTD is approximately 58 years.
There appears to be a slight male preponderance, although
this finding has varied between reports. It is remarkable Early loss of sympathy or empathy
that about 40% of FTD patients present with a positive Patients may lose the ability to empathize with others,
family history for dementia. This frequency of genetic eti- become insensitive to loved ones’ emotional expressions
ology is higher when we compare it with most other neu- and needs (Bartchowski et al., 2018). They become dis-
rodegenerative diseases (Riedl et al., 2014). tant, cold, and indifferent in relationships, even with close
friends and family. Often there is little or no concern for
bvFTD SYMPTOMS, ANATOMICAL the effect of their behavior on loved ones (Rankin, 2005).
CORRELATES, AND OVERLAP WITH In a voxel-based morphometry analysis, Rankin and col-
PRIMARY PSYCHIATRIC DISORDERS leagues showed that the loss of empathy correlated with
Patients with bvFTD demonstrate a gradual and progres- atrophy in the right anterior temporal and medial frontal
sive but insidious decline in social conduct, apathy,emo- regions (Rankin et al., 2006). Similarly, Rosen and col-
tional blunting, and loss of insight. Some patients present leagues have shown related impairment in emotion recog-
with a prodromal phase with marriage discourse, alienation nition with atrophy in the right lateral inferior temporal
of friends and family, poor financial decisions, problems at and right middle temporal gyri (Rosen et al., 2006).
work, new addictions, and criminal activities. Furthermore,
the overlap with primary psychiatric conditions is high, Early perseverative or compulsive behavior
leading to a diagnosis of depression, bipolar disorder, or
a midlife crisis (Woolley et al., 2011; Wald€o et al., 2015). Patients with bvFTD often display obsessive–compulsive
For the purpose of this chapter, we will review the core behaviors (71%). This varies from simple motor repetitive
symptoms of bvFTD and its anatomical correlates. movements, like foot tapping or pacing, to more complex
movements and language rituals. Repetition of stock
Early behavioral disinhibition words or phrases becomes common. Patients tend to be
ritualistic around meals without any variation in the daily
Behavioral disinhibition is one the most classic and rec- diet. Hoarding is common (Pressman and Miller, 2014).
ognized symptoms in bvFTD patients, present in nearly Some patients become more mentally rigid and resistant
76% of the patients (Rascovsky et al., 2011). Within the to changes in scheduled routines or plans (Cerami and
first years, patients tend to break social norms and rules. Cappa, 2013). Obsessive–compulsive behavior has been
As the disinhibition become more evident, patients related with networks involving orbitofrontal and anterior
become more impulsive, addiction prone, irritable, and cingulate cortexes, basal ganglia, and thalamus.
likely to commit antisocial behaviors. The disinhibition
syndrome correlates with atrophy in the orbitofrontal
Hyperorality and dietary changes
cortex (Rosen et al., 2005; Pressman and Miller, 2014).
Manoochehri and Huey (2012) note that one way dis- In bvFTD, changes in food preferences and habits are
inhibition manifests is that individuals become overly usually present. In the clinic, we have seen patients that
friendly and start conversations that are inappropriately become vegetarian or exclusively eat meat or sweets.
explicit or personal. Offensive jokes, comments, or sex- Others eat way beyond the point of satiety (Woolley
ual remarks; encroachment on the personal space of et al., 2007). Attempts to eat nondigestible objects are
others; childish behavior; and a general lack of etiquette also seen in later stages with relative frequency. The
are common. exact anatomical correlation of this symptom is not clear,
FRONTOTEMPORAL DEMENTIA 35
but Woolley has associated it with atrophy in the right bipolar disorder (Wald€o et al., 2015). For this reason,
orbitofrontal-insular-striatal brain network (Woolley misdiagnosis is seen in two directions, with some bvFTD
et al., 2007), while Piguet has correlated abnormal eating patients being initially misinterpreted as suffering from
with atrophy in the posterior hypothalamus (Piguet, a psychiatric disorder, while others with a primary
2011; Piguet et al., 2011b). psychiatric disorder being misdiagnosed as bvFTD.
Recent studies (Snowden et al., 2012; Shinagawa et al.,
Decline in executive function 2014; Wald€o et al., 2015) have pointed out the relative fre-
quency of psychotic symptoms in FTD patients, and the
In contrast to Alzheimer’s disease (AD), cognitive reports vary from 10% to 32%. These differences are
decline in bvFTD tends to be less dramatic in the early related to the instruments used for the evaluation of psy-
stages (Gregory et al., 1999). The cognitive symptoms chotic symptoms and the genetic and pathology profile
of bvFTD are mainly related to poor judgment, inatten- of the sample. A higher prevalence of psychotic symptoms
tiveness and distractibility, loss of planning ability, and in bvFTD seems to be associated with chromosome 9 open
disorganization. These patients are overly trusting and reading frame 72 (C9orf72) mutations where hallucina-
become susceptible to financial scams (Rabinovici and tions and delusions are reported as high as 50% (Sharon
Miller, 2010; Pressman and Miller, 2014). The executive et al., 2012; Snowden et al., 2012; Kertesz et al., 2013).
dysfunction correlates with atrophy in the dorsolateral Mahoney and colleagues hypothesize that thalamic
and medial prefrontal cortex (Rosen et al., 2005). and cerebellar projections could be related to the neuro-
psychiatric features associated with this mutation,
