Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

739818

research-article2017
TPP0010.1177/2045125317739818Therapeutic Advances in PsychopharmacologyJ. Young et al.

Therapeutic Advances in Psychopharmacology Review

Frontotemporal dementia: latest evidence


Ther Adv Psychopharmacol

2018, Vol. 8(1) 33­–48

and clinical implications DOI: 10.1177/


https://doi.org/10.1177/2045125317739818
https://doi.org/10.1177/2045125317739818
2045125317739818

© The Author(s), 2017.


Reprints and permissions:
Juan Joseph Young, Mallika Lavakumar, Deena Tampi, Silpa Balachandran http://www.sagepub.co.uk/
journalsPermissions.nav
and Rajesh R. Tampi

Abstract
Background: Frontotemporal dementia (FTD) describes a cluster of neurocognitive syndromes
that present with impairment of executive functioning, changes in behavior, and a decrease
in language proficiency. FTD is the second most common form of dementia in those younger
than 65 years and is expected to increase in prevalence as the population ages. This goal
in our review is to describe advances in the understanding of neurobiological pathology,
classification, assessment, and treatment of FTD syndromes.
Methods: PubMed was searched to obtain reviews and studies that pertain to advancements in
genetics, neurobiology, neuroimaging, classification, and treatment of FTD syndromes. Articles
were chosen with a predilection to more recent preclinical/clinical trials and systematic reviews.
Results: Recent reviews and trials indicate a significant advancement in the understanding
of molecular and neurobiological clinical correlates to variants of FTD. Genetic and
histopathologic markers have only recently been discovered in the past decade. Current
therapeutic modalities are limited, with most studies reporting improvement in symptoms
with nonpharmacological interventions. However, a small number of studies have reported
improvement of behavioral symptoms with selective serotonin reuptake inhibitor (SSRI)
treatment. Stimulants may help with disinhibition, apathy, and risk-taking behavior.
Memantine and cholinesterase inhibitors have not demonstrated efficacy in ameliorating
FTD symptoms. Antipsychotics have been used to treat agitation and psychosis, but safety
concerns and side effect profiles limit utilization in the general FTD population. Nevertheless,
recent breakthroughs in the understanding of FTD pathology have led to developments in
pharmacological interventions that focus on producing treatments with autoimmune, genetic,
and molecular targets.
Conclusion: FTD is an underdiagnosed group of neurological syndromes comprising multiple
variants with distinct neurobiological profiles and presentations. Recent advances suggest
there is an array of potential novel therapeutic targets, although data concerning their
effectiveness are still preliminary or preclinical. Further studies are required to develop Correspondence to:
pharmacological interventions, as there are currently no US Food and Drug administration Rajesh R. Tampi
MetroHealth Medical
approved treatments to manage FTD syndromes. Center, Case Western
Reserve University
School of Medicine,
2500 MetroHealth Drive,
Keywords:  classification, frontotemporal dementia, frontotemporal lobar degeneration, Cleveland, OH 44109, USA
genetics, neurobiology, treatment rajesh.tampi@gmail.com
Juan Joseph Young
Mallika Lavakumar
Received: 21 February 2017; revised manuscript accepted: 26 September 2017. Silpa Balachandran
Department of Psychiatry,
MetroHealth Medical
Center, Cleveland, OH,
USA Case Western
Reserve University,
Introduction executive functioning, behavior, and language. It Cleveland, OH, USA
Frontotemporal dementia (FTD) is a term used is considered the third most common form of Deena Tampi
to describe a group of neurocognitive disorders dementia following Alzheimer’s disease (AD) and Mercy Regional Medical
Center, 3700 Kolbe Rd,
that encompass progressive dysfunction in dementia with Lewy bodies.1 As per its namesake, Lorain, OH 44053, USA.

journals.sagepub.com/home/tpp 33
Therapeutic Advances in Psychopharmacology 8(1)

it is a cluster of syndromes that result from degen- estimated that the prevalence of FTD between
eration of the frontal and temporal lobes, and is the ages of 45–65 years ranged from 15 to 22 per
subdivided into two categories that are unique in 100,000 people, with incidence estimates rang-
respect to their predominating presentations; ing from 2.7 to 4.1 per 100,000 members in the
namely, the behavioral subtype that accounts for same age range. Overall, they estimated approxi-
about half of FTD cases, and the language sub- mately 20,000 to 30,000 cases of FTD in the
type.2 The language subtype is further subdivided USA alone. Yet, these numbers may actually be
into nonfluent and semantic variants of primary underestimating the occurrence of FTD syn-
progressive aphasia (PPA), which are character- dromes. This is especially evident, as older cases
ized by diverging localizations and underlying of FTD begin to echo symptoms of AD further
cerebral dysfunction.2,3 The semantic and nonflu- confounding the delineation between the neuro-
ent variants of PPA are both characterized by cognitive disorders.6 Further emphasizing the
prominent language impairment, but also differ limitations of FTD epidemiologic studies,
in neurological localization, severity of specific Lambert and colleagues8 conducted a systematic
symptoms, and the overall course of their presen- review with the goal of estimating prevalence
tations. Illustrative of this, the semantic variant rates of early-onset dementia. They found a sig-
exhibits bilateral anterior temporal lobe atrophy nificant disparity between studies estimating the
and is associated with dysfunction of emotional prevalence of early-onset FTD with estimates
processing, compulsions, and decline in language ranging from 1.0 to 15.4 per 100,000 members
skills. In contrast, the nonfluent variant com- of the population. Adding to this, it appears that
monly presents with greater left hemisphere atro- the data presented was principally collected from
phy, and is associated with speech problems populations studied in the UK, mainland Europe,
earlier while behavioral disturbances develop later and Japan, suggesting that the information gath-
in the disease course.3 As the population of geri- ered may not be generalizable to the world-wide
atric patients grow and neurocognitive disorders population as a whole. For this reason, standard-
become more prevalent, there will be an increased ization of study designs will likely aid in more
need for physicians with an expert understanding precisely defining FTD epidemiology globally, a
of the diverse clinical findings that define the het- focus that future studies should pursue in the
erogeneous FTD subtypes. This review will focus future.
on the most recent findings concerning FTD
neurobiology, current classification and assess-
ment systems, and the most up-to-date expert Classification and assessment systems
consensus on the treatment of this unique collec- Researchers have developed multiple methodol-
tion of syndromes. ogies to classify FTD disorders using biological
and molecular signatures. However, diagnosis is
still obtained clinically due to the absence of
Epidemiology definitive biomarkers. The international consen-
According to the World Health Organization sus criteria for the behavioral variant of FTD
(WHO), there are an estimated 47.5 million peo- (bvFTD) was published by the International
ple who have dementia with another 7.7 million Behavioral Variant FTD Criteria Consortium9
new cases every year.4 Although AD is the most to develop a sensitive standard for early screen-
common form of dementia, contributing to 60– ing and management of bvFTD. To be diag-
70% of cases, mixed types exist, and the overlap nosed, a patient must show progressive
between different forms of dementia is sizeable. deterioration of behavior or cognition by obser-
FTD is the second most common cause of vation or history provided by a reliable caretaker
dementia in patients aged < 65 years,5 with a fur- or informant in order to be diagnosed. Possible
ther 25% of dementia cases in patients older than bvFTD is diagnosed if three or more of the fol-
65 years also attributed to FTD disorders.6 lowing are present: (a) early behavioral disinhi-
However, determining the total population with bition described as socially inappropriate
underlying FTD pathology is difficult due to the behavior, loss of manners/decorum, impulsivity
low disease frequency in a relatively large at-risk and rash actions; (b) early apathy or inertia; (c)
population. Additionally, studies estimating inci- early loss of empathy or sympathy, including
dence and prevalence rates of FTD are limited diminished response to other people’s need
by the inherent difficulty identifying FTD disor- or sympathies, and diminished social interest;
ders.6,7 In the USA, Knopman and Roberts7 (d) early perseverative or compulsive/ritualistic

34 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

behavior (i.e. simple repetitive movements, knowledge; (b) surface dyslexia (i.e. inability to
complex, compulsive, or ritualistic behavior, ste- recognize words as a whole) or dysgraphia; (c)
reotypy of speech); (e) hyperorality and dietary spared repetition; (d) spared speech production.
changes, including altered food preferences, For an imaging-supported diagnosis, there must
binge eating, increased consumption of alcohol either be predominant anterior temporal lobe
and cigarettes, oral exploration or consumption atrophy or predominant anterior temporal hypop-
of inedible objects; (f) neuropsychological pro- erfusion/hypometabolism on SPECT or PET.
file demonstrates deficits in executive function- For a definite pathology diagnosis, there must be
ing and relative sparing of episodic memory and histopathologic evidence or the presence of a
visuospatial functioning. Probable bvFTD diag- known pathogenic mutation.
nosis would need to meet criteria for possible
bvFTD, but also includes imaging suggestive of The nonfluent variant of PPA (nfvPPA) diagno-
frontal or anterior atrophy on magnetic reso- sis requires at least one of two core features
nance imaging (MRI) or computed tomography (agrammatism in language production or effort-
(CT). Alternatively, positron emission tomogra- ful, halting speech not consistent with apraxia of
phy (PET) or single-photon emission computed speech), and two out of three of the following
tomography (SPECT) imaging demonstrating features: (a) impaired comprehension of complex
hypoperfusion or hypometabolism in the frontal sentences; (b) spared single-word comprehen-
or anterior temporal regions could be used to sion; (c) spared object knowledge. Imaging-
diagnose probable bvFTD. Definitive bvFTD supported nfvPPA variant diagnosis is obtained
with definite FTD pathology is diagnosed when when there is imaging that demonstrates pre-
a patient meets criteria for possible bvFTD and dominant left posterior-fronto-insular atrophy
has one or both of the following: histopathologi- on MRI or predominantly left posterior fronto-
cal evidence of FTD or if there is evidence of a insular hypoperfusion or hypometabolism on
known pathogenic mutation. bvFTD is excluded SPECT or PET. Alternatively, nfvPPA with defi-
if a patient’s presentation is better accounted for nite pathology diagnosis is present when there is
by other nondegenerative nervous system or histopathologic evidence or the presence of a
medical disorders, a psychiatric diagnosis, or known pathogenic mutation.
biomarkers are strongly indicative of other neu-
rocognitive disorders or neurodegenerative pro- Like other PPA subtypes, individuals with the
cesses such as AD.9 logopenic variant of PPA exhibit language impair-
ment but with more specific difficulties in con-
Gorno-Tempini and colleagues developed a com- frontation naming and syntax. These individuals
mon framework for the classification of PPA sub- are more likely to have speech interrupted with
types to provide consistency in clinical diagnosis filling sounds, frequent pauses, phonological
and standardize PPA assessments for future clini- errors, and impaired sentence repetition.11 The
cal studies.10 According to this framework, PPA is diagnosis of logopenic variant PPA requires the
diagnosed when a patient exhibits prominent dif- following core features to be present: impaired
ficulty with language wherein the deficits are the single-word retrieval in spontaneous speech and
principal cause of impairment of daily activities, naming, and impaired repetition of sentences and
with aphasia being the most prominent deficit at phrases. Additionally, three of four of the follow-
symptom onset and during the initial phase of the ing features must also be present for a diagnosis:
disease. PPA diagnosis is excluded if the present- (a) speech (phonologic) errors in spontaneous
ing symptoms are more consistent with other speech and naming; (b) spared single-word com-
neurocognitive disorders, medical conditions, prehension and object knowledge; (c) spared
neurodegenerative processes, or a psychiatric motor speech; (d) absence of frank agrammatism.
diagnosis. PPA is also excluded if there are prom- As with other PPA-variant diagnoses, imaging-
inent initial episodic memory, visual memory, supported diagnosis is made with corresponding
visuospatial impairments, and initial behavioral abnormalities in imaging. In the logopenic vari-
disturbance. Patients may be diagnosed with the ant, MRI, SPECT, or PET scans must demon-
semantic variant of PPA (svPPA) if they exhibit strate predominant left posterior perisylvian or
impaired confrontation naming (i.e. difficulty parietal atrophy/hypoperfusion/hypometabolism.
naming or recognizing objects or drawings) and Definite histopathologic evidence or the presence
impaired single-word comprehension, and have of a known pathogenic mutation would allow for
at least three of the following: (a) impaired object a definite pathology specifier.

journals.sagepub.com/home/tpp 35
Therapeutic Advances in Psychopharmacology 8(1)

Familial frontotemporal dementia caused predominance of early language involvement


by frontotemporal dementia-associated compared with MAPT or C9orf72 mutations.
mutations Extrapyramidal symptoms occur between 40%
FTD is a highly heritable disorder despite varying and 60% of PGRN-associated FTD cases.57,60
heritability among different clinical syndromes Interestingly, PGRN mutations, though thought
and subtypes due to a range of gene mutations.12 to be pathogenic, do not appear to increase the
Up to half of FTD cases with autosomal-dominant risk of developing FTLD.54
inheritance report a family history of FTD.13
Familial FTD also accounts for approximately one FTD patients with the C9orf72 expansion muta-
third to half of all FTD cases and presents more tion present more commonly as bvFTD, motor
commonly as bvFTD than other FTD sub- neuron disease, or a combination of the two,
types.6,14 Mutations in microtubule-associated although clinical presentations tend to be hetero-
protein tau (MAPT),15–25 progranulin (PGRN),26–38 geneous early in the disease process.41,61 Apathy,
and chromosome 9 open reading frame 72 disinhibition, and loss of empathy are prominent
(C9orf72) expansion mutations32,39–50 have been behavioral features in this type of familial FTD.62
found to be the most common causes of familial Complex stereotyped behaviors are common, but
FTD.51,52 Although FTD presentations are rela- language decline is rarer compared with patients
tively homogenous early in the disease course, with MAPT or PGRN mutations. Patients typi-
different biological correlates and varying genetic cally have profound disruption of executive func-
mutations ultimately result in diverging clinical tioning, may have reduced spontaneous speech,
courses.53 MAPT-associated familial FTD typi- echolalia, perseveration, impaired visual and ver-
cally presents younger than FTD associated with bal episodic memory, apraxia, anomia, and dys-
other mutations, and presents as bvFTD with or calculia.63 C9orf72 expansion mutations exhibit
without Parkinsonian symptoms, as well as with an autosomal-dominant pattern of inheritance, is
or without language decline. MAPT mutations implicated in disease anticipation that affect suc-
occur on average at a frequency of 6–11% of all cessive generations,44 and is considered the most
FTD subjects, is inherited in an autosomal-dom- common cause of ALS-FTD (amyotrophic lateral
inant manner, has high penetrance, and may have sclerosis associated with frontotemporal dysfunc-
a shorter duration of illness relative to other muta- tion).42,43,49 However, the minimum repeat length
tions.17,54,55 The most prominent behavioral fea- required to indicate increased risk of disease is
tures with MAPT mutations include disinhibition, unknown.47 These expansion mutations are
stereotyped repetitive behaviors, and obsessions. highly associated with transactive response DNA-
Notably, apathy is less common than in PGRN binding protein (TDP-43) pathology, and the
and C9orf72 cases. Semantic impairment is more presence of neuronal cytoplasmic and intranu-
common later during the disease course. Episodic clear inclusions that are immunoreactive to ubiq-
memory loss may occur and mimic early onset uitin proteasome system markers. A confounding
AD.56 MAPT mutations have been implicated in factor to consider when diagnosing this subtype is
the formation of hyperphosphorylated tau in cor- the possibility of AD-like biomarkers present in
tical and subcortical grey and white matter sug- the cerebrospinal fluid.60 Psychiatric presenta-
gesting a neuroanatomical correlation in the tions, including delusions, auditory/visual/tactile
development of clinical symptoms.55 hallucinations, somatoform symptoms, agitation,
and anxiety may be observed. Parkinson-like
PGRN mutations present with more variable symptoms are common and may present as gait
clinical presentations, but most commonly pre- disturbance, tremor, akinesis, and rigidity. Most
sent as bvFTD and less commonly as a nfvPPA patients do not respond to levodopa when
presentation.57 Mutations occur at a frequency of administered.60,62
5–20% in the FTD population, are inherited in
an autosomal-dominant manner, and have rela- Two other studied pathognomonic mutations
tively low penetrance until the age of 70.17,58,59 found in a minority of cases include mutations in
Apathy and social withdrawal are prominent in vasolin-containing protein (VCP) and transactive
their presentation, and 10–30% of cases may pre- DNA-binding protein (TARDBP).64 VCP muta-
sent with episodic memory impairment.60 tions have been implicated in ubiquitin–proteas-
Neuropsychiatric symptoms are common and ome pathway dysfunction, disruption of TDP-43
patients may report delusions, hallucinations, or localization, and inducing neuronal apoptosis.65
exhibit ritualistic behaviors. There is also a Individuals with VCP mutations historically

36 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

present as the triad of autosomal-dominant tau tangles similar to those found in AD.86 The
hereditary inclusion body myopathy with Paget’s tauopathy present in this variant involves hyper-
disease of the bone and frontotemporal dementia phosphorylated tau protein that forms insoluble
(IBMPFD).66–69 FTD symptoms typically include filamentous inclusions, which adversely affect
behavioral changes, executive dysfunction, and axonal transport regulation, microtubule forma-
aphasia.70 Consequently, a number of European tion, and microtubule stabilization. These
and Asian cases detailing progressive myopathy changes lead to a decrease in microtubule binding
with signs and symptoms consistent with FTD and an increase in tau aggregation, the latter of
have been associated with VCP mutations.71–77 Of which induces neurotoxicity and nerve cell dys-
particular note is one study78 that investigated function.87 Tau aggregates in FTLD-tau subtypes
how the location of VCP gene mutations in 27 vary in the degree of phosphorylation and the
families related to the progression and severity of amount of tau isoforms they contain. This has
symptoms. Critically, the investigators found that prompted their subclassifications into predomi-
the onset of neurocognitive deterioration fore- nantly 3R or 4R (e.g. corticobasal degeneration,
shadowed a rapid progression of the disease lead- progressive supranuclear palsy, argyrophilic grain
ing to an average life span of 6 years. disease, globular glial tauopathy) tau isoform dis-
orders.59,88 These subtypes of FTLD tauopathies
Alternatively, TARDBP was found to be a major also have distinct anatomical and histopathologi-
component of ubiquitin-positive and tau-negative cal correlates. In general, FTD is associated with
inclusions in patients that have been diagnosed severe neuronal pathology, and spherical argyro-
with both amyotrophic lateral sclerosis (ALS) and philic neuronal cytoplasmic inclusions composed
FTD.79 TARDBP has also been implicated in primarily of 3R tau isoforms.64 Progressive supra-
both behavioral and semantic FTD cases with co- nuclear palsy (PSP), a movement disorder that
occurring motor neuron disease (MND).80 presents with rigidity, bradykinesia, and ophthal-
However, other studies have reported cases of moplegia, is characterized by significant degener-
individuals with TARDBP mutations that have ation of subcortical regions including the
developed FTD without the progression of ALS subthalamic nucleus, midbrain, substantia nigra,
or MND symptoms.81,82 Clinical presentations of globus pallidus, and striatum. Postmortem exam-
patients with TARDBP mutations are highly het- inations of PSP patients demonstrate spherical
erogeneous and may present with apathy, poor and flame-shaped neurofibrillary tangles, tufted
executive functioning, poor language comprehen- astrocytes, and inclusions composed predomi-
sion, or disinhibition.83 It has been suggested that nantly of the 4R tau isoform.89 FTD tauopathies
mutations of TARDBP have led to increased may also exhibit the corticobasal syndrome,
translocation of TDP-43 causing dysregulation of which may present as rigidity, bradykinesia, dys-
neuronal endoplasmic reticulum calcium signal- tonia, apraxia, and alien limb phenomenon, is
ing resulting in an increase in B-cell lymphoma 2 characterized by substantia nigra depigmenta-
(Bcl-2) mediated neuronal apoptosis.84 tion, globus pallidus atrophy, and asymmetric
and focal cortical atrophy. This syndrome
exhibits similar traits compared with PSP, but
Neurobiology develop more white matter involvement and has
FTD is classified into distinct, major subtypes unique ring-shaped collections called astrocytic
based on their respective protein-based inclusions plaques caused by tau accumulation in
and underlying molecular pathologies. Namely, astrocytes.59,89
they are separated into the tau-based frontotem-
poral lobar degeneration (FTLD)-tau subtype, Alternatively, neuronal inclusions immunoreac-
the FTLD-TDP subtype associated with TDP- tive to ubiquitin are characteristic of the FTLD-
43, the FTLD-FET subtype that is associated TDP and FTLD-FET forms.85,90 In FTLD-TDP,
with the fused in sarcoma (FUS) proteinopathy these neuronal inclusions are primarily composed
and other FET [i.e. FUS, EWSR1 (EWS RNA- of TDP-43, which has been associated with mul-
binding protein 1), TAF15 (TATA-box binding tiple genetic markers thought to be pathogno-
protein associated factor 15)] family proteins, and monic of FTD. These include mutations of
FTLD-ubiquitin–proteasome system (UPS).59,85 PGRN, C9orf72, and VCP.86,91 In mice, accu-
(See Table 1 for a summary of FTD tauopathies) mulation of the insoluble, phosphorylated cyto-
FTLD-tau accounts for approximately 40% of plasmic TDP-43 and loss of nuclear TDP-43 is
all FTD cases, and is characterized by misfolded associated with brain atrophy, muscle

journals.sagepub.com/home/tpp 37
Therapeutic Advances in Psychopharmacology 8(1)

Table 1.  Frontotemporal dementia tauopathies.59

Tauopathies Presentation Histopathology Tau isoform


Pick’s disease bvFTD or nfvPPA Pick bodies, argyrophilic neuronal 3R
cytoplasmic inclusions
Progressive Rigidity, ophthalmoplegia, Spherical and flame-shaped 4R
supranuclear palsy bradykinesia neurofibrillary tangles, tufted
astrocytes
Corticobasal Rigidity, bradykinesia, dystonia, White matter degeneration with –
degeneration apraxia, alien limb phenomenon ring-shaped astrocytic plaques
FTD, frontotemporal dementia; bvFTD, behavioral variant FTD; nfvPPA, nonfluent variant primary progressive aphasia.

denervation, motor neuron loss, and progressive charged multivesicular body protein 2B
motor impairments that are commonly observed (CHMP2B) gene that leads to dysfunction in the
both in FTLD-TDP and ALS suggesting a com- endosomal ESCRTIII complex that causes dis-
mon neuropathological pathway.92 FTLD-TDP ruption of endosomal traffic.98–101 A decline in
is further subdivided into Types A, B, C, and D levels of CHMP2B results in a decrease of hip-
that differ in the constitution of pathological pocampal dendritic branching and reduced excit-
inclusions and their distribution within the corti- atory synapse activity effectively diminishing
ces.93 Patients with FTLD-TDP Type A have synaptic plasticity and causing neurodegenerative
predominantly neuronal cytoplasmic inclusions deficits.102 This subtype typically presents with
and dystrophic neurites with TDP-43 immunore- changes in behavior and personality (i.e. disinhi-
active lesions in the second layer of the cortices. bition, apathy, loss of emotion, dyscalculia, pro-
FTLD-TDP type B features more neuronal cyto- gressive aphasia that leads to mutism), as well
plasmic inclusions in the superficial and deeper extrapyramidal motor symptoms as the disease
cortical layers. FTLD-TDP type C has a signifi- progresses.98 However, CHMP2B mutations are
cant amount of dystrophic neurites demonstrat- relatively rare,103 and do not appear to increase
ing a ‘corkscrew appearance’ through the cortical the risk of development of FTD.104
layers. FTLD-TDP type D is associated with
multiple neuronal intranuclear inclusions and
dystrophic neurites with fewer neuronal cytoplas- Neuroimaging
mic inclusions.85,93 Protein immunoreactivity to A recent review105 of neuroimaging studies have
members of the FET protein family, with FUS detailed advances in quantifying brain atrophy in
being the more well-known FTD-associated pro- FTD using volumetric MRI measurements and
teinopathy, is the last major subgroup of FTD by observing hypometabolism in specific regions
pathology, and is associated with co-aggregation using [18F] fluorodeoxyglucose PET (18F-FDG
of FET proteins into ubiquitinated inclusions PET). Volumetric MRI studies they reviewed
found in severe cases of sporadic FTD.59,86 Of focused mainly on cerebral loss of grey matter
note, FUS protein involvement in the pathology with the goal of improving accurate diagnosis and
of certain FTD subtypes is suggestive of a com- characterization of FTD variants. In these stud-
mon neuropathologic mechanism and close rela- ies, MRI imaging demonstrated disruption of the
tionship between the FTD disorders and ALS, salience network, default mode network, left
since FUS has been described as a common cause hemisphere language and semantic networks, and
of autosomal-dominant ALS.92,94 Interestingly, executive control networks that imply pathoana-
FTD-associated proteinopathies including tau, tomical relationships in FTD syndromes.
TDP-43, and FUS have demonstrated aggrega- Notably, one study has demonstrated how base-
tion and seeding behavior in both in vivo and in line salience network resting state activity may
vitro models, suggesting FTD pathologies have predict behavioral changes in FTD.106 MRI stud-
prion-like characteristics.86,87 Lastly, the FTLD- ies of bvFTD patients in particular show losses in
UPS subtype is characterized by ubiquitin-immu- the prefrontal cortex, anterior temporal regions,
noreactive inclusions that do not label for tau, insula, anterior cingulate, striatum, and thala-
TDP-43, or FET proteins.12,95–97 This subtype is mus, which differentiate bvFTD from AD with
commonly associated with a mutation in the relatively high sensitivity and specificity. A recent

38 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

retrospective observational study also suggests cortices during the early stages of the disease but
that there are specific anatomical phenotypes spreads outwards as the disorder progresses.
based on the salience and semantic appraisal net- Orbitofrontal, dorsolateral, medial prefrontal cor-
works that may contribute in classifying the oth- tex, and anterior poles display hypometabolism in
erwise heterogeneous bvFTD subgroups.107 The bvFTD patients’ PET scans. Likewise, svPPA
svPPA exhibits more asymmetric temporal lobe patients exhibit more asymmetrical presentations
atrophy on imaging scans with primarily anterior with exclusively left temporal lobe hypometabo-
and inferior regional atrophy more commonly lism, nfvPPA imaging shows hypometabolism in
seen in the left hemisphere. The earliest changes the left frontal and superior temporal regions, and
observed are grey matter losses in the inferior logopenic variants demonstrate a left parietotem-
temporal and fusiform gyri, temporal pole, and poral hypometabolism extending to anterior tem-
parahippocampal entorhinal cortex. Notably, poral and frontal regions.105,108 However, it is
temporal pole atrophy could be seen on MRI important to note that there is considerable indi-
imaging despite a patient’s continued ability to vidual variability when using imaging to classify
maintain activities of daily functioning. However, FTD disorders as a consequence of heterogeneity
the severity of functional decline does later cor- in genetic associations and the underlying patho-
relate with the extent of atrophy as it progresses to logical causes that are yet to be fully understood.
orbitofrontal, inferior frontal, insular, and ante- Consequently, neurobiological and imaging
rior cingulate cortices anteriorly. While both vari- markers remain ancillary modalities in the diag-
ants of PPA share the trait of primarily left nosis and classification of FTD subtypes in con-
hemisphere atrophy, the nfvPPA differs from trast to the gold standard of clinical evaluations.
svPPA in distribution during the course of the Please see Table 2 for a summary of neuroimag-
disease. In nfvPPA, atrophic losses may begin in ing findings commonly found in FTD variants.
the inferior frontal gyrus (particularly in the pars
opercularis), dorsolateral prefrontal cortex, supe-
rior temporal gyrus, and insula. Over time, these Management and treatment
losses will include the prefrontal and temporal There is a lack of quality studies and reports con-
lobe structures in the right hemisphere. Ipsilateral cerning the management of problematic behav-
anterior frontal, lateral temporal, and anterior iors in FTD with mainly sparse case reports and
parietal lobes are also affected. Lastly, the logo- case series detailing their effectiveness.
penic PPA subtype has a more posterior profile Nevertheless, nonpharmacological strategies are
involving the left temporoparietal and posterior considered the preferred intervention before
cingulate atrophy in the early stages of disease resorting to pharmacological therapies that may
progression. In summary, bvFTD subtypes pre- exacerbate medical comorbidities that affect more
sent with frontal and temporal lobe pathology elderly patients. The main purpose of nonphar-
that includes key tracts such as the corpus callo- macological interventions is to prevent disruptive
sum and cingulum, while PPA subtypes are more behaviors, provide symptom relief, and lessen
focal and asymmetric presenting with white mat- caregiver distress. Environmental approaches aim
ter changes in specific networks associated with to decrease irritability, aggression, and anxiety
language processing. Other pathologically defined that arise from patients’ difficulty processing
forms also exhibit specific atrophic patterns. PSP information from the array of daily stimuli. These
is associated with midbrain atrophy, and TDP-43 interventions include reducing noise, limiting
proteinopathies present with a variation of sym- stimulation, simplifying social parameters by lim-
metric and asymmetric atrophic findings accord- iting interaction to small groups of people, and
ing to the underlying TDP type. facilitating the removal of complicated daily activ-
ities that may confuse, and therefore agitate,
18F FDG-PET imaging studies also demonstrate patients. There should be special care in imple-
patterns of hypometabolism that correlate with menting hearing aids, and optimizing sensory
areas of atrophy, and are useful in differentiating stimulation to limit patient discomfort.
AD from FTD. In FTD, PET studies typically Introducing games or old hobbies may reduce
demonstrate hypometabolism in the anterior disinhibition and inappropriate behavior. Exercise
frontotemporal regions including the cingulate has been suggested to reduce behavioral symp-
gyri, uncus, insula, subcortical areas, basal gan- toms as well. Confronting and challenging psy-
glia, and medial thalamic regions. Hypometabolism chotic symptoms may lead to more agitation, so
is limited to frontal, parietal, and temporal reassurance and distracting patients are

journals.sagepub.com/home/tpp 39
Therapeutic Advances in Psychopharmacology 8(1)

Table 2.  Frontotemporal dementia neuroimaging correlations.105

Variant MRI findings PET/SPECT findings


bvFTD Atrophy in prefrontal cortex, anterior temporal Decreased FA in uncinate fasciculus,
regions, anterior cingulate, the striatum, genu of corpus callosum and cingulum,
thalamus, the insula superior and inferior longitudinal
fasciculi, inferior fronto-occipital
fasciculus
nfvPPA Predominant left hemisphere cortical Decreased FA in left uncinate fasciculus,
loss; atrophy predominantly affecting pars inferior longitudinal fasciculus tracts,
opercularis in the inferior frontal gyrus corpus callosum, cingulum; notable
extending to prefrontal and temporal cortices, sparing of occipital white matter,
caudate and putamen atrophy bilaterally brainstem, cerebellum
svPPA Predominant left hemisphere cortical loss; Decreased FA in left superior longitudinal
grey matter loss predominantly in inferior fasciculus, corpus callosum, left
temporal and fusiform gyri, temporal pole, cingulum, left orbitofrontal, inferior
parahippocampal cortex, entorhinal cortex; frontal, anterior temporal, inferior
losses extend to anterior cingulate, orbitofrontal, parietal white matter regions
inferior frontal, and insular cortices
Logopenic Atrophy of left temporoparietal and posterior Notable white matter losses in left
PPA cingulate frontoparietal regions including left
superior longitudinal fasciculus and
arcuate fasciculus
FTD, frontotemporal dementia; bvFTD, behavioral variant FTD; FA, fractional anistotrophy; MRI, magnetic resonance
imaging; nfvPPA, nonfluent variant primary progressive aphasia; PET, positron emission tomography; PPA, primary
progressive aphasia; SPECT, single-photon emission computed tomography; svPPA, semantic variant primary
progressive aphasia.

considered to be more acceptable alternatives in improvement and may have caused impairment
decreasing behavioral symptoms. Clinicians have in several neuropsychological tests including vis-
attempted to modify compulsions, apathy, and ual discrimination tasks, errors in paired-associ-
lack of motivation using reward systems, although ates learning task, and delayed pattern recognition
there are limited data concerning this interven- memory accuracy. Discrepancies between study
tion. Cognitive behavioral therapy is currently results were attributed to subjects responding
being studied as a possible option to modulate more favorably to lower doses of paroxetine sug-
behavior, but studies are still preliminary.109,110 gesting a higher rate of tolerability at 20 mg daily
compared with higher dosages. Additionally, par-
There are currently no US Food and Drug oxetine may be more effective when prescribed
Administration (FDA) approved pharmacologi- earlier in the disease process, although it is diffi-
cal therapies for FTD.111 There is limited evi- cult to determine without more in-depth and
dence that antidepressants such as selective higher-powered studies.
serotonin reuptake inhibitors (SSRIs) may be
useful for impulsivity, irritability, eating behavior, Citalopram is another SSRI that has been
and disinhibition.112 For example, one small, ran- recently shown to provide some improvement in
domized-controlled, open-label 14-month trial113 FTD patient behavior. Herrmann and colleagues
reported improvement of behavioral symptoms in found improvement with a citalopram challenge
subjects with ages ranging from 64 to 68 years at 40 mg daily dosages that led to a decrease
after being prescribed paroxetine at dosages of up in behavioral symptoms including irritability,
to 20 mg/day. Notably, subjects tolerated parox- depression, apathy, and disinhibition, while also
etine well and exhibited improvement in improving overall NPI scores.115 More recently,
Neuropsychiatric Inventory (NPI) scoring. in a randomized double-blinded placebo-con-
However, a follow-up study114 using placebo con- trolled crossover study, bvFTD patients who
trols with a similarly small subject population were prescribed 30 mg daily dosages of citalo-
determined that paroxetine administration at pram demonstrated improved disinhibition
dosages up to 40 mg/day provided little objective symptoms in addition to a partial restoration of

40 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

serotonergic neurotransmission in dysfunctional process should take into account the limited data
prefrontal cortical systems as measured by mag- provided by these stimulant studies due to their
netoencephalography and low power and questionable generalizability, as
electroencephalography.116 well as possible side effects and tolerability issues
of stimulants in more elderly patients.
Studies of sertraline efficacy in treating FTD
symptoms are limited to mainly observational Memantine and cholinesterase inhibitors have
studies. One study comparing stereotypical not demonstrated clinically significant efficacy in
behavior in FTD and AD subjects suggested that treating FTD patients, and may also hasten cog-
stereotypical movements in FTD could be nitive decline or worsen behavioral symp-
decreased with sertraline administration at a dos- toms.125,126 This is likely due to a lack of consistent
age range of 50–100 mg/day.117 A case study of a evidence of a cholinergic deficit in FTD without
53-year-old ALS-FTD patient indicated that ser- concomitant pathology of other major neurocog-
traline treatment could also decrease inappropri- nitive disorders.127 However, at least one study
ate sexual behavior in addition to decreasing reported some efficacy in improving behavioral
aggressive behavior which was helpful in mini- symptoms with galantamine treatment in an
mizing caregiver burden.118 aphasic subgroup of FTD without a preponder-
ance of serious adverse events, although no effect
Alternatively, treatment with trazodone at dos- was found in overall treatment of the general
ages of at least 300 mg/day over 12 weeks has FTD subject population.128
been reported to be helpful in decreasing symp-
toms of problematic eating, agitation, irritability, Complaints of insomnia may be due to behavioral
dysphoria, and depression, although adverse dysfunction or neuronal degeneration causing
events were more prominent in subjects being impairment of the sleep/wake cycle. However,
prescribed the psychotropic. Typical emergent primary sleep disorders such as restless leg syn-
side effects included fatigue, dizziness, and hypo- drome may also contribute to insomnia and can
tension, but the authors reported that these symp- be treated with alpha-2-delta calcium channel
toms were mild in severity.119 At least one ligands (e.g. gabapentin, pregabalin) or dopamin-
open-label 12-week trial showed a treatment ergic agents. Agitation may be controlled by atyp-
response with fluvoxamine administration from a ical antipsychotics such as risperidone, olanzapine,
dosage range of 50–150 mg/day (mean dose was quetiapine, and clozapine, but they are not FDA-
110 mg/day) improving stereotypic behavior, eat- approved and could be detrimental in a demen-
ing behavior, and roaming behavior.120 tia-associated agitation due to increased risk of
mortality and unwanted side effects.111,129,130 Of
Stimulants have been proposed as alternative note, antipsychotics have been suggested to be
treatments due to their mechanism of action in more helpful in treating psychotic symptoms in
elevating extracellular catecholamine concentra- patients with the C9orf72 subtype of FTD, but
tions to improve cognitive functioning and lessen once again, data is limited with increased risk of
frontostriatal and orbitofrontal dysfunction extrapyramidal side effects and morbidity in
reported to affect risk-taking behavior in FTD dementia patients preventing safe utilization of
patients.121–123 Exemplifying this, one study123 these neuroleptics.131 Consequently, due to the
found that eight patients who were prescribed limited data and power of pharmacologic treat-
methylphenidate at 40 mg daily dosages demon- ment studies in FTD populations; nonpharmaco-
strated decreased risk-taking behavior with over- logical interventions and caregiver support are the
all improvement in general behavior. Additionally, preferred management options.125
a randomized double-blinded crossover trial
comparing dextroamphetamine and quetiapine Due to the limited contemporary psychopharma-
use in FTD patients found that dextroampheta- cologic interventions available to physicians, sev-
mine administration decreased disinhibition and eral novel approaches are being researched to
apathy symptoms, while being more tolerable develop more effective treatments for FTD
than quetiapine, which caused sedation in their symptoms. One such approach is the use of the
subject population.124 Despite these studies sug- neuropeptide, oxytocin, to aid in mediating
gesting the safety and efficacy of stimulants in behavior and improving apathy and empathy
treating specific symptoms common in the FTD in svPPA patients.132 Another study suggested
population, a clinician’s medical decision-making agomelatine, a melatonin receptor agonist, may

journals.sagepub.com/home/tpp 41
Therapeutic Advances in Psychopharmacology 8(1)

improve apathy in FTD patients.133 A preclinical decreasing FTD-specific symptomatology and


trial reported active immunization against tau difficult behavior. Additionally, the extensive list
aggregates using anti-tau antibodies may improve of symptoms that are being targeted for interven-
cognition through decreasing tauopathy.134 A tion typically necessitates a variety of therapies
study investigating the effectiveness of inhibiting for clinical improvement, which fundamentally
tau aggregation in bvFTD patients using leuco- complicates treatment planning. However, this
methylthioninium was recently completed, but renewed interest in FTD research gives hope for
detailed results are pending release and publica- future advancements that aim to increase diag-
tion.135 Other targets for therapy include disrupt- nostic capabilities, and expand treatment options
ing the downstream effects of PGRN and through novel therapeutic targets.
C9orf72 mutations.111 For example, antisense
oligonucleotides are being used to target the Funding
transcription elongation factor SUPT4H1/ This research received no specific grant from any
SUPT5H to bi-directionally suppress C9orf72 funding agency in the public, commercial, or not-
RNA transcription.136 Other proposed methods for-profit sectors.
involve elevating PGRN levels through molecu-
lar manipulation of biological pathways using Conflict of interest statement
suberoylanilide hydroxamic acid, androgen treat- Mallika Lavakumar received honorarium from
ment, and bafilomycin A1.135 Oakstone and receives research funding from the
Health Research Services Administration.
Rajesh R Tampi receives honorarium from
Conclusion Oakstone and royalties from Lippincott Williams
The entirety of the FTD clinical syndrome is & Wilkins.
now known to be composed of complex and het-
erogeneous variants that vary in presentation,
genetic alterations, and pathological processes.
Consequently, there is a need for physicians to References
become well versed in the diagnostic criteria set 1. Bang J, Spina S and Miller BL. Frontotemporal
dementia. Lancet 2015; 386: 1672–1682.
forth by consortiums and expert consensus in
order to properly assess and manage FTD 2. Kurz A, Kurz C, Ellis K, et al. What is
patients. According to recent epidemiologic frontotemporal dementia? Maturitas 2014; 79:
studies, FTD is likely to be underdiagnosed due 216–219.
to the inherent difficulty accurately diagnosing 3. Bott NT, Radke A, Stephens ML, et al.
these disorders. These findings underline the Frontotemporal dementia: diagnosis, deficits
need to standardize and update current criteria and management. Neurodegener Dis Manag
to increase diagnostic sensitivity, and allow for 2014; 4: 439–454.
better management of these variants at an earlier 4. World Health Organization. Dementia fact
stage of the disease course. There have been sev- sheet, http://www.who.int/mediacentre/
eral breakthroughs in genetic and neuroimaging factsheets/fs362/en/ (2016). Accessed January
studies that have elucidated the pathophysiologi- 22, 2017.
cal characteristics of these FTD subtypes in the
5. Rosso SM, Donker Kaat L, Baks T, et al.
past decade. However, FTD pathology is still
Frontotemporal dementia in The Netherlands:
relatively unknown and requires further investi- patient characteristics and prevalence estimates
gations to produce more effective diagnostic bio- from a population-based study. Brain 2003;
markers and therapies, especially as later course 126: 2016–2022.
presentations begin to mimic AD symptomatol-
ogy. Current management guidelines recom- 6. Onyike CU and Diehl-Schmid J. The
epidemiology of frontotemporal dementia. Int
mend a focus on nonpharmacological
Rev Psychiatry 2013; 25: 130–137.
interventions, as these have been found to be the
most studied and effective practices at the dis- 7. Knopman DS and Roberts RO. Estimating
posal of clinicians. Contemporary pharmacologi- the number of persons with frontotemporal
cal treatment options are still lacking due to a lobar degeneration in the US population. J Mol
deficit of comprehensive randomized controlled Neurosci 2011; 45: 330–335.
trials to determine efficacy and safety, although 8. Lambert MA, Bickel H, Prince M, et al.
there is limited evidence of SSRI efficacy in Estimating the burden of early onset dementia;

42 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

systematic review of disease prevalence. Eur J 20. Ghetti B, Oblak AL, Boeve BF, et al. Invited
Neurol 2014; 21: 563–569. review: frontotemporal dementia caused
by microtubule-associated protein tau
9. Rascovsky K, Hodges JR, Knopman D, et al.
gene (MAPT) mutations: a chameleon for
Sensitivity of revised diagnostic criteria for the
neuropathology and neuroimaging. Neuropathol
behavioural variant of frontotemporal dementia.
Appl Neurobiol 2015; 41: 24–46.
Brain 2011; 134: 2456–2477.
21. Ehrlich M, Hallmann AL, Reinhardt P,
10. Gorno-Tempini ML, Hillis AE, Weintraub
et al. Distinct neurodegenerative changes in
S, et al. Classification of primary progressive
an induced pluripotent stem cell model of
aphasia and its variants. Neurology 2011; 76:
frontotemporal dementia linked to mutant TAU
1006–1014.
protein. Stem Cell Reports 2015; 5: 83–96.
11. Leyton CE and Hodges JR. Towards a clearer
22. Lee SE, Tartaglia MC, Yener G, et al.
definition of logopenic progressive aphasia. Curr
Neurodegenerative disease phenotypes in
Neurol Neurosci Rep 2013; 13: 396.
carriers of MAPT p.A152T, a risk factor for
12. Rohrer JD, Guerreiro R, Vandrovcova J, et al. The frontotemporal dementia spectrum disorders
heritability and genetics of frontotemporal lobar and Alzheimer disease. Alzheimer Dis Assoc
degeneration. Neurology 2009; 73: 1451–1456. Disord 2013; 27: 302–309.
13. Rademakers R, Neumann M and Mackenzie IR. 23. Coppola G, Chinnathambi S, Lee JJ, et al.
Advances in understanding the molecular basis Evidence for a role of the rare p.A152T variant
of frontotemporal dementia. Nat Rev Neurol in MAPT in increasing the risk for FTD-
2012; 8: 423–434. spectrum and Alzheimer’s diseases. Hum Mol
Genet 2012; 21: 3500–3512.
14. Capozzo R, Sassi C, Hammer MB, et al.
Clinical and genetic analyses of familial and 24. Rovelet-Lecrux A, Hannequin D, Guillin O,
sporadic frontotemporal dementia patients in et al. Frontotemporal dementia phenotype
Southern Italy. Alzheimers Dement 2017; 13: associated with MAPT gene duplication. J
858–869. Alzheimers Dis 2010; 21: 897–902.
15. Moreno F, Indakoetxea B, Barandiaran M, et al. 25. Hutton M, Lendon CL, Rizzu P, et al.
The unexpected co-occurrence of GRN and Association of missense and 5’-splice-site
MAPT p.A152T in Basque families: clinical mutations in tau with the inherited dementia
and pathological characteristics. PLoS One FTDP-17. Nature 1998; 393: 702–705.
2017; 12: e0178093.
26. Sudre CH, Bocchetta M, Cash D, et al. White
16. Rademakers R, Cruts M and van Broeckhoven matter hyperintensities are seen only in GRN
C. The role of tau (MAPT) in frontotemporal mutation carriers in the GENFI cohort.
dementia and related tauopathies. Hum Mutat Neuroimage Clin 2017; 15: 171–180.
2004; 24: 277–295.
27. Kao AW, McKay A, Singh PP, et al.
17. Boeve BF and Hutton M. Refining Progranulin, lysosomal regulation and
frontotemporal dementia with parkinsonism neurodegenerative disease. Nat Rev Neurosci
linked to chromosome 17: introducing FTDP- 2017; 18: 325–333.
17 (MAPT) and FTDP-17 (PGRN). Arch
28. Jackman K, Kahles T, Lane D, et al.
Neurol 2008; 65: 460–464.
Progranulin deficiency promotes post-ischemic
18. Tacik P, Sanchez-Contreras M, DeTure blood-brain barrier disruption. J Neurosci 2013;
M, et al. Clinicopathologic heterogeneity in 33: 19579–19589.
frontotemporal dementia and parkinsonism
29. Paternico D, Premi E, Gazzina S, et al. White
linked to chromosome 17 (FTDP-17) due to
matter hyperintensities characterize monogenic
microtubule-associated protein tau (MAPT)
frontotemporal dementia with granulin
p.P301L mutation, including a patient with
mutations. Neurobiol Aging 2016; 38: 176–180.
globular glial tauopathy. Neuropathol Appl
Neurobiol 2017; 43: 200–214. 30. Baker M, Mackenzie IR, Pickering-Brown
SM, et al. Mutations in progranulin cause
19. Biswas MH, Almeida S, Lopez-Gonzalez
tau-negative frontotemporal dementia linked to
R, et al. MMP-9 and MMP-2 contribute
chromosome 17. Nature 2006; 442: 916–919.
to neuronal cell death in iPSC models
of frontotemporal dementia with MAPT 31. Caroppo P, Le Ber I, Camuzat A, et al.
mutations. Stem Cell Reports 2016; 7: Extensive white matter involvement in patients
316–324. with frontotemporal lobar degeneration: think

journals.sagepub.com/home/tpp 43
Therapeutic Advances in Psychopharmacology 8(1)

progranulin. JAMA Neurol 2014; 71: 1562– 42. DeJesus-Hernandez M, Mackenzie IR, Boeve
1566. BF, et al. Expanded GGGGCC hexanucleotide
repeat in noncoding region of C9ORF72 causes
32. Lashley T, Rohrer JD, Mahoney C, et al.
chromosome 9p-linked FTD and ALS. Neuron
A pathogenic progranulin mutation and
2011; 72: 245–256.
C9orf72 repeat expansion in a family with
frontotemporal dementia. Neuropathol Appl 43. Renton AE, Majounie E, Waite A, et al. A
Neurobiol 2014; 40: 502–513. hexanucleotide repeat expansion in C9ORF72
is the cause of chromosome 9p21-linked ALS-
33. Chen-Plotkin AS, Unger TL, Gallagher
FTD. Neuron 2011; 72: 257–268.
MD, et al. TMEM106B, the risk gene for
frontotemporal dementia, is regulated by 44. Van Mossevelde S, van der Zee J, Gijselinck I,
the microRNA-132/212 cluster and affects et al. Clinical evidence of disease anticipation in
progranulin pathways. J Neurosci 2012; 32: families segregating a C9orf72 repeat expansion.
11213–11227. JAMA Neurol 2017; 74: 445–452.
34. Tapia L, Milnerwood A, Guo A, et al. 45. Devenney E, Hornberger M, Irish M, et al.
Progranulin deficiency decreases gross neural Frontotemporal dementia associated with the
connectivity but enhances transmission at C9ORF72 mutation: a unique clinical profile.
individual synapses. J Neurosci 2011; 31: JAMA Neurol 2014; 71: 331–339.
11126–11132.
46. Farg MA, Sundaramoorthy V, Sultana JM, et al.
35. Gijselinck I, Van Broeckhoven C and Cruts C9ORF72, implicated in amytrophic lateral
M. Granulin mutations associated with sclerosis and frontotemporal dementia, regulates
frontotemporal lobar degeneration and related endosomal trafficking. Hum Mol Genet 2014;
disorders: an update. Hum Mutat 2008; 29: 23: 3579–3595.
1373–1386.
47. Rohrer JD, Isaacs AM, Mizielinska S, et al.
36. van Swieten JC and Heutink P. Mutations C9orf72 expansions in frontotemporal dementia
in progranulin (GRN) within the spectrum and amyotrophic lateral sclerosis. Lancet Neurol
of clinical and pathological phenotypes of 2015; 14: 291–301.
frontotemporal dementia. Lancet Neurol 2008;
7: 965–974. 48. Lee SE, Khazenzon AM, Trujillo AJ, et al.
Altered network connectivity in frontotemporal
37. Cruts M, Gijselinck I, van der Zee J, et al. dementia with C9orf72 hexanucleotide repeat
Null mutations in progranulin cause ubiquitin- expansion. Brain 2014; 137: 3047–3060.
positive frontotemporal dementia linked
to chromosome 17q21. Nature 2006; 442: 49. Irwin DJ, McMillan CT, Brettschneider J,
920–924. et al. Cognitive decline and reduced survival in
C9orf72 expansion frontotemporal degeneration
38. Snowden JS, Pickering-Brown SM, Mackenzie and amyotrophic lateral sclerosis. J Neurol
IR, et al. Progranulin gene mutations associated Neurosurg Psychiatry 2013; 84: 163–169.
with frontotemporal dementia and progressive
non-fluent aphasia. Brain 2006; 129: 50. Khan BK, Yokoyama JS, Takada LT, et al.
3091–3102. Atypical, slowly progressive behavioural variant
frontotemporal dementia associated with
39. Mahoney CJ, Beck J, Rohrer JD, et al. C9ORF72 hexanucleotide expansion. J Neurol
Frontotemporal dementia with the C9ORF72 Neurosurg Psychiatry 2012; 83: 358–364.
hexanucleotide repeat expansion: clinical,
neuroanatomical and neuropathological 51. Rohrer JD, Nicholas JM, Cash DM, et al.
features. Brain 2012; 135: 736–750. Presymptomatic cognitive and neuroanatomical
changes in genetic frontotemporal dementia in
40. Majounie E, Renton AE, Mok K, et al. the Genetic Frontotemporal dementia Initiative
Frequency of the C9orf72 hexanucleotide (GENFI) study: a cross-sectional analysis.
repeat expansion in patients with amyotrophic Lancet Neurol 2015; 14: 253–262.
lateral sclerosis and frontotemporal dementia:
a cross-sectional study. Lancet Neurol 2012; 11: 52. Ferrari R, Hernandez DG, Nalls MA, et al.
323–330. Frontotemporal dementia and its subtypes: a
genome-wide association study. Lancet Neurol
41. Hsiung GY, DeJesus-Hernandez M, Feldman
2014; 13: 686–699.
HH, et al. Clinical and pathological features
of familial frontotemporal dementia caused by 53. Piguet O, Brooks WS, Halliday GM, et al.
C9ORF72 mutation on chromosome 9p. Brain Similar early clinical presentations in familial
2012; 135: 709–722. and non-familial frontotemporal dementia.

44 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

J Neurol Neurosurg Psychiatry 2004; 75: 65. Gitcho MA, Strider J, Carter D, et al. VCP
1743–1745. mutations causing frontotemporal lobar
degeneration disrupt localization of TDP-43
54. Pickering-Brown SM, Rollinson S, Du Plessis
and induce cell death. J Biol Chem 2009; 284:
D, et al. Frequency and clinical characteristics
12384–12398.
of progranulin mutation carriers in the
Manchester frontotemporal lobar degeneration 66. Watts GD, Wymer J, Kovach MJ, et al.
cohort: comparison with patients with MAPT Inclusion body myopathy associated with Paget
and no known mutations. Brain 2008; 131: disease of bone and frontotemporal dementia
721–731. is caused by mutant valosin-containing protein.
Nat Genet 2004; 36: 377–381.
55. Ng AS. Genetics of frontotemporal dementia
in Asia: advancing knowledge through 67. Neumann M, Mackenzie IR, Cairns NJ,
collaboration. Neurology 2015; 85: 2060–2062. et al. TDP-43 in the ubiquitin pathology of
frontotemporal dementia with VCP gene
56. Snowden JS, Adams J, Harris J, et al. Distinct mutations. J Neuropathol Exp Neurol 2007; 66:
clinical and pathological phenotypes in 152–157.
frontotemporal dementia associated with
MAPT, PGRN and C9orf72 mutations. 68. Forman MS, Mackenzie IR, Cairns NJ,
Amyotroph Lateral Scler Frontotemporal Degener et al. Novel ubiquitin neuropathology in
2015; 16: 497–505. frontotemporal dementia with valosin-
containing protein gene mutations.
57. Moreno F, Indakoetxea B, Barandiaran M, et al. J Neuropathol Exp Neurol 2006; 65:
“Frontotemporoparietal” dementia: clinical 571–581.
phenotype associated with the c.709–1G>A
PGRN mutation. Neurology 2009; 73: 1367– 69. Bersano A, Del Bo R, Lamperti C, et al.
1374. Inclusion body myopathy and frontotemporal
dementia caused by a novel VCP mutation.
58. Gass J, Cannon A, Mackenzie IR, et al. Neurobiol Aging 2009; 30: 752–758.
Mutations in progranulin are a major cause
of ubiquitin-positive frontotemporal lobar 70. Jacquin A, Rouaud O, Soichot P, et al.
degeneration. Hum Mol Genet 2006; 15: Psychiatric presentation of frontotemporal
2988–3001. dementia associated with inclusion body
myopathy due to the VCP mutation (R155H)
59. Mackenzie IR and Neumann M. Molecular in a French family. Case Rep Neurol 2013; 5:
neuropathology of frontotemporal dementia: 187–194.
insights into disease mechanisms from
postmortem studies. J Neurochem 2016; 71. Kazamel M, Sorenson EJ, McEvoy KM, et al.
138(Suppl. 1): 54–70. Clinical spectrum of valosin containing
protein (VCP)-opathy. Muscle Nerve 2016;
60. Woollacott IO and Rohrer JD. The clinical 54: 94–99.
spectrum of sporadic and familial forms of
frontotemporal dementia. J Neurochem 2016; 72. Gidaro T, Modoni A, Sabatelli M, et al. An
138(Suppl. 1): 6–31. Italian family with inclusion-body myopathy
and frontotemporal dementia due to mutation
61. Patel AN and Sampson JB. Cognitive profile in the VCP gene. Muscle Nerve 2008; 37:
of C9orf72 in frontotemporal dementia and 111–114.
amyotrophic lateral sclerosis. Curr Neurol
Neurosci Rep 2015; 15: 59. 73. Komatsu J, Iwasa K, Yanase D, et al. Inclusion
body myopathy with Paget disease of the bone
62. Takada LT and Sha SJ. Neuropsychiatric and frontotemporal dementia associated with a
features of C9orf72-associated behavioral novel G156S mutation in the VCP gene. Muscle
variant frontotemporal dementia and Nerve. Epub ahead of print 19 July 2013. DOI:
frontotemporal dementia with motor neuron 10.1002/mus.23960.
disease. Alzheimers Res Ther 2012; 4: 38.
74. Kimonis VE, Fulchiero E, Vesa J, et al. VCP
63. Suhonen NM, Haanpaa RM, Korhonen V, et al. disease associated with myopathy, Paget disease
Neuropsychological profile in the C9ORF72 of bone and frontotemporal dementia: review of
associated behavioral variant frontotemporal a unique disorder. Biochim Biophys Acta 2008;
dementia. J Alzheimers Dis 2017; 58: 479–489. 1782: 744–748.
64. Rohrer JD, Lashley T, Schott JM, et al. Clinical 75. Shi Z, Hayashi YK, Mitsuhashi S, et al.
and neuroanatomical signatures of tissue Characterization of the Asian myopathy patients
pathology in frontotemporal lobar degeneration. with VCP mutations. Eur J Neurol 2012; 19:
Brain 2011; 134: 2565–2581. 501–509.

journals.sagepub.com/home/tpp 45
Therapeutic Advances in Psychopharmacology 8(1)

76. Haubenberger D, Bittner RE, Rauch-Shorny 88. Lee VM, Goedert M and Trojanowski JQ.
S, et al. Inclusion body myopathy and Paget Neurodegenerative tauopathies. Annu Rev
disease is linked to a novel mutation in the VCP Neurosci 2001; 24: 1121–1159.
gene. Neurology 2005; 65: 1304–1305.
89. Josephs KA, Hodges JR, Snowden JS, et al.
77. Kim EJ, Park YE, Kim DS, et al. Inclusion Neuropathological background of phenotypical
body myopathy with Paget disease of bone variability in frontotemporal dementia. Acta
and frontotemporal dementia linked to VCP Neuropathol 2011; 122: 137–153.
p.Arg155Cys in a Korean family. Arch Neurol
2011; 68: 787–796. 90. Josephs KA, Holton JL, Rossor MN, et al.
Frontotemporal lobar degeneration and
78. Mehta SG, Khare M, Ramani R, et al. ubiquitin immunohistochemistry. Neuropathol
Genotype-phenotype studies of VCP-associated Appl Neurobiