You are on page 1of 8

REVIEW

Diagnosis and Management of Behavioral Variant


Frontotemporal Dementia
Peter S. Pressman and Bruce L. Miller
Frontotemporal dementia was documented over a century ago. The last decade, however, has seen substantial changes in our
conceptions of this increasingly recognized disorder. Different clinical variants have been delineated, the most common of which is the
behavioral variant (bvFTD). Updated diagnostic criteria have been established. New histopathological findings and genetic etiologies
have been discovered. Research continues to uncover molecular mechanisms by which abnormal proteins accumulate in degenerating
brain tissue. Novel neuroimaging techniques suggest that functional networks are diminished in bvFTD that might be relevant to
empathy and social behavior. Despite rapid advances in our understanding of bvFTD, the disease is still under-recognized and commonly
misdiagnosed. The result is inappropriate patient care. Recognizing the various presentations of bvFTD and its histological and genetic
subtypes might further diagnosis, treatment, and research.

semantic variant of primary progressive aphasia (svPPA), and


Key Words: Behavioral variant frontotemporal dementia, bvFTD, nonfluent variant primary progressive aphasia (nfvPPA) (7,8)
frontotemporal dementia, frontotemporal lobar degeneration, (Table S1 in Supplement 1). A third variant of PPA, logopenic
FTD, Pick’s disease variant primary progressive aphasia is associated with atrophy of
the temporoparietal junction and histopathological findings of
Alzheimer’s disease (AD).

T
he frontal lobes of the brain influence human personality,
motivation, attention, executive function, emotions, social The focus of this review is on the most common FTD subtype,
capability, and language. Although the Czech neuropsychia- bvFTD. This dementia shares many symptoms with primary
trist Arnold Pick first described a progressive aphasia in 1892, psychiatric disorders, including schizophrenia, obsessive-
selective degeneration of the frontal and temporal lobes compulsive disorder, borderline personality disorder, and bipolar
remained relatively obscure for approximately a century (1,2). In disorder. In addition, neurodegenerative diseases with prominent
the last few decades, Pick’s disease, now called frontotemporal movement abnormalities including progressive supranuclear
dementia (FTD), has become recognized as the second most palsy (PSP) (9,10), corticobasal degeneration (CBD) (11,12), and
common neurodegenerative dementia in patients under the age amyotrophic lateral sclerosis (ALS) often present as bvFTD or
of 65 and the third most common neurodegenerative dementia nfvPPA (13).
in all age groups (3,4). The neuropathological classification of FTLD subtypes is based
The term FTD refers to a heterogeneous group of syndromes upon the types of misfolded protein aggregates found within the
caused by progressive and selective degeneration of the frontal brain such as tau, trans-activator regulatory DNA binding protein
lobes, temporal lobes, or both with relative preservation of the 43 (TDP-43) (14), or fused in sarcoma (FUS) (15,16). The neuro-
posterior cortical regions. This focal anterior-predominant neuro- pathological changes in patients with ALS, CBD, and PSP are
degeneration leads to disorders of behavior, language, and strongly linked with the presence of tau (PSP, CBD) or TDP-43
executive function. (ALS) aggregates.
Frontotemporal dementia has acquired several different Symptomatically, patients with FTD might present at a
names over the years, including Pick’s disease, frontal lobe relatively young age, and many are mistaken for nonprogressive
dementia of the non-Alzheimer’s type, frontotemporal lobar psychiatric diseases more commonly seen in young adulthood
degeneration, and dementia of the frontal type. Frontotemporal such as schizophrenia and bipolar disorder. Approximately 50% of
dementia remains the most common term in use. We use the patients in our bvFTD cohort were initially diagnosed with a
term FTD to describe the clinical syndrome and frontotemporal psychiatric illness before being recognized as having bvFTD (17).
lobar degeneration (FTLD) for the pathological diagnosis (5). Improper diagnosis prevents patients and families from properly
Pick’s disease is now reserved for pathological findings of planning for the future and often leads to inappropriate and
argyrophilic, tau-positive neuronal inclusions (Pick bodies) (6). potentially harmful treatments.
This clear separation between syndromic and pathological diag-
noses is fundamental to a clear understanding of FTD, because
identical symptoms might be caused by many different molecular Epidemiology
and cellular mechanisms.
Frontotemporal dementia is currently divided into three major The incidence of FTD seems to peak during the 6th and 7th
subtypes: behavioral variant frontotemporal dementia (bvFTD), decades of life, at 2.2, 3.3, and 8.9/100,000 person-years at 40–49,
50–59, and 60–69, respectively. Mean age at onset varies some-
From the Memory and Aging Center, Department of Neurology, University what across FTD subtypes, with bvFTD having the earliest onset,
of California at San Francisco, San Francisco, California. followed by svPPA and then nvfPPA (18). The average survival
Address correspondence to Peter Pressman, M.D., Department of Neurol- time from diagnosis to death ranges from 3 years for bvFTD
ogy, University of California at San Francisco, Memory and Aging patients with motor neuron disease to approximately 12 years in
Center, Clinical Instructor, Campus Box 1207, 675 Nelson Rising Lane, patients with semantic variant PPA (19). The mean age at
Suite 190, San Francisco, CA 94143-1207; E-mail: ppressman@memory. presentation for bvFTD is approximately 58 years, although cases
ucsf.edu. presenting as early as the 2nd decade and as late as the 9th
Received May 9, 2013; revised Oct 31, 2013; accepted Nov 1, 2013. decade have been reported (18,20).

0006-3223/$36.00 BIOL PSYCHIATRY 2014;75:574–581


http://dx.doi.org/10.1016/j.biopsych.2013.11.006 & 2014 Society of Biological Psychiatry
P.S. Pressman and B.L. Miller BIOL PSYCHIATRY 2014;75:574–581 575

The overall prevalence of FTD has likely been underestimated Patients with bvFTD often display obsessive-compulsive
due to misdiagnosis but might range from 15 to 22/100,000 (21). behavior that varies from simple repetitive motions, like foot
The prevalence in Cambridgeshire in the United Kingdom was tapping or pacing, to frequent playing of computer games or
estimated at 15/100,000 in the age group of 45–64 years, similar bizarre rituals. Hoarding is common. Some patients become more
to early-onset AD (4). Among patients with progressive dementia, mentally rigid and resistant to changes in scheduled routines or
the prevalence of a non-Alzheimer’s related frontotemporal plans (31).
atrophy with neuronal loss and gliosis on autopsy has been Eating habits frequently change in patients with bvFTD. Some
estimated at between 12% and 16%, but prevalence varies patients develop a strong preference for sweets or eat beyond
enormously from center to center on the basis of the age of the point of satiety (32). Family members might have to lock
the cohort and the interest of the clinicians and neuropatholo- kitchen cabinets and be wary in grocery stores to stop the patient
gists in FTLD (22). from stealing food. Hyperorality and attempts to eat nonedible
In one study from the State of California AD centers, the objects might also occur.
clinical frequency of FTD in patients with dementia varied across Although patients might exhibit either increased or decreased
different ethnicities, present in 4.7% of Caucasians, 4.2% of Asians activity in the beginning of their disease course, all eventually
and Pacific Islanders, 2.4% in Latinos, and 2.4% in African develop symptoms of apathy and inertia. This might progress to
Americans. Many of these cases were diagnosed during the mutism and immobility in the end stages of the disease.
1990s when FTD was rarely recognized, and the frequencies for Anatomically, apathy is thought to correlate with dysfunction of
all ethnicities are low (23). There might be a slight male brain circuits that involve the right anterior cingulate and caudate
preponderance, although this finding has varied between reports. (30,33).
Neuropsychological testing in patients with bvFTD frequently
reveals executive dysfunction, which often correlates with tissue
Clinical Presentations loss in the dorsolateral prefrontal cortex. Although memory can
be better than in AD, episodic memory can be impaired even in
In 2011 an international consortium developed revised criteria early stages of the disease, which might contribute to misdiag-
for the diagnosis of bvFTD (24) (Table S2 in Supplement 1). The noses of AD (34). By contrast, drawing and other visuospatial
new criteria were designed to increase diagnostic sensitivity functions are often remarkably spared in all of the FTD clinical
compared with previous criteria by Neary et al. (25). In a subtypes.
comparison of the new criteria with histological evidence of In addition, many patients with bvFTD demonstrate changes
FTLD, the sensitivity was 85% and specificity was 95% for with language, including but not restricted to stereotypy of
probable bvFTD (26). speech and echolalia (25). As the disease progresses, semantic
People with bvFTD frequently have limited insight into their deficits and progressive reduction of speech occur. These
illness, making an informant critically important when collecting changes represent a disruption of dominant lobar language
the patient history (27). Family members of patients often do not networks.
initially recognize personality changes as being due to a degen- There are few physical examination findings specific for bvFTD.
erative illness. Patients might present first to psychiatrists or Frontotemporal dementia might be associated with classical
primary care physicians with behavioral symptoms. They might findings of typical Parkinson’s disease, and it is also associated
also present to marriage counselors, to human resources offices in with two atypical parkinsonian disorders, PSP and CBD. The
the work environment, to addiction programs, and to the criminal classical PSP findings of vertical gaze palsy and axial-
system. Symptoms of a global dementia become more apparent predominant parkinsonism might occur early or emerge later in
as the disease progresses. some patients with tau-related FTLD. Similarly, asymmetric par-
Initial personality changes often include social disinhibition kinsonism, alien limb phenomena, hemineglect, and apraxia, the
and impulsivity. Social norms might be infringed upon with corticobasal syndrome, is often associated with FTLD neuro-
offensive remarks or inappropriate behaviors. Patients might lose pathology. Amyotrophic lateral sclerosis accompanies bvFTD in
the ability to empathize with others, be insensitive to the distress approximately 15% of cases (35,36) and can be suggested by the
of someone else, or become distant and detached in their presence of upper and lower motor neuron findings. Both
relationships. Loss of empathy correlated with atrophy in the parkinsonism and motor neuron disease, however, can be caused
right anterior temporal and medial frontal regions in a large-scale by a variety of other disorders that must be taken into
voxel-based morphometry analysis (28). An impaired recognition consideration.
of emotions has been found to correlate with volume loss in the
right lateral inferior temporal and right middle temporal gyri (29).
Patients might become more irritable and might commit Clinicopathological Correlation: Histopathology and
antisocial or even criminal acts. Such acts, however, are usually Genetics
not malevolent but rather poorly thought out or impulsive in
nature, such as driving through a stop sign while being followed Behavioral variant FTD involves atrophy of the orbitofrontal,
by a policeman. In some instances, patients are overly friendly anterior cingulate, anterior insular, and anterior temporal cortices,
and start conversations that are inappropriately explicit or particularly within the right hemisphere (37). Functional studies
personal. Often, these patients are overly trusting and become have increasingly demonstrated network connectivity disruption
susceptible to financial scams. Such behavior might reflect an in addition to modular and structural atrophy. Different degen-
inability to judge potential for reward or punishment. Often, this erative diseases have been associated with unique networks that
correlates with atrophy in the orbitofrontal cortex, a region that might explain particular symptoms. For example, AD has been
has been associated with weighting potential outcomes of future tied to changes in the default mode network, progressive supra-
actions. The subgenual cingulate and medial prefrontal and nuclear palsy with changes beginning in the midbrain, and
anterior temporal cortices might also be involved (30). primary progressive aphasia with dominant hemisphere language

www.sobp.org/journal
576 BIOL PSYCHIATRY 2014;75:574–581 P.S. Pressman and B.L. Miller

networks (38–40). Behavioral variant FTD is associated with bvFTD or nfvPPA in whom progranulin mutations are absent.
changes in the salience network, which comprises the orbital- Type B is typical for FTD with motor neuron disease, and type C is
frontal cortex, anterior cingulate cortex, anterior insula, and present in the vast majority of patients with svPPA (Figures S4–S6
presupplementary motor area. This network is posited to facilitate in Supplement 1).
selection and prioritization of the myriad stimuli presented to us, Mutations in the TDP-43 gene itself rarely cause FTD, usually
many laden with potential rewards or punishments. An executive with ALS. The two more common genetic mutations associated
control network linking dorsolateral frontal and parietal neo- with TDP-43 pathology are progranulin and C9orf72 (chromosome
cortices might then mediate planning and actions on the basis of 9 open reading frame 72) (47,48). Mutations in the progranulin
this salient information (41). gene (GRN) accounted for approximately 8% of all familial forms
Histologically, components of the salience network such as the of FTD. At 62 years, the mean age of onset is slightly higher than
anterior insula and anterior cingulate contain Von Economo other forms of FTD. In contrast to MAPT mutations, GRN mutations
neurons. These spindle-shaped neurons are greatly expanded in usually lead to asymmetric cerebral atrophy and in addition to
number in socially complex animals, including cetaceans, ele- bvFTD might be associated with nfvPPA. Like MAPT mutations,
phants, and the great apes, which has led to the hypothesis that progranulin mutations might lead to parkinsonism (47).
von Economo neurons have evolved in relation to social behav- The GRN mutations cause haploinsufficiency, resulting in levels
iors. Behavioral variant FTD is associated with selective loss of of serum progranulin that are approximately a third of normal
these neurons, the reason for which is a very active area of (49). How low progranulin levels mediate neurodegeneration is
research (42). Furthermore, FTLD is associated with frontal and unknown but is under active study. Mouse models suggest that
anterior temporal atrophy with microvacuolation and astrogliosis low levels of progranulin are associated with decreased neural
most prominent in layers II and III (36,43). connectivity (50), and progranulin seems to play a role as a
Neurodegenerative diseases are characterized by the accu- neuronal growth factor. Additionally, low progranulin levels lead
mulation of abnormal protein deposits on immunohistochemistry. to accelerated inflammation (51,52). In a recent study Miller et al.
Nearly all bvFTD cases fall within one of three histopathological (53) demonstrated that patients with GRN mutations exhibited a
groupings: tau, TDP-43, or FUS (5) (Table S3 in Supplement 1). higher frequency of autoimmune disorders, including sarcoid,
In addition, AD might occasionally mimic FTD with pathology Sjogren syndrome, rheumatoid arthritis, lupus, and chronic
found in the frontal lobes (44). For each proteinopathy, there are lymphocytic colitis and a high peripheral tumor necrosis factor
possible genetic contributions. Genetics might play a role in up to level.
40% of cases, including direct inheritance in at least 10% (45). A noncoding GGGGCC hexanucleotide expansion in the C9
Although there is significant variation between individuals open reading frame is strongly associated with both FTD and ALS
with a mutation, on a population level each genetic variant is (54,55). Aggregates of a dipeptide-repeat protein generated from
associated with slightly different symptoms and patterns of the GGGGCC hexanucleotide are found with C9orf72 mutations in
neurodegeneration. various brain regions including the cerebellum (56,57). C9orf72
mutations account for roughly 50% of familial FTD cases in our
center. Other reports give a range of 13%–26% among familial
Tau FTD cases, compared with 11%–22% for MAPT and 6%–22% for
GRN (58).
Tau is important in the pathogenesis of a variety of neuro-
Many C9orf72 bvFTD patients exhibit bizarre behavior, includ-
degenerative conditions, including AD and FTD. Tau protein
ing obsessive-compulsive behaviors, odd rituals, and psychotic
normally helps to maintain microtubular structure. There are six
features, making this mutation particularly relevant to psychiatric
isoforms of tau, three of which have three microtubule binding
practitioners (59,60). Some relatively young patients with slowly
repeats, and three of which have four repeats. Approximately half
progressive psychiatric symptoms might actually suffer from early
of all patients with bvFTD have tau aggregates: three microtubule
FTD. Clinical testing is now commercially available for C9orf72
binding repeats tau in Pick’s disease, and four microtubule
mutations (60,61). On magnetic resonance imaging (MRI), patients
binding repeats tau in PSP and CBD (Figures S1–S3 in
with the C9orf72 mutation are more likely to have atrophy in
Supplement 1). In addition to bvFTD, tauopathies are a cause of
dorsolateral, medial orbitofrontal, anterior temporal, parietal,
approximately 70% of nfvPPA cases in our center.
occipital, and cerebellar regions, compared with anteromedial
Mutations in the microtubule-associated protein tau gene
temporal atrophy in MAPT gene mutations (46).
(MAPT) have been found in one series to be present in
A smaller proportion of patients with FTD have pathology
approximately 32% of patients with both FTD and a positive
without TDP-43 or tau aggregates (5%). Most of these cases have
family history (20). We have found MAPT mutations to be less
FUS protein deposits (62,63). Age of onset in this population
common, and they account for approximately 17% of familial
tends to be younger (mean 48 years), and they might present
forms of FTD in our center. Over 40 different mutations of the
with psychiatric symptoms.
MAPT gene have been identified. These mutations tend to cause
These distinctive genetic and neuropathological subtypes
bifrontal and anterior temporal atrophy (46).
might demand different treatment options in the future. For
example, because GRN mutations cause a protein deficiency,
Trans-Activator Regulatory DNA Binding Protein 43 studies are underway to elevate levels of that protein in patients
with the mutation (64). As with progranulin mutations, TDP-43
The TDP-43 protein is found in approximately half of bvFTD type C has been associated with an increased risk of autoimmune
cases on histological examination and is seen in all cases of FTD- disorders, suggesting a unique biochemical pathway that might
ALS. There are three major patterns of TDP-43 pathology: Type A, respond to immunomodulation (53). Even while these treatments
Type B, and Type C, which correlate with different forms of FTD. are in development, recognition of the genetic, histologic, and
The FTLD TDP-43 type A is characterized by inclusions that occur syndromic variability of these diseases might help prevent
with progranulin mutations but can be seen in other patients with misdiagnosis.

www.sobp.org/journal
P.S. Pressman and B.L. Miller BIOL PSYCHIATRY 2014;75:574–581 577

Differential Diagnosis Diagnosis of Frontotemporal Dementia


A careful history, combined with laboratory studies and A diagnosis of FTD is made primarily by clinical assessment.
neuroimaging, can usually exclude reversible mimics of FTD such Despite potential diagnostic pitfalls, the presence of abnormal
as neurological infections, metabolic disorders, vascular disease, social conduct, change of eating habits, stereotyped behaviors,
and paraneoplastic conditions. and apathy in the relative absence of memory or visuospatial
Patients with bvFTD might exhibit symptoms suggestive of deficit will usually lead to the correct diagnosis (24).
obsessive-compulsive disorder, bipolar disorder, depression, or A short neuropsychological battery to compare different
schizophrenia. An onset of symptoms in patients in middle age cognitive domains can be an efficient way of aiding the differ-
should lead to consideration of the inclusion of bvFTD in the ential diagnosis. Neuropsychological evaluation should show
differential diagnosis (17). Other neurodegenerative disorders can primarily executive dysfunction, with relative sparing of visuospa-
be mistaken for FTD-spectrum disorders, particularly AD, vascular tial ability. Memory should be relatively spared but might be
dementia, and dementia with Lewy bodies. Both normal pressure involved especially later in the disease. The National Institutes of
hydrocephalus and low intracranial pressure syndromes have Health EXAMINER battery is designed to test executive functions
sometimes been mistaken for FTD, although their imaging and can classify bvFTD and AD with 73% accuracy (70).
features should immediately draw attention to a non-FTD The 2011 bvFTD criteria require neuroimaging data in the form
diagnosis (65). of selective atrophy on MRI or hypometabolism on positron
An entity initially described by John Hodges and Chris Kipps, emission tomography (PET) scan to transform a diagnosis from
“slowly progressive FTD” can be a difficult diagnostic dilemma “possible” into “probable” bvFTD (24). Early in the disease,
(66). These are patients who seem to meet the bvFTD phenotype however, imaging might be normal. The bvFTD patients typically
but progress slowly or not at all and do not show atrophy on MRI. show atrophy in the frontoinsular structures and other regions of
Some of these individuals have FTD, and gene carriers can the salience network (Figure 1). The svPPA patients show atrophy
progress very slowly. In other patients the initial history is at the temporal poles, and nfvPPA patients will show volume loss
misleading or incomplete, and further history-taking clarifies the in the left perisylvian region. Progressive supranuclear palsy
bvFTD diagnosis. Psychiatric disorders can also account for some classically shows midbrain atrophy with relative sparing of the
of these bvFTD phenotypical patients. Although sometimes a pons (46,71). Corticobasal syndrome often demonstrates dorsal
psychiatric syndrome might be misdiagnosed as FTD, in our atrophy around the central sulcus (11). Patterns of atrophy might
experience it is more common to initially diagnose a dementia as also suggest an underlying histological or genetic etiology.
being nondegenerative (67,68). Furthermore, neuroimaging studies might help exclude alterna-
Statistically, someone under the age of 65 is approxi- tive disorders such as vascular dementia or normal pressure
mately as likely to have AD as FTD (4). Early-onset AD hydrocephalus.
frequently does not have the classical amnestic presentation Functional neuroimaging studies such as fluorodeoxyglucose-
of later-onset AD but might instead have prominent execu- PET might be more sensitive than MRI in early stages, and can be
tive dysfunction or language dysfunction (69). Conversely, clinically useful in distinguishing FTD from AD (72). Fluorodeoxy-
memory loss might be a feature of FTD. Interpersonal glucose-PET reveals hypometabolism of frontal, anterior cingulate,
dysfunction is unusual for AD and might be a useful clinical and anterior temporal regions in FTD, in contrast to temporopar-
distinguishing feature of bvFTD. ietal and posterior cingulate hypometabolism in AD (Figure 2).

Figure 1. A T1 magnetic resonance image of the brain in axial, coronal, and sagittal cuts, shown in neurological convention, showing severe anterior
predominant atrophy with hydrocephalus ex vacuo in a patient with advanced behavioral variant frontotemporal dementia.

www.sobp.org/journal
578 BIOL PSYCHIATRY 2014;75:574–581 P.S. Pressman and B.L. Miller

Figure 2. Axial fluorodeoxyglucose positron emission tomography images of a patient with advanced behavioral variant frontotemporal dementia, shown
in neurological convention, demonstrating hypometabolism in the anterior lobes bilaterally but right more so than left. Image courtesy of Pia Ghosh of the
University of California at San Francisco Memory and Aging Center. SUVR, standard uptake values ratios.

The increasing prominence of ligand imaging such as florbetapir discussed early in the disease course (78). Physical safety around
in the diagnosis of AD can help suggest the presence of AD the home should also be considered as well as early retirement if
pathology but should not necessarily be thought to exclude the patient is still working (79).
comorbid frontotemporal dementia. Tau ligands might also Exercise has been shown to benefit mood, cognition, and
become more widely available for research in the near future (73). overall health in patients with dementia (80). Physical therapy
Other functional imaging techniques such as functional MRI might be helpful in patients with mobility problems such as
have shown promise in distinguishing affected systems in parkinsonism, including reducing the risk of falls. Speech therapy
research but have not yet been used in the diagnosis of might have some benefits for patients with language deficits (81).
individuals. Similarly, electroencephalograms have distinguished Problematic behavior can be managed by assessing for causes of
FTD from AD but are not used in routine diagnosis. pain or delirium and might also be handled with a combination of
Cerebrospinal fluid tau levels can be high or low in FTD and redirection, distraction, and offering simple choices. Most patients
cannot help to rule in FTD. A cutoff of 1.06 in the tau/amyloid- benefit from a stable and structured environment. Finally, care-
beta ratio had a 79% sensitivity and 97% specificity in distinguish- giver stress is significant in FTD and should be addressed as early
ing syndromic FTD from AD (74). as possible.
Genetic testing is available for several mutations that cause Pharmacological treatment of neurobehavioral symptoms
familial FTD. Such testing should be done selectively and guided in frontotemporal dementia should ideally suit typical changes
by the family history, clinical syndrome, and results of any in neurotransmitter systems occurring with the disease (82,83). In
pathological studies. Genetic counseling is advisable for those nondemented populations, decreased brain levels of serotonin
who wish to know the results of any testing done (75). are associated with many behavioral problems seen in FTD (84).
Frontotemporal dementia has been shown to have serotonergic
Treatment network disruption on the basis of autopsy, neuroimaging,
and cerebrospinal fluid studies (84–87). Some studies have
The proper treatment of FTD requires an individually tailored suggested that serotonin selective reuptake inhibitors are effec-
approach. Because no treatment is yet available that changes the tive in helping with various symptoms of FTD, including dis-
course of the neurodegenerative disease, the focus of medical inhibition, impulsivity, repetitive behaviors, and eating disorders
therapy is on symptomatic relief (76). The difficult behaviors (88,89). Trazodone has been effective in treating agitation and
typical for bvFTD can place a great deal of stress on caregivers. aggression in FTD and might also function as a sleep aid if
Education and counseling about the neurological basis of the necessary (90).
disease can offer some relief to frustrated friends and family. Dopaminergic function is altered not only in association with
Resources such as The Association for Frontotemporal Degener- extrapyramidal symptoms but might also be related to agitated
ation (http://www.theaftd.org/) might be useful. behavior (88,91). Antipsychotic medications can help with neuro-
Many nonpharmacological interventions can assist the man- behavioral symptoms in FTD (92,93). Because many patients with
agement of a patient with frontotemporal dementia (77). Safety is FTD are predisposed toward parkinsonian symptoms, however,
a serious concern due to possible impulsivity and impaired they are likely vulnerable to extrapyramidal side effects (94). For
judgment. Financial guidance and driving safety should be this reason, antipsychotics are usually used after behavioral

www.sobp.org/journal
P.S. Pressman and B.L. Miller BIOL PSYCHIATRY 2014;75:574–581 579

modification and serotonin selective reuptake inhibitors. Agents for Frontotemporal Lobar Degeneration. Acta Neuropathologica 114:
with relatively less D2 receptor antagonism such as quetiapine 5–22.
are preferred if antipsychotics are used (95). Furthermore, patients 6. Kertesz A, Hillis A, Munoz D (2003): Frontotemporal degeneration,
Pick’s disease, Pick complex, and Ravel. Ann Neurol 54(Suppl 5):S1.
and family members should be cautioned about the increased 7. Rabinovici GD, Miller BL (2010): Frontotemporal lobar degeneration:
mortality associated with antipsychotic use in elderly patients Epidemiology, pathophysiology, diagnosis and management. CNS
with dementia. Drugs 24:375–398.
The cholinergic system is relatively preserved in FTD (96), 8. Gorno-Tempini ML, Hillis AE, Weintraub S, Kertesz A, Mendez M,
unlike AD. Trials have not shown a convincing benefit for Cappa SF, et al. (2011): Classification of primary progressive aphasia
cholinesterase inhibitors in cognition or behavior (97,98). Several and its variants. Neurology 76:1006–1014.
9. Chiu WZ, Papma JM, de Koning I, Donker Kaat L, Seelaar H,
other drugs, including memantine, lithium, and some anticonvul- Reijs AEM, et al. (2012): Midcingulate involvement in progressive
sants, have been tested in FTD with negative or inconclusive supranuclear palsy and tau positive frontotemporal dementia. J
results (99,100). Risks of toxicity are generally deemed to out- Neurol Neurosurg Psychiatry 83:910–915.
weigh theoretical benefits. 10. Arai T, Ikeda K, Akiyama H, Shikamoto Y, Tsuchiya K, Yagishita S, et al.
(2001): Distinct isoforms of tau aggregated in neurons and glial cells in
brains of patients with Pick’s disease, corticobasal degeneration and
Conclusions progressive supranuclear palsy. Acta Neuropathologica 101:167–173.
11. Lee SE, Rabinovici GD, Mayo MC, Wilson SM, Seeley WW, DeArmond
Frontotemporal dementia is increasingly recognized as not SJ, et al. (2011): Clinicopathological correlations in corticobasal
degeneration. Ann Neurol 70:327–340.
only a common cause of early-onset dementia but also a cause of
12. Rankin KP, Mayo MC, Seeley WW, Lee S, Rabinovici G, Gorno-Tempini
manic, depressive, obsessive-compulsive, or psychotic symptoms. ML, et al. (2011): Behavioral variant frontotemporal dementia with
The syndrome of bvFTD can result from a number of different corticobasal degeneration pathology: Phenotypic comparison to
proteinopathies, and some cases have a genetic etiology. Recog- bvFTD with Pick’s disease. J Mol Neurosci 45:594–608.
nizing the core features of bvFTD as well as the variety of possible 13. Lomen-Hoerth C (2011): Clinical phenomenology and neuroimaging
presentations can avoid misdiagnosis. This will permit the correlates in ALS-FTD. J Mol Neurosci 45:656–662.
14. Armstrong RA, Carter D, Cairns NJ (2012): A quantitative study of the
appropriate selection of symptomatic therapies and might iden- neuropathology of 32 sporadic and familial cases of frontotemporal
tify subtypes that might benefit from future therapies. Promising lobar degeneration with TDP-43 proteinopathy (FTLD-TDP). Neuro-
areas of research include the role of von Economo neurons, PET path Appl Neurobiol 38:25–38.
imaging with tau ligands, autoimmune treatments for TDP-C 15. Lee SE, Seeley WW, Poorzand P, Rademakers R, Karydas A, Stanley
disorders, and protein replacement for progranulin mutations. CM, et al. (2012): Clinical characterization of bvFTD due to FUS
neuropathology. Neurocase 18:305–317.
16. Rohrer JD, Lashley T, Schott JM, Warren JE, Mead S, Isaacs AM, et al.
We would like to thank the Tau Consortium, the Consortium for (2011): Clinical and neuroanatomical signatures of tissue pathology
Frontotemporal Dementia Research, and the teams of the Primary in frontotemporal lobar degeneration. Brain 134:2565–2581.
Program Grant and the Alzheimer’s Disease Research Center of the 17. Woolley JD, Khan BK, Murthy NK, Miller BL, Rankin KP (2011): The
Memory and Aging Center of the University of California, San diagnostic challenge of psychiatric symptoms in neurodegenerative
Francisco. This publication was made possible by Grant number disease: Rates of and risk factors for prior psychiatric diagnosis in
patients with early neurodegenerative disease. J Clin Psychiatry 72:
P50AG023501 and Grant number P01AG019724 from the National
126–133.
Institutes of Health National Institute on Aging. Its contents are 18. Johnson JK, Diehl J, Mendez MF, Neuhaus J, Shapira JS, Forman M,
solely the responsibility of the authors and do not necessarily et al. (2005): Frontotemporal lobar degeneration: Demographic
represent the official views of the National Institute on Aging or characteristics of 353 patients. Arch Neurol 62:925–930.
National Institutes of Health. 19. Roberson ED, Hesse JH, Rose KD, Slama H, Johnson JK, Yaffe K, et al.
Dr. Miller is a board member of the Larry L. Hillblom Foundation, (2005): Frontotemporal dementia progresses to death faster than
Alzheimer disease. Neurology 65:719–725.
the John Douglas French Foundation, and the Tau Consortium. Dr. 20. Rosso SM, Donker Kaat L, Baks T, Joosse M, de Koning I, Pijnenburg
Miller has consulted for Tau Rx, Allon Therapeutics, Bristol-Myers Y, et al. (2003): Frontotemporal dementia in The Netherlands: Patient
Squibb, Siemens Molecular Imaging, and Eli Lilly US. He receives characteristics and prevalence estimates from a population-based
funding from the National Institutes of Health/National Institute of study. Brain 126:2016–2022.
Aging. Dr. Miller receives royalties from Cambridge University Press, 21. Onyike CU, Diehl-Schmid J (2013): The epidemiology of frontotem-
Neurocase, and Guilford Publications. poral dementia. Int Rev Psychiatry 25:130–137.
22. Greicius MD, Geschwind MD, Miller BL (2002): Presenile dementia
Dr. Pressman reports no biomedical financial interests or syndromes: An update on taxonomy and diagnosis. J Neurol Neuro-
potential conflicts of interest. surg Psychiatry 72:691–700.
23. Hou C, Yaffe K, Perrez-Stable E, Miller B (2006): Frequency of
Supplementary material cited in this article is available online at dementia etiologies in four ethnic groups. Dement Geriatr Cogn
http://dx.doi.org/10.1016/j.biopsych.2013.11.006. Disord 22:42–47.
24. Rascovsky K, Hodges JR, Knopman D, Mendez MF, Kramer JH,
1. Pick A (1892): Uber die beziehungen der senilen hirnatropie zur Neuhaus J, et al. (2011): Sensitivity of revised diagnostic criteria for
aphasie. Pragen Medizinischen Wochenschrift 17:165–167. the behavioural variant of frontotemporal dementia. Brain 134:
2. Pasquier F, Petit H (1997): Frontotemporal dementia: Its rediscovery. 2456–2477.
Eur Neurol 38:1–6. 25. Neary D, Snowden J, Gustafson L, Passant U, Stuss D, Black S, et al.
3. Brunnstrom H, Gustafson L, Passant U, Englund E (2009): Prevalence (1998): Frontotemporal lobar degeneration: A consensus on clinical
of dementia subtypes: A 30-year retrospective survey of neuro- diagnostic criteria. Neurology 51:1546–1554.
pathological reports. Arch Gerontol Geriatriatr 49:146–149. 26. Harris JM, Gall C, Thompson JC, Richardson AMT, Neary D, Plessis Dd,
4. Ratnavalli E, Brayne C, Dawson K, Hodges JR (2002): The prevalence et al. (2013): Sensitivity and specificity of FTDC criteria for behavioral
of frontotemporal dementia. Neurology 58:1615–1621. variant frontotemporal dementia. Neurology 80:1–7.
5. Cairns NJ, Bigio EH, Mackenzie IR, Neumann M, Lee VM, Hatanpaa KJ, 27. Mendez M, Shapira JS (2005): Loss of insight and functional neuro-
et al. (2007): Neuropathologic diagnostic and nosologic criteria for imaging in frontotemporal dementia. J Neuropsychiatry Clin Neurosci
frontotemporal lobar degeneration: Consensus of the Consortium 17:413–416.

www.sobp.org/journal
580 BIOL PSYCHIATRY 2014;75:574–581 P.S. Pressman and B.L. Miller

28. Rankin KP, Gorno-Tempini ML, Allison SC, Stanley CM, Glenn S, (Arg493X) mutation: An international initiative. Lancet Neurol 6:
Weiner MW, et al. (2006): Structural anatomy of empathy in neuro- 857–868.
degenerative disease. Brain 129:2945–2956. 50. Tapia L, Milnerwood A, Guo A, Mills F, Yoshida E, Vasuta C, et al.
29. Rosen HJ, Wilson MR, Schauer GF, Allison S, Gorno-Tempini ML, (2011): Progranulin deficiency decreases gross neural connectivity
Pace-Savitsky C, et al. (2006): Neuroanatomical correlates of impaired but enhances transmission at individual synapses. J Neurosci 31:
recognition of emotion in dementia. Neuropsychologia 44:365–373. 11126–11132.
30. Rosen H, Allison S, Schauer G, Gorno-Tempini M, Weiner M, Miller B 51. Kao AW, Eisenhut RJ, Martens LH, Nakamura A, Huang A, Bagley JA,
(2005): Neuroanatomical correlates of behavioural disorders in et al. (2011): A neurodegenerative disease mutation that accelerates
dementia. Brain 128:2612–2625. the clearance of apoptotic cells. Proc Natl Acad Sci U S A 108:
31. Perry DC, Whitwell JL, Boeve BF, Pankratz VS, Knopman DS, Petersen 4441–4446.
RC, et al. (2012): Voxel-based morphometry in patients with 52. Ward ME, Miller BL (2011): Potential mechanisms of progranulin-
obsessive-compulsive behaviors in behavioral variant frontotempo- deficient FTLD. J Mol Neurosci 45:574–582.
ral dementia. Eur J Neurol 19:911–917. 53. Miller Z, Rankin K, Graff-Radford N, Takada L, Sturm V, Cleveland C,
32. Wooley J, Gorno-Tempini M, Seeley W, Rankin K, Lee S, Mathews B, et al. (2013): TDP-43 frontotemporal lobar degeneration and auto-
et al. (2007): Binge eating is associated with right orbitofrontal- immune disease. J Neurol Neurosurg Psychiatry.
insular-striatal atrophy in frontotemporal dementia. Neurology 69: 54. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M,
1424–1433. Rutherford NJ, et al. (2011): Expanded GGGGCC hexanucleotide
33. Eslinger PJ, Moore P, Antani S, Anderson C, Grossman M (2012): repeat in noncoding region of C9ORF72 causes chromosome 9p-
Apathy in frontotemporal dementia: Behavioral and neuroimaging linked FTD and ALS. Neuron 72:245–256.
correlates. Behav Neurol 25:127–136. 55. Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs
34. Hodges JR, Davies RR, Xuereb JH, Casey B, Broe M, Bak TH, et al. JR, et al. (2011): A hexanucleotide repeat expansion in C9ORF72 is
(2004): Clinicopathological correlates in frontotemporal dementia. the cause of chromosome 9p21-linked ALS-FTD. Neuron 72:257–268.
Ann Neurol 56:399–406. 56. Ash PE, Bieniek KF, Gendron TF, Caulfield T, Lin WL, Dejesus-
35. Gustafson L (1987): Frontal lobe degeneration of non-Alzheimer Hernandez M, et al. (2013): Unconventional translation of C9ORF72
type. II. Clinical picture and differential diagnosis. Arch Gerontol GGGGCC expansion generates insoluble polypeptides specific to
Geriatr 6:209–223. c9FTD/ALS. Neuron 77:639–646.
36. Brun A (1987): Frontal lobe degeneration of non-Alzheimer type. I. 57. Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, et al.
Neuropathology. Arch Gerontol Geriatr 6:193–208. (2013): The C9orf72 GGGGCC repeat is translated into aggregating
37. Seeley W, Crawford R, Rascovsky K, Kramer J, Weiner M, Miller B, dipeptide-repeat proteins in FTLD/ALS. Science 339:1335–1338.
et al. (2008): Frontal paralimbic network atrophy in very mild 58. Majounie E, Abramzon Y, Renton AE, Perry R, Bassett SS, Pletnikova
behavioral variant frontotemporal dementia. Arch Neurol 65: O, et al. (2012): Repeat expansion in C9ORF72 in Alzheimer’s disease.
249–255. N Engl J Med 366:283–284.
38. Seeley WW, Crawford RK, Zhou J, Miller BL, Greicius MD (2009): 59. Sha SJ, Takada LT, Rankin KP, Yokoyama JS, Rutherford NJ, Fong JC,
Neurodegenerative diseases target large-scale human brain net- et al. (2012): Frontotemporal dementia due to C9ORF72 mutations:
works. Neuron 62:42–52. Clinical and imaging features. Neurology 79:1002–1011.
39. Gardner RC, Boxer AL, Trujillo A, Mirsky JB, Guo CC, Gennatas ED, 60. Snowden JS, Rollinson S, Thompson JC, Harris JM, Stopford CL,
et al. (2013): Intrinsic connectivity network disruption in progressive Richardson AM, et al. (2012): Distinct clinical and pathological
supranuclear palsy. Ann Neurol 73:603–616. characteristics of frontotemporal dementia associated with
40. Greicius MD, Srivastava G, Reiss AL, Menon V (2004): Default-mode C9ORF72 mutations. Brain 135:693–708.
network activity distinguishes Alzheimer’s disease from healthy 61. Khan BK, Yokoyama JS, Takada LT, Sha SJ, Rutherford NJ, Fong JC,
aging: Evidence from functional MRI. Proc Natl Acad Sci U S A 101: et al. (2012): Atypical, slowly progressive behavioural variant fronto-
4637–4642. temporal dementia associated with C9ORF72 hexanucleotide expan-
41. Seeley WW, Menon V, Schatzberg AF, Keller J, Glover GH, Kenna H, sion. J Neurol Neurosurg Psychiatry 83:358–364.
et al. (2007): Dissociable intrinsic connectivity networks for salience 62. Neumann M, Rademakers R, Roeber S, Baker M, Kretzschmar HA,
processing and executive control. J Neurosci 27:2349–2356. Mackenzie IRA (2009): A new subtype of frontotemporal lobar
42. Seeley WW, Carlin DA, Allman JM, Macedo MN, Bush C, Miller BL, degeneration with FUS pathology. Brain 132:2922–2931.
et al. (2006): Early frontotemporal dementia targets neurons unique 63. Urwin H, Josephs KA, Rohrer JD, Mackenzie IR, Neumann M, Authier
to apes and humans. Ann Neurol 60:660–667. A, et al. (2010): FUS pathology defines the majority of tau- and TDP-
43. McKhann GM, Albert MS, Grossman M, Miller B, Dickson D, 43-negative frontotemporal lobar degeneration. Acta Neuropatho-
Trojanowski JQ, et al. (2001): Clinical and pathological diagnosis of logica 120:33–41.
frontotemporal dementia: Report of the Work Group on Frontotem- 64. Cenik B, Sephton CF, Dewey CM, Xian X, Wei S, Yu K, et al. (2011):
poral Dementia and Pick’s Disease. Arch Neurol 58:1803–1809. Suberoylanilide hydroxamic acid (vorinostat) up-regulates progranu-
44. Alladi S, Xuereb J, Bak T, Nestor P, Knibb J, Patterson K, et al. (2007): lin transcription: Rational therapeutic approach to frontotemporal
Focal cortical presentations of Alzheimer’s disease. Brain 130: dementia. J Biol Chem 286:16101–16108.
2636–2645. 65. Wicklund MR, Mokri B, Drubach DA, Boeve BF, Parisi JE, Josephs KA
45. Goldman JS, Rademakers R, Huey ED, Boxer AL, Mayeux R, Miller BL, (2011): Frontotemporal brain sagging syndrome: An SIH-like pre-
et al. (2011): An algorithm for genetic testing of frontotemporal lobar sentation mimicking FTD. Neurology 76:1377–1382.
degeneration. Neurology 76:475–483. 66. Kipps CM, Hodges JR, Hornberger M (2010): Nonprogressive behav-
46. Boeve BF, Boylan KB, Graff-Radford NR, DeJesus-Hernandez M, ioural frontotemporal dementia: Recent developments and clinical
Knopman DS, Pedraza O, et al. (2012): Characterization of fronto- implications of the ‘bvFTD phenocopy syndrome’. Curr Opin Neurol
temporal dementia and/or amyotrophic lateral sclerosis associated 23:628–632.
with the GGGGCC repeat expansion in C9ORF72. Brain 135:765–783. 67. Davies R, Kipps C, Mitchell J, Kril J, Halliday G, Hodges J (2006):
47. Baker M, Mackenzie IR, Pickering-Brown SM, Gass J, Rademakers R, Progression in frontotemporal dementia: Identifying a benign
Lindholm C, et al. (2006): Mutations in progranulin cause tau- behavioral variant by magnetic resonance imaging. Arch Neurol 63:
negative frontotemporal dementia linked to chromosome 17. Nature 1627–1631.
442:916–919. 68. Woolley J, Wilson M, Hung E, Gorno-Tempini M, Miller B, Shim J
48. Cruts M, Gijselinck I, van der Zee J, Engelborghs S, Wils H, Pirici D, (2007): Frontotemporal dementia and mania. Am J Psychiatry 164:
et al. (2006): Null mutations in progranulin cause ubiquitin-positive 1811–1816.
frontotemporal dementia linked to chromosome 17q21. Nature 442: 69. Mendez MF, Lee AS, Joshi A, Shapira JS (2012): Nonamnestic
920–924. presentations of early-onset Alzheimer’s disease. Am J Alzheimers
49. Rademakers R, Baker M, Gass J, Adamson J, Huey E, Mommeni P, Dis Other Demen 27:413–420.
et al. (2007): Phenotypic variability associated with progranulin 70. Possin KL, Feigenbaum D, Rankin KP, Smith GE, Boxer AL,
haploinsufficiency in patients with the common 1477C-T Wood K, et al. (2013): Dissociable executive functions in behavioral

www.sobp.org/journal
P.S. Pressman and B.L. Miller BIOL PSYCHIATRY 2014;75:574–581 581

variant frontotemporal and Alzheimer dementias. Neurology 80: 86. Procter A, Qurne M, Francis P (1999): Neurochemical features of
2180–2185. frontotemporal dementia. Dement Geriatr Cogn Disord 10(Suppl 1):80–84.
71. Massey L, Miscallef C, Paviour D, O’Sullivan S, Ling H, Williams D, 87. Franceschi M, Anchisi D, Pelati O, Zuffi M, Matarrese M, Moresco RM,
et al. (2012): Conventional magnetic resonance imaging in con- et al. (2005): Glucose metabolism and serotonin receptors in the
firmed progressive supranuclear palsy and multiple system atrophy. frontotemporal lobe degeneration. Ann Neurol 57:216–225.
Mov Disord 27:1754–1762. 88. Manoochehri M, Huey ED (2012): Diagnosis and management of
72. Womack KB, Diaz-Arrastia R, Aizenstein HJ, Arnold SE, Barbas NR, behavioral issues in frontotemporal dementia. Curr Neurol Neurosci
Boeve BF, et al. (2011): Temporoparietal hypometabolism in fronto- Report 12:528–536.
temporal lobar degeneration and associated imaging diagnostic 89. Swartz J, Miller B, Lesser I, Darby AL (1997): Frontotemporal
errors. Arch Neurol 68:329–337. dementia: Treatment response to serotonin selective reuptake
73. Maruyama M, Shimada H, Suhara T, Shinotoh H, Ji B, Maeda J, et al. inhibitors. J Clin Psychiatry 58:212–216.
(2013): Imaging of tau pathology in a tauopathy mouse model and 90. Lebert F, Stekke W, Hasenbroekz C, Pasquier F (2004): Frontotem-
in Alzheimer patients compared to normal controls. Neuron 79: poral dementia: A randomised, controlled trial with trazodone.
1094–1108. Dement Geriatr Cogn Disord 17:355–359.
74. Bian H, Swieten JV, Leight S, Massimo L, Wood E, Forman M, et al. 91. Engelborghs S, Vloeberghs E, Le Bastard N, Van Buggenhout M,
(2008): CSF biomarkers in frontotemporal lobar degeneration with Marien P, Somers N, et al. (2008): The dopaminergic neurotransmit-
known pathology. Neurology 70:1827–1835. ter system is associated with aggression and agitation in fronto-
75. Goldman JS, Adamson J, Karydas A, Miller BL, Hutton M (2007): New temporal dementia. Neurochem Int 52:1052–1060.
genes, new dilemmas: FTLD genetics and its implications for 92. Moretti R, Torre P, Antonello R, Cazzato G, Griggio S, Bava A (2003):
families. Am J Alzheimers Dis Other Demen 22:507–515. Olanzapine as a treatment of neuropsychiatric disorders of Alz-
76. Perry R, Miller B (2001): Behavior and treatment in frontotemporal heimer’s disease and other dementias: A 24-month follow-up of 68
dementia. Neurology 56:S46–S51. patients. Am J Alzheimers Dis Other Demen 18:205–214.
77. Merrilees J (2007): A model for management of behavioral symp- 93. Fellgiebel A, Müller M, Hiemke C, Bartenstein P, Schreckenberger M
toms in frontotemporal lobar degeneration. Alzheimer Dis Assoc (2007): Clinical improvement in a case of frontotemporal dementia
Disord 21:S64–S69. under aripiprazole treatment corresponds to partial recovery of
78. Piguet O, Hornberger M, Mioshi E, Hodges J (2011): Behavioural- disturbed frontal glucose metabolism. World J Biol Psychiatry 8:123–126.
variant frontotemporal dementia: Diagnosis, clinical staging, and 94. Pijnenburg Y, Sampson E, Harvey R, Fox N, Rossor M (2003):
management. Lancet Neurol 10:162–172. Vulnerability to neuroleptic side effects in frontotemporal lobar
79. Talerico K, Evans L (2001): Responding to safety issues in fronto- degeneration. Int J Geriatr Psychiatry 18:67–72.
temporal dementias. Neurology 56:S52–S55. 95. Komossa K, Rummel-Kluge C, Schmid F, Hunger H, Schwarz S,
80. Cheng ST, Chow PK, Song YQ, Yu EC, Chan AC, Lee TM, Lam JH Srisurapanont M, et al. (2010): Quetiapine versus other atypical
(2013): Mental and physical activities delay cognitive decline in older antipsychotics for schizophrenia. Cochrane Database Syst Rev
persons with dementia [published online ahead of print February 6]. CD006625.
Am J Geriatr Psychiatry. 96. Hansen L, Deteresa R, Tobias H, Alford M, Terry R (1988): Neocortical
81. Robinson K (2001): Rehabilitation applications in caring for patients morphometry and cholinergic neurochemistry in Pick’s disease. Am J
with Pick’s disease and frontotemporal dementias. Neurology 56:S56. Pathol 131:507–518.
82. Huey E, Putnam K, Grafman J (2006): A systematic reviw of neuro- 97. Hu B, Ross L, Neuhaus J, Knopman D, Kramer J, Boeve B, et al. (2010):
transmitter deficits and treatments in frontotemporal dementia. Off-label medication use in frontotemporal dementia. Am J Alz-
Neurology 66:17–22. heimers Dis Other Demen 25:128–133.
83. Bowen DM, Procter AW, Mann DM, Snowden JS, Esiri MM, Neary D, 98. Mendez M, Shapira J, MucMurtray A, Licht E (2007): Preliminary
Francis PT (2008): Imbalance of a serotonergic system in frontotem- findings: Behavioral worsening on donepezil in patients with
poral dementia: Implication for pharmacotherapy. Psychopharmacol- frontotemporal dementia. Am J Geriatr Psychiatry 15:84–87.
ogy 196:603–610. 99. Boxer AL, Knopman DS, Kaufer DI, Grossman M, Onyike C, Graf-
84. Sparks D, Markesbery W (1991): Altered serotonergic and cholinergic Radford N, et al. (2013): Memantine in patients with frontotemporal
synaptic markers in Pick’s disease. Arch Neurol 48:796–799. lobar degeneration: A multicentre, randomised, double-blind,
85. Engelborghs S, Vloeberghs E, Maertens K, Marescau B, De Deyn PP placebo-controlled trial. Lancet Neurol 12:149–156.
(2004): Evidence for an association between the CSF HVA:5-HIAA 100. Freedman M (2007): Frontotemporal dementia: Recommendations
ratio and aggressiveness in frontotemporal dementia but not in for therapeutic studies, designs, and approaches. Can J Neurol Sci
Alzheimer’s disease. J Neurol Neurosurg Psychiatry 75:1080. 34(Suppl 1):S118–S124.

www.sobp.org/journal

You might also like