Pseudobulbar affect in FTD with MND including hallucinations and/or delusions (Takada and
Pseudobulbar affect (PBA) is a syndrome in which emo- Sha, 2012). These brain areas also seem to be particularly
tional or affective motor control becomes dysregulated as affected in C9orf72 carriers, possibly explaining the
a result of brain damage from a neurologic disease or as a higher prevalence of psychotic symptoms in patients
result of brain injury. This syndrome is often misdiag- with this genetic mutation (Mahoney et al., 2012).
nosed as a mood disorder caused by a primary psychiatric Recent studies have shown reduction of right medial pul-
disease. PBA among other causes may be seen in the vinar volume and neuron number in patients with schizo-
setting of FTD with MND (FTD-MND), which must phrenia, the same region affected in C9orf72 mutation
be distinguished from depression or other primary psy- carriers which may explain symptom overlap. Another
chiatric conditions. Laughter or crying are emotions that bvFTD subtype in which psychosis is high is FTLD with
are dysregulated with PBA (Olney et al., 2011). fused in sarcoma (FUS) pathology (Snowden et al.,
The cardinal feature of the PBA is the presence of epi- 2012) (Table 3.1).
sodes of laughter and/or crying, often triggered by a dis-
crete stimulus. These episodes present with paroxysm, Table 3.1
tend to be uncontrollable and involuntary, and may occur
Frequent symptoms in bvFTD patients and the anatomical
even in the absence of any congruent changes in the
correlates (Galimberti et al., 2015)
mood of the patient. The duration tends to be short. These
features help in the differentiation of PBA from mood Symptoms Frequency (%) Anatomical correlates
disorders, where the emotional state tends to last longer
and their emotional response is primarily a reflection of a Apathy 84 Right anterior cingulate,
specific situation or reaction. Several scales are available medial frontal lobe, and
to identify and characterize PBA (Newsom-Davis et al., caudate
1999; Smith et al., 2004). However, this is a clinical diag- Disinhibition 76 Right orbitofrontal cortex
nosis where the level of suspicion and clinical training Loss empathy 73 Right anterior temporal and
play important roles. This syndrome is effectively treated orbito-insular cortex
with a combination compound of dextromethorphan and Repetitive 71 Right supplementary motor
motor/ area and network
quinidine sulfate (Sauve, 2016).
compulsive involving orbitofrontal
behavior and anterior cingulate
Psychotic symptoms cortices, basal ganglia, and
thalamus
As discussed previously, bvFTD may present with prom-
Hyperorality 59 Network involving
inent neuropsychiatric symptoms and shares many and dietary orbitofrontal-insular-
symptoms with primary psychiatric disorders including changes striatal region and the
schizophrenia, obsessive compulsive disorder, border- posterior hypothalamus
line schizoid or antisocial personality disorder, and
36 B. MILLER AND J.J. LLIBRE GUERRA
The presence of overlapping symptoms between FTD Table 3.2
and specific psychiatric disorders can pose various prob- International consensus criteria for behavioral variant FTD
lems related to differential diagnosis, prognosis, and (Rascovsky et al., 2011)
treatment, but neuropsychologic assessment may help
to differentiate the FTD profile from non-FTD patients. I. Neurodegenerative disease
During the neuropsychologic testing, some core The following symptom must be present to meet criteria for
bvFTD:
symptoms of bvFTD may be evident, as some patients
A. Shows progressive deterioration of behavior and/or
make inappropriate comments to their examiners, appear
cognition by observation or history (as provided by a
cold or emotionally blunted, or appear inattentive and knowledgeable informant)
disinterested. During the examination, these patients
often make rule violations and perseverate. The magni- II. Possible bvFTD
tude of these findings is usually greater in bvFTD than Three of the following behavioral/cognitive symptoms (A–F)
must be present to meet criteria. Ascertainment requires that
in primary psychiatric disorders. Neuropsychologic test-
symptoms be persistent or recurrent, rather than single or rare
ing in bvFTD also frequently reveals executive dysfunc-
events
tion, which often correlates with tissue loss in the A. Early* behavioral disinhibition [one of the following
dorsolateral prefrontal cortex. In bvFTD, patient memory symptoms (A.1–A.3) must be present]:
tends to be better than in AD, but some authors have
pointed out some degree of impairment in episodic mem- A.1. Socially inappropriate behavior
ory in the early stages of FTD (Hodges et al., 2004). In A.2. Loss of manners or decorum
comparison with AD patients, bvFTD patients tend to
A.3. Impulsive, rash or careless actions
show higher scores of apathy, euphoria, disinhibition,
aberrant motor behavior, and eating abnormalities on B. Early apathy or inertia [one of the following symptoms
the Neuropsychiatric Inventory (NPI) (Levy et al., (B.1–B.2) must be present]:
1996; Liscic et al., 2007). On the other hand, drawing B.1. Apathy
and other visuospatial functions are often relatively
spared in FTD clinical subtypes (Rabinovici and B.2. Inertia
Miller, 2010). With the progression of the disease, C. Early loss of sympathy or empathy [one of the following
changes in language, like stereotypy of speech, semantic symptoms (C.1–C.2) must be present]:
deficits, and progressive reduction, may occur as a result
C.1. Diminished response to other people’s needs and feelings
of a disruption of dominant lobar language networks
(Pressman and Miller, 2014). C.2. Diminished social interest, interrelatedness or personal
warmth

bvFTD DIAGNOSIS CRITERIA D. Early perseverative, stereotyped or compulsive/ritualistic


behavior [one of the following symptoms (D.1–D.3) must
The diagnosis of bvFTD requires a clinical and neurop- be present]:
sychologic approach supported by the use of neuroimag-
D.1. Simple repetitive movements
ing or genetics studies. Most of the patients with bvFTD
present with lack of insight and gradual onset. For this D.2. Complex, compulsive, or ritualistic behaviors
reason, in the assessment of a patient with suspected D.3. Stereotypy of speech
bvFTD, it is extremely important to interview caregivers
regarding changes in patient behavior and cognition E. Hyperorality and dietary changes [one of the following
symptoms (E.1–E.3) must be present]:
(Cerami and Cappa, 2013; Pressman and Miller, 2014).
Improper diagnosis prevents patients and families E.1. Altered food preferences
from planning for the future and often leads to inappro-
E.2. Binge eating, increased consumption of alcohol or
priate and potentially harmful treatments, with FTD cigarettes
caregivers reporting that the delay to proper diagnosis
is one of the most frustrating aspects of their experience. E.3. Oral exploration or consumption of inedible objects
In 2011, the International Behavioral Variant FTD F. Neuropsychologic profile: executive/generation deficits
Consortium (FTDC) used pathologically confirmed cases with relative sparing of memory and visuospatial functions
of FTLD to develop sensitive and specific clinical criteria [all of the following symptoms (F.1–F.3) must be present]:
for diagnosing bvFTD, making possible a more accurate
F.1. Deficits in executive tasks
diagnosis (see Table 3.2) (Rascovsky et al., 2011).
The international research criteria for bvFTD empha- F.2. Relative sparing of episodic memory
sized six core symptoms for the diagnosis, and the pres- F.3. Relative sparing of visuospatial skills
ence of any three of these is sufficient for a diagnosis of
FRONTOTEMPORAL DEMENTIA 37
Table 3.2 than 50% of the FTLD patients proved to be τ-negative
but ubiquitin staining positive, so two main groups were
Continued
identified FTLD-τ and FTLD-ubiquitin (FTLD-U) or
unknown molecular pathology (Cairns et al., 2007;
III. Probable bvFTD
All of the following symptoms (A–C) must be present to meet MacKenzie et al., 2010). Later on, 80%–95% of FTLD-
criteria. U inclusions were found to be composed of transactive
A. Meets criteria for possible bvFTD response (TAR) DNA binding protein 43kDa (TDP-43)
(Neumann et al., 2006). Most of the remaining 5% of
B. Exhibits significant functional decline (by caregiver report
cases, who were τ- and TDP-43-negative, had inclusions
or as evidenced by Clinical Dementia Rating Scale or
of FUS. Thus, they were referred to as FTLD-FUS.
Functional Activities Questionnaire scores)
Correlations have been noted between the clinical
C. Imaging results consistent with bvFTD [one of the FTLD subtypes and underlying proteinopathies, but a
following (C.1–C.2) must be present]: strict one-to-one relationship is lacking. Nearly all
C.1. Frontal and/or anterior temporal atrophy on MRI or CT bvFTD cases fall within one of three histopathologic
groupings: τ, TDP-43, or FUS, each with a possibly
C.2. Frontal and/or anterior temporal hypoperfusion or
unique genetic contribution.
hypometabolism on PET or SPECT
Positive family history is observed in 40%–50% of
IV. bvFTD with definite FTLD pathology the FTLD patients, and in the bvFTD phenotype, a pos-
Criterion A and either criterion B or C must be present to meet itive family history could be present in 30% of the
criteria patients when symptoms of MND are present (Lashley
A. Meets criteria for possible or probable bvFTD et al., 2011; Goldman, 2012).
B. Histopathologic evidence of FTLD on biopsy or at Molecular genetic studies have identified various
postmortem genes with specific pathogenic mutations in FTD:
e.g., the microtubule-associated protein τ gene (MAPT),
C. Presence of a known pathogenic mutation
granulin (GRN), chromosome 9 open reading frame
V. Exclusionary criteria for bvFTD 72 (C9orf72), TARDBP, the gene encoding valosin-
Criteria A and B must be answered negatively for any bvFTD containing protein (VCP), and the charged multivesicu-
diagnosis. Criterion C can be positive for possible bvFTD but lar body protein 2B gene (CHMP2B) (see Table 3.3).
must be negative for probable bvFTD
Other genes appear to modify the susceptibility to dis-
A. Pattern of deficits is better accounted for by other
ease, but this remains a new and active area for
nondegenerative nervous system or medical disorders
investigation.
B. Behavioral disturbance is better accounted for by a For the purpose of this chapter we focus our attention
psychiatric diagnosis on C9orf72, GRN, and MAPT mutations and their path-
C. Biomarkers strongly indicative of Alzheimer’s disease or ologic correlations in bvFTD. These three genes are the
other neurodegenerative process most common and together explain at least 17%–40% of
the familial FTLD (DeJesus-Hernandez et al., 2011;
*As a general guideline “early” refers to symptom presentation within
Gijselinck et al., 2012). C9orf72 and GRN mutations
the first 3 years. are associated with TDP-43 deposits and MAPT with τ
deposition.
τ protein is involved in maintaining the microtubular
structure in the neuronal axon. Six isoforms have been
possible bvFTD. Probable FTD will be considered in the
described, three of which have three microtubule binding
presence of significant functional decline and imaging
repeats (3R), and three of which have four repeats (4R).
findings consistent with the disease or a known causal
τ is important in the pathogenesis of a variety of neuro-
genetic mutation. The diagnosis of definite bvFTD is
degenerative conditions, including AD and FTLD.
given if the possible and probable criteria are supported
Mutations in MAPT have been found to represent from
by a finding of FTLD pathology.
17% to 32% of patients with both FTD and a positive
family history (Rosso et al., 2003; Pressman and
CLINICAL AND PATHOLOGIC
Miller, 2014). MAPT mutations on chromosome 17q21
CORRELATION: HISTOPATHOLOGY
were the first to be linked to familial FTD. These muta-
AND GENETICS
tions were identified in families with an autosomal dom-
Initially, FTLD was pathologically identified by the pres- inant clinical syndrome of disinhibition, dementia, and
ence of frontotemporal atrophy and gliosis, with neurons Parkinsonism. Now, more than 50 different MAPT muta-
and glial cells containing inclusions of hyperphosphory- tions have been reported in 134 FTLD families (Bang
lated τ protein. This was referred to as FTLD-τ. More et al., 2015).
Table 3.3
Genes related with FTLD

C9orf72 MAPT GRN TARDBP VCP FUS CHMP2B

Cr. Loc 9p21.2 17q21.32 17q21.32 1p36.22 9p13.3 16p11.2 3p11.2


Protein Unknown τ Progranulin TDP-43 ATP binding FUS protein deposits Multivesicular body
protein 2B
Function Unknown Microtubule Growth factor, DNA/RNA modulation Intracellular transport DNA repair/regulates ESCRT-III complex
stabilization inflammation transcription production
Age of onset 51 (36–68) 44 (21–63) 56 (47–87) Early ALS Variable Early ALS 50–60
20–60
Frequency in FTD 20%–50% 40%–50% 5%–25% Rare Rare 5% Rare
Syndrome FTD, ALS, FTD- FTD, PSP, CBD FTD, CBD, PD, AD ALS, rarely FTD Paget’s with IBM, FTD, ALS, FTD, essential FTD
ALS, ALS tremor
psychiatric
symptoms
Brain inclusion TDP A/B τ TDP A ? TDP D Unknown
FRONTOTEMPORAL DEMENTIA 39
FTLD-τ is mainly associated with bvFTD and symp- MND (bvFTD-MND), or ALS, usually beginning at
toms of MND tend to be rare. MAPT mutations present the age of 55 (DeJesus-Hernandez et al., 2011; Sharon
with significant variability in clinical expression even et al., 2012; Snowden et al., 2012).
in the same family. In these families, symptoms are often These differences in genetic and neuropathologic sub-
present by the age of 50; however, the range for age of types suggest the need for different treatment approaches
onset is wide and strongly overlaps with other types of based on differential target engagement in the future
FTLD. It is remarkable that many carriers of MAPT management of the disease.
mutations present with a long psychiatric prodromal C9orf72, chromosome 9 open reading frame 72;
phase (Bang et al., 2015). MAPT, microtubule-associated protein τ; GRN, granulin;
TDP-43 protein is found in at least one half of all TARDBP, transactive response DNA binding protein
bvFTD cases on histologic examination and is present 43 kDa; VCP, valosin-containing protein; FUS, fused
in nearly all cases with FTD with amyotrophic lateral scle- in sarcoma; CHMP2B, charged multivesicular body
rosis (FTD-ALS). MacKenzie et al. (2011), in the new protein 2B; IBM, inclusion body myositis; ESCRT, endo-
international FTLD classification, elicited four FTLD- somal sorting complexes required for transport.
TDP subtypes, A–D, based on the cortical distribution,
intracellular location and morphology of the TDP inclu-
NEUROIMAGING IN bvFTD
sions. Based on large clinicopathologic studies (Josephs
et al., 2011), the most common FTLD-TDP subtype is In the diagnostic evaluation of bvFTD patients, fronto-
type A accounting for 41%–49% of the cases, followed temporal atrophy or hypometabolism with structural
by type B with 28%–34% and type C with 17%–25%, and/or functional neuroimaging transforms a diagnosis
while type D is rare (Sieben et al., 2012). FTLD-TDP- from possible bvFTD into probable bvFTD
43 type A is seen in patients with bvFTD or nfvPPA, type (Rascovsky et al., 2011; Pressman and Miller, 2014).
B is typical for FTD-MND, and type C is seen with svPPA Furthermore, neuroimaging studies may help exclude
and rarely with other FTD subtypes. alternative disorders such as tumors, vascular dementia,
The two more common genetic mutations associated or normal pressure hydrocephalus.
with TDP-43 pathology are GRN and C9orf72 (Baker FTLD is associated with diverse patterns of atrophy
et al., 2006; Cruts et al., 2006). In the case of the progra- of frontal and anterior temporal lobes. Different patterns
nulin gene (PGRN) more than 69 different GRN muta- of atrophy have been described in correlation with the
tions have been reported in 231 families (Cruts et al., clinical phenotypes. All of them are characterized by pro-
2012). GRN mutations usually present by the age of gressive cell loss, stereotypical spread, and abnormal
60 years, having an age of onset that is slightly higher protein aggregation.
than for other forms of FTD. It is remarkable that vari- Patterns of neuroimaging findings can differentiate
ability in penetrance has been described, with 50%– bvFTD from other FTD syndromes, AD, and other
60% of the mutation carriers being affected by the age dementias (Muñoz-Ruiz et al., 2012), with important
of 60 years, and 90%–95% by the age of 70 years results coming from Seeley and colleagues who have
(Sieben et al., 2012). GRN mutations are often associated demonstrated that bvFTD is associated with atrophy of
with asymmetric cerebral atrophy and, in addition to the orbitofrontal, anterior cingulate, anterior insular,
bvFTD, may be associated with nfvPPA and Parkinson- and anterior temporal cortices, particularly within the
ism. Motor neuron symptoms are rare (Chen-Plotkin right hemisphere (Lee et al., 2016). This group has also
et al., 2011). shown that changes in the salience network, which is
C9orf72 encodes a ubiquitously expressed protein of comprised of the orbitofrontal cortex, anterior cingulate
unknown function. In the normal population, the size of cortex, anterior insula, and presupplementary motor area,
the GGGGCC repeat ranges from 3 to 25 units; a noncod- may underlie the selective vulnerability and the stereo-
ing GGGGCC hexanucleotide expansion in this gene of typical spread of this neurodegenerative illness (Seeley
more than 400 base pairs is strongly associated with both et al., 2009).
FTD and ALS (DeJesus-Hernandez et al., 2011; Renton bvFTD is associated with frontal and temporal lobe
et al., 2011). At the University of California, San Fran- atrophy (Fig. 3.1) but also involves other structures like
cisco Memory and Aging Center, mutations in this gene the thalamus, striatum, and hypothalamus. Typically the
account for roughly 50% of familial FTD, other reports atrophy pattern is more pronounced on the right than left
give a range of 13%–26% among familial FTD cases frontotemporal areas (Lee et al., 2016; Muñoz-Ruiz
(Majounie et al., 2012). et al., 2012).
C9orf72 hexanucleotide repeat expansion is the Patterns of atrophy differ slightly among the genetic
most common known genetic cause of FTD and ALS forms of FTLD. Anteromedial temporal atrophy has been
and most often presents with bvFTD, bvFTD with associated with mutations in MAPT (Rohrer et al., 2010),
40 B. MILLER AND J.J. LLIBRE GUERRA

Fig. 3.1. A T1 magnetic resonance image of the brain in sagittal, axial, and coronal cuts, shown in neurologic convention, showing
severe anterior predominant atrophy with hydrocephalus ex vacuo in a patient with advanced behavioral variant frontotemporal
dementia (bvFTD).

Syndrome-specific regional atrophy patterns: patients vs controls Atrophy max


= seed ROI
AD bvFTD SD PNFA CBS
L IFG R PMC
+35 +11 +14 +10 +40 6

R FI 4.4
A R Ang L TPole
Intrinsic functional connectivity networks: healthy controls
5

3.6
B
Structural covariance networks: healthy controls
6

4.6
C
Fig. 3.2. Convergent syndromic atrophy, healthy intrinsic connectivity networks (ICN), and healthy structural covariance (SC)
patterns (A) Five distinct clinical syndromes showed dissociable atrophy patterns, whose cortical maxima (circled) provided seed
ROIs for ICN and SC analyses. (B) ICN mapping experiments identified five distinct networks anchored by the five syndromic
atrophy seeds. (C) Healthy subjects further showed gray matter volume covariance patterns that recapitulated results shown in
(A and B). For visualization purposes, results are shown at P < 0.00001 uncorrected (A and C) and P < 0.001 corrected height
and extent thresholds (B). In (A–C), results are displayed on representative sections of the MNI template brain. In coronal and
axial images, the left side of the image corresponds to the left side of the brain. ANG ¼ angular gyrus; FI ¼ frontoinsula;
IFGoper ¼ inferior frontal gyrus, pars opercularis; PMC ¼ premotor cortex; TPole ¼ temporal pole (Seeley et al., 2009).

whereas mutations in GRN have been associated with anterior temporal atrophy (Beck et al., 2008; Rohrer
asymmetric temporoparietal atrophy, and C9orf72 muta- et al., 2010).
tions tend to present symmetric atrophy, predominantly Patterns of atrophy may also help in the differential
involving dorsolateral, medial, and orbitofrontal lobes, diagnosis, with Seeley et al. (2009) pointing out differen-
anterior temporal lobes, cerebellum, and medial thala- tial neuroimaging profiles in AD, bvFTD, svPPA,
mus. Sporadic forms usually present with frontal and nfvPPA, and CBD (Fig. 3.2). Even though these atrophy
FRONTOTEMPORAL DEMENTIA 41
Table 3.4
Differential diagnosis of bvFTD

Psychiatric disorders Neurodegenerative disorders

Obsessive–compulsive Alzheimer’s disease


disorder
Bipolar disorder Chronic traumatic encephalopathy
Fig. 3.3. Axial fluorodeoxyglucose positron emission tomog- Depression Other FTD syndromes
raphy (FDG-PET) images of a patient with advanced bvFTD, Schizophrenia Movement disorders
demonstrating hypometabolism in the anterior lobes bilater- Treatable conditions that can mimic FTD
ally, but right more so than left.
Metabolic disturbances Cerebrovascular disease
patterns in structural MRI are extremely helpful in the Nutritional deficiencies CNS tumors
diagnostic approach, it is important to note that early CNS infections Paraneoplastic conditions
in the disease, structural imaging can be normal. In these Substance abuse Normal pressure hydrocephalus
early stages, functional neuroimaging studies such as Heavy metal toxicity Low intracranial pressure
fluorodeoxyglucose positron emission tomography
(FDG-PET) may be slightly more sensitive than MRI
for the diagnosis (Womack et al., 2011). Characteristic
patterns of regional glucose hypometabolism on FDG- bvFTD patients are often initially misdiagnosed with a
PET, a marker of neuronal injury and neurodegeneration, major depressive disorder, bipolar affective disorder, or
have been identified in association with the common schizophrenia (Gálvez-Andres et al., 2007; Woolley
neurodegenerative dementias and can be clinically useful et al., 2007, 2011; Velakoulis et al., 2009).
with a sensitivity of 97% and a specificity of 86% for dis- In one study, 28% of patients had a prior psychiatric
tinguishing AD from FTD (Foster et al., 2007). FDG- diagnosis, and the mean delay between receiving psychi-
PET reveals hypometabolism of frontal, anterior cingu- atric and neurodegenerative disease diagnoses was
late and anterior temporal regions in FTD, in contrast 33 months. Women with bvFTD received a psychiatric
to temporoparietal and posterior cingulate hypometabo- diagnosis more often than men. This study suggests that
lism in AD (Fig. 3.3). physicians should consider a more comprehensive eval-
A significant problem in bvFTD is predicting the uation in patients who present with new-onset behav-
molecular subtype from the clinical syndrome. Amyloid ioral, emotional, or cognitive changes after age 40 in
imaging is useful for distinguishing FTLD syndromes order to rule out bvFTD or another neurodegenerative
from AD, and eventually, it is hoped that τ imaging will condition (Woolley et al., 2011).
help predict the risk of developing the disease, assess dis- Others neurodegenerative disorders can be mistaken
ease progression, and measure the efficacy of therapeu- for FTD-spectrum disorders, particularly AD, vascular
tics. In the last few years, the search for a sensitive and dementia, and dementia with Lewy bodies. Early age
specific τ binding ligand has intensified and τ neuroim- of onset AD (EOAD) can be challenging to differentiate
aging agents are under development. from bvFTD because frequently it does not have the clas-
sical amnestic presentation of later onset AD. In some
cases, there may be a prominent executive or language
DIFFERENTIAL DIAGNOSIS dysfunction as well.
It is also important to make reference to the FTD phe-
Lack of an early or clear diagnosis of bvFTD may lead to
nocopy. This term was introduced by John Hodges and
delayed and inappropriate treatment, as well as family
Chris Kipps and may represent a diagnostic dilemma
and patient distress. bvFTD syndromes can be mistaken
(Kipps et al., 2010), describing patients who seem to
for other neurodegenerative or psychiatric syndromes
meet the bvFTD criteria but do not appear to progress
(Table 3.4), and in early stages, the diagnosis is more
clinically and do not show atrophy on MRI.
challenging, as it is important to exclude treatable condi-
FTD phenocopy pattern usually falls into four main
tions that can mimic FTLD. A comprehensive clinical
subtypes:
and neuropsychologic evaluation, combined with labora-
tory studies and neuroimaging, are essential components 1. FTD by proxy
of the required workup. 2. Self-diagnosed FTD
Dementias are often mistaken for psychiatric disease, 3. Primary psychiatric disorder
and bvFTD patients are at highest risk for misdiagnosis. 4. Slow FTD
42 B. MILLER AND J.J. LLIBRE GUERRA
(Franceschi et al., 2005; Huey et al., 2006). Disruption in
In FTD by proxy, the caregiver becomes convinced
the dopaminergic system, with low levels of dopamine
that their loved one has bvFTD. This idea is often rein-
metabolites and severely reduced presynaptic dopamine
forced by reading about the disease on the Internet or
transporters in the putamen and caudate of FTD patients,
though discussion with someone familiar with the
seems to be part of the FTD neuropharmacology
disease. A similar story is seen in self-diagnosed FTD.
(Sj€ogren et al., 1998; Rinne et al., 2002). There is no evi-
Sometimes a psychiatric syndrome is misdiagnosed
dence supporting a deficit of acetylcholine in this disease
as FTD. Usually, however, patients with primary psychi-
(Hansen et al., 1988). Consequently, the pharmacologic
atric conditions like bipolar disorder have symptoms that
management of neurobehavioral symptoms should ide-
begin earlier in life, show relatively normal neuroimag-
ally suit the typical changes in neurotransmitter systems
ing, respond better to psychoactive compounds, and do
occurring with the disease.
not progress to dementia. Finally, there are gene carriers
In 1997 Swartz and colleagues first treated 11 FTD
who may initially show normal imaging and progress
subjects with fluoxetine, sertraline, or paroxetine and
very slowly. In particular, some patients with C9 muta-
found improvement in disinhibition, apathy, carbohy-
tions have been reported to have a very low progression
drate cravings, and compulsive behaviors. In 2005 Men-
rate and long prodromal phase (Bang et al., 2015).
dez and colleagues described decreased verbal and motor
stereotypies when sertraline was used in this group of
MANAGEMENT OF BEHAVIORAL patients. A meta-analysis of studies using serotonergic
SYMPTOMS IN FTD drugs was conducted by Huey et al. in 2006, and signif-
There is no treatment available that changes the course of icant decrease in behavioral symptoms as measured by
the bvFTD, so the focus of medical therapy is on symp- the NPI was observed (Huey et al., 2006). In total, the
tomatic relief. An individualized management plan, with evidence suggests that serotonin selective reuptake
both nonpharmacologic and pharmacologic interven- inhibitors (SSRIs) (fluoxetine, sertraline, paroxetine, flu-
tions, should be created. voxamine, and citalopram) are effective in helping with
various symptoms of FTD, including disinhibition,
Nonpharmacologic interventions impulsivity, repetitive behaviors, and eating disorders
(Sj€ogren et al., 1998; Huey et al., 2006; Manoochehri
Nonpharmacologic interventions should focus on the and Huey, 2012).
safety and well-being of the patient. Discussion with fam- Some patients with bvFTD will require the use of anti-
ily around financial issues, such as limiting access to credit psychotic medications for the treatment of severe neuro-
cards and bank accounts, driving safety, and achieving an behavioral symptoms. These compounds are usually
environment that assures the physical safety of the patient considered if there is no success with behavioral modifi-
(Talerico and Evans, 2001; Piguet et al., 2011a) and early cation or SSRIs. Diminished dopaminergic function in
retirement should be considered if the patient is currently these patients makes them vulnerable to extrapyramidal
employed (Pressman and Miller, 2014). side effects. Therefore, agents with relatively less D2
Exercises and physical therapy may be also helpful in receptor antagonism, such as quetiapine, are preferred
patients with impending problems with movement. Chen if antipsychotics are needed (Pijnenburg et al., 2003;
and colleagues (Cheng et al., 2014) have shown a benefit Komossa et al., 2011; Asmal et al., 2013).
in mood, cognition, and overall health in patients with Acetylcholinesterase inhibitors are not recommended
dementia when regular exercise routines are performed. in this group of patients, as they do not have a cholinergic
Physical therapy may be helpful in patients with mobility deficit like AD or DLB, and trials have not shown a con-
problems such as Parkinsonism and may help to reduce vincing benefit for cholinesterase inhibitors in cognition
the risk of falls. or behavior (Mendez et al., 2007). AChIs are incorrectly
The presence of hyperorality in some patients may prescribed in approximately 40% of bvFTD patients
require special care from caregivers to prevent excessive (Pressman and Miller, 2014). Memantine is an NMDA
weight gain or the dangerous placement of inedible receptor antagonist used for slowing cognitive decline
objects in the mouth (Manoochehri and Huey, 2012). with moderate AD, but no significant difference in
behavioral or cognitive function was found in bvFTD
when compared with a placebo (Diehl-Schmid et al.,
Pharmacologic interventions
2008; Boxer et al., 2009; Vercelletto et al., 2011). Several
Studies in the neurochemistry of bvFTD have shown a other drugs, like lithium and some anticonvulsants, have
serotoninergic network disruption, with decreased sero- been tested in FTD with negative or inconclusive results.
tonin levels and 5-HT1A and 5-HT2A receptors in fron- Risks of toxicity are generally deemed to outweigh
totemporal regions and neuronal loss in the raphe nuclei theoretical benefits (Pressman and Miller, 2014).
FRONTOTEMPORAL DEMENTIA 43
CONCLUSIONS Chen-Plotkin AS, Martinez-Lage M, Sleiman PMA et al.
(2011). Genetic and clinical features of progranulin-
FTD is increasingly recognized as a leading cause of early- associated frontotemporal lobar degeneration. Arch
onset dementia. bvFTD is the most common syndrome in Neurol 68: 488–497.
FTD and can be associated with behavior changes mim- Cruts M, Gijselinck I, van der Zee J et al. (2006). Null muta-
icking manic, depressive, obsessive–compulsive, or other tions in progranulin cause ubiquitin-positive frontotem-
psychiatric syndromes, and causing social and family dis- poral dementia linked to chromosome 17q21. Nature
ruption. bvFTD can result from a number of different pro- 442: 920–924.
teinopathies, and some cases have a genetic etiology. Cruts M, Theuns J, Van Broeckhoven C (2012). Locus-specific
A comprehensive clinical and neuropsychologic evalua- mutation databases for neurodegenerative brain diseases.
Hum Mutat 33: 1340–1344.
tion, combined with laboratory studies and neuroimaging,
DeJesus-Hernandez M, MacKenzie IR, Boeve BF et al. (2011).
can help in recognizing the core features of bvFTD and in Expanded GGGGCC hexanucleotide repeat in noncoding
avoiding misdiagnosis. The pharmacologic management region of C9ORF72 causes chromosome 9p-linked FTD
of behavioral symptoms currently relies on the effect of and ALS. Neuron 72: 245–256.
SSRIs, with some efficacy in reducing disinhibition, repet- Diehl-Schmid J, F€ orstl H, Perneczky R et al. (2008).
itive behaviors, and hyperorality. Small doses of atypical A 6-month, open-label study of memantine in patients with
antipsychotics may be required when there is no success frontotemporal dementia. Int J Geriatr Psychiatry 23:
with SSRIs. There is no evidence supporting the use of 754–759.
cholinesterase inhibitors or memantine. Promising areas Eslinger PJ, Moore P, Antani S et al. (2012). Apathy in fron-
of research include PET imaging with τ ligands, which totemporal dementia: behavioral and neuroimaging corre-
may make it possible to identify subgroups that could lates. Behav Neurol 25: 127–136.
Foster NL, Heidebrink JL, Clark CM et al. (2007). FDG-PET
benefit from future therapies targeting the underlying
improves accuracy in distinguishing frontotemporal
pathology with disease. dementia and Alzheimer’s disease. Brain 130: 2616–2635.
Franceschi M, Anchisi D, Pelati O et al. (2005). Glucose
REFERENCES metabolism and serotonin receptors in the frontotemporal
lobe degeneration. Ann Neurol 57: 216–225.
Asmal L, Flegar SJ, Wang J et al. (2013). Quetiapine versus Galimberti D, Dell’Osso B, Altamura AC et al. (2015).
other atypical antipsychotics for schizophrenia. Cochrane Psychiatric symptoms in frontotemporal dementia: epide-
Database Syst Rev (11): CD006625. miology, phenotypes, and differential diagnosis. Biol
Baker M, MacKenzie IR, Pickering-Brown SM et al. (2006). Psychiatry 78: 684–692.
Mutations in progranulin cause tau-negative frontotemporal Gálvez-Andres A, Blasco-Fontecilla H, González-Parra S
dementia linked to chromosome 17. Nature 442: 916–919. et al. (2007). Secondary bipolar disorder and Diogenes syn-
Bang J, Spina S, Miller BL (2015). Frontotemporal dementia. drome in frontotemporal dementia: behavioral improve-
Lancet (London, England) 386: 1672–1682. ment with quetiapine and sodium valproate. J Clin
Bartchowski Z, Gatla S, Khoury S et al. (2018). Empathy Psychopharmacol 27: 722–723.
changes in neurocognitive disorders: a review. Ann Clin Gijselinck I, Van Langenhove T, van der Zee J et al. (2012).
Psychiatry 30 (3): 220–232. A C9orf72 promoter repeat expansion in a Flanders-
Beck J, Rohrer JD, Campbell T et al. (2008). A distinct clinical, Belgian cohort with disorders of the frontotemporal lobar
neuropsychological and radiological phenotype is associ- degeneration-amyotrophic lateral sclerosis spectrum: a
ated with progranulin gene mutations in a large UK series. gene identification study. Lancet Neurol 11: 54–65.
Brain 131: 706–720. Goldman JS (2012). New approaches to genetic counseling
Boxer AL, Lipton AM, Womack K et al. (2009). An open-label and testing for Alzheimer’s disease and frontotemporal
study of memantine treatment in 3 subtypes of frontotem- degeneration. Curr Neurol Neurosci Rep 12: 502–510.
poral lobar degeneration. Alzheimer Dis Assoc Disord 23: Gregory CA, Serra-Mestres J, Hodges JR (1999). Early diag-
211–217. nosis of the frontal variant of frontotemporal dementia:
Cairns NJ, Bigio EH, MacKenzie IRA et al. (2007). how sensitive are standard neuroimaging and neuropsycho-
Neuropathologic diagnostic and nosologic criteria for logic tests? Neuropsychiatry Neuropsychol Behav Neurol
frontotemporal lobar degeneration: consensus of the con- 12: 128–135.
sortium for frontotemporal lobar degeneration. Acta Hansen LA, Deteresa R, Tobias H et al. (1988). Neocortical
Neuropathol 114: 5–22. morphometry and cholinergic neurochemistry in Pick’s
Cerami C, Cappa SF (2013). The behavioral variant of fronto- disease. Am J Pathol 131: 507–518.
temporal dementia: linking neuropathology to social cogni- Hodges JR, Davies RR, Xuereb JH et al. (2004). Ann Neurol
tion. Neurol Sci 34: 1267–1274. 56: 399–406.
Cheng S-T, Chow PK, Song Y-Q et al. (2014). Mental and Huey ED, Putnam KT, Grafman J (2006). A systematic review
physical activities delay cognitive decline in older persons of neurotransmitter deficits and treatments in frontotem-
with dementia. Am J Geriatr Psychiatry 22: 63–74. poral dementia. Neurology 66: 17–22.
44 B. MILLER AND J.J. LLIBRE GUERRA
Josephs KA, Hodges JR, Snowden JS et al. (2011). between hippocampal volumetry, tensor-based morphom-
Neuropathological background of phenotypical variability etry and voxel-based morphometry. PLoS One 7: e52531.
in frontotemporal dementia. Acta Neuropathol 122: Neumann M, Sampathu DM, Kwong LK et al. (2006).
137–153. Ubiquitinated TDP-43 in frontotemporal lobar degeneration
Kertesz A, Cyn Ang L, Jesso S et al. (2013). Psychosis and hal- and amyotrophic lateral sclerosis. Science 314: 130–133.
lucinations in frontotemporal dementia with the C9ORF72 Newsom-Davis IC, Abrahams S, Goldstein LH et al. (1999).
mutation: a detailed clinical cohort. Cogn Behav Neurol 26: The emotional lability questionnaire: a new measure of
146–154. emotional lability in amyotrophic lateral sclerosis.
Kipps CM, Hodges JR, Hornberger M (2010). Nonprogressive J Neurol Sci 169: 22–25.
behavioural frontotemporal dementia: recent develop- Olney NT, Goodkind MS, Lomen-Hoerth C et al. (2011).
ments and clinical implications of the ‘bvFTD phenocopy Behaviour, physiology and experience of pathological
syndrome’. Curr Opin Neurol 23: 628–632. laughing and crying in amyotrophic lateral sclerosis.
Knopman DS, Roberts RO (2011). Estimating the number of Brain 134: 3458–3469.
persons with frontotemporal lobar degeneration in the Onyike CU, Diehl-Schmid J (2013). The epidemiology of
US population. J Mol Neurosci 45: 330–335. frontotemporal dementia. Int Rev Psychiatry 25: 130–137.
Komossa K, Rummel-Kluge C, Schwarz S et al. (2011). Piguet O (2011). Eating disturbance in behavioural-variant
Risperidone versus other atypical antipsychotics for frontotemporal dementia. J Mol Neurosci 45 (3): 589–593.
schizophrenia. Cochrane Database Syst Rev (1): Piguet O, Hornberger M, Mioshi E et al. (2011a). Behavioural-
CD006626. variant frontotemporal dementia: diagnosis, clinical stag-
Lanata SC, Miller BL (2016). The behavioural variant fronto- ing, and management. Lancet Neurol 10: 162–172.
temporal dementia (bvFTD) syndrome in psychiatry. Piguet O, Petersen Å, Yin Ka Lam B et al. (2011b). Eating and
J Neurol Neurosurg Psychiatry 87: 501–511. hypothalamus changes in behavioral-variant frontotem-
Lashley T, Rohrer JD, Bandopadhyay R et al. (2011). poral dementia. Ann Neurol 69: 312–319.
A comparative clinical, pathological, biochemical and Pijnenburg YAL, Sampson EL, Harvey RJ et al. (2003).
genetic study of fused in sarcoma proteinopathies. Brain Vulnerability to neuroleptic side effects in frontotemporal
134: 2548–2564. lobar degeneration. Int J Geriatr Psychiatry 18: 67–72.
Lee S, Sias A, Mandelli M et al. (2016). Network degeneration Pressman PS, Miller BL (2014). Diagnosis and management of
and dysfunction in presymptomatic C9orf72 expansion behavioral variant frontotemporal dementia. Biol
carriers. Neuroimage Clin 14: 286–297. Psychiatry 75 (7): 574–581.
Levy ML, Miller BL, Cummings JL et al. (1996). Alzheimer Rabinovici G, Miller B (2010). Frontotemporal lobar degener-
disease and frontotemporal dementias. Behavioral distinc- ation: epidemiology, pathophysiology, diagnosis and man-
tions. Arch Neurol 53: 687–690. agement. CNS Drugs 24 (5): 375–398.
Liscic RM, Storandt M, Cairns NJ et al. (2007). Clinical and Rankin KP (2005). Self awareness and personality change in
psychometric distinction of frontotemporal and Alzheimer dementia. J Neurol Neurosurg Psychiatry 76: 632–639.
dementias. Arch Neurol 64: 535. Rankin KP, Gorno-tempini ML, Allison SC et al. (2006).
MacKenzie IRA, Neumann M, Bigio EH et al. (2010). Structural anatomy of empathy in neurodegenerative dis-
Nomenclature and nosology for neuropathologic subtypes ease. Brain 129: 2945–2956.
of frontotemporal lobar degeneration: an update. Acta Rascovsky K, Hodges JR, Knopman D et al. (2011).
Neuropathol 119: 1–4. Sensitivity of revised diagnostic criteria for the behavioural
MacKenzie IRA, Neumann M, Baborie A et al. (2011). variant of frontotemporal dementia. Brain 134: 2456–2477.
A harmonized classification system for FTLD-TDP pathol- Renton AE, Majounie E, Waite A et al. (2011). A hexanucleotide
ogy. Acta Neuropathol 122: 111–113. repeat expansion in C9ORF72 is the cause of chromosome
Mahoney CJ, Beck J, Rohrer JD et al. (2012). Frontotemporal 9p21-linked ALS-FTD. Neuron 72: 257–268.
dementia with the C9ORF72 hexanucleotide repeat expan- Riedl L, MacKenzie IR, F€ orstl H et al. (2014). Frontotemporal
sion: clinical, neuroanatomical and neuropathological fea- lobar degeneration: current perspectives. Neuropsychiatr
tures. Brain 135: 736–750. Dis Treat 10: 297–310.
Majounie E, Renton A, Mok K et al. (2012). Frequency of the Rinne JO, Laine M, Kaasinen V et al. (2002). Striatal dopa-
C9orf72 hexanucleotide repeat expansion in patients with mine transporter and extrapyramidal symptoms in fronto-
amyotrophic lateral sclerosis and frontotemporal dementia: temporal dementia. Neurology 58: 1489–1493.
a cross-sectional study. Lancet Neurol 11: 297–298. Rohrer JD, Ridgway GR, Modat M et al. (2010). Distinct pro-
Manoochehri M, Huey ED (2012). Diagnosis and management files of brain atrophy in frontotemporal lobar degeneration
of behavioral issues in frontotemporal dementia. Curr caused by progranulin and tau mutations. Neuroimage 53:
Neurol Neurosci Rep 12: 528–536. 1070–1076.
Mendez MF, Shapira JS, McMurtray A et al. (2007). Rosen HJ, Allison SC, Schauer GF et al. (2005).
Preliminary findings: behavioral worsening on donepezil Neuroanatomical correlates of behavioural disorders in
in patients with frontotemporal dementia. Am J Geriatr dementia. Brain 128: 2612–2625.
Psychiatry 15: 84–87. Rosen HJ, Wilson MR, Schauer GF et al. (2006).
Muñoz-Ruiz MÁ, Hartikainen P, Koikkalainen J et al. (2012). Neuroanatomical correlates of impaired recognition of
Structural MRI in frontotemporal dementia: comparisons emotion in dementia. Neuropsychologia 44: 365–373.
FRONTOTEMPORAL DEMENTIA 45
Rosso SM, Kaat LD, Baks T et al. (2003). Frontotemporal Takada LT, Sha SJ (2012). Neuropsychiatric features of
dementia in The Netherlands: patient characteristics and C9orf72-associated behavioral variant frontotemporal
prevalence estimates from a population-based study. dementia and frontotemporal dementia with motor neuron
Brain 126: 2016–2022. disease. Alzheimers Res Ther 4: 38.
Sauve W (2016). Recognizing and treating pseudobulbar Talerico KA, Evans LK (2001). Responding to safety issues in
affect. CNS Spectr S1: 34–44. frontotemporal dementias. Neurology 56: S52–S55.
Seeley WW, Crawford RK, Zhou J et al. (2009). Velakoulis D, Walterfang M, Mocellin R et al. (2009).
Neurodegenerative diseases target large-scale human brain Frontotemporal dementia presenting as schizophrenia-like
networks. Neuron 62: 42–52. psychosis in young people: clinicopathological series and
Sharon JS, Takada LT, Rankin KP et al. (2012). review of cases. Br J Psychiatry 194: 298–305.
Frontotemporal dementia due to C9ORF72 mutations. Vercelletto M, Boutoleau-Bretonniere C, Volteau C et al.
Neurology 79: 1002–1011. (2011). Memantine in behavioral variant frontotemporal
Shinagawa S, Nakajima S, Plitman E et al. (2014). dementia: negative results. J Alzheimers Dis 23: 749–759.
Psychosis in frontotemporal dementia. J Alzheimers Dis Wald€ o ML, Gustafson L, Passant U et al. (2015). Psychotic
42: 485–499. symptoms in frontotemporal dementia: a diagnostic
Sieben A, Van Langenhove T, Engelborghs S et al. (2012). The dilemma? Int Psychogeriatr. 27: 531–539.
genetics and neuropathology of frontotemporal lobar Womack KB, Diaz-Arrastia R, Aizenstein HJ et al. (2011).
degeneration. Acta Neuropathol 124: 353–372. Temporoparietal hypometabolism in frontotemporal lobar
Sj€
ogren M, Minthon L, Passant U et al. (1998). degeneration and associated imaging diagnostic errors.
Decreased monoamine metabolites in frontotemporal demen- Arch Neurol 68: 329–337.
tia and Alzheimer’s disease. Neurobiol Aging 19: 379–384. Woolley JD, Gorno-Tempini ML, Seeley WW et al. (2007).
Smith RA, Berg JE, Pope LE et al. (2004). Validation of the Binge eating is associated with right orbitofrontal-insular-
CNS emotional lability scale for pseudobulbar affect (path- striatal atrophy in frontotemporal dementia. Neurology 69:
ological laughing and crying) in multiple sclerosis patients. 1424–1433.
Mult Scler 10: 679–685. Woolley JD, Khan BK, Murthy NK et al. (2011). The diagnos-
Snowden JS, Rollinson S, Thompson JC et al. (2012). Distinct tic challenge of psychiatric symptoms in neurodegenera-
clinical and pathological characteristics of frontotemporal tive disease: rates of and risk factors for prior psychiatric
dementia associated with C9ORF72 mutations. Brain 135: diagnosis in patients with early neurodegenerative disease.
693–708. J Clin Psychiatry 72: 126–133.

You might also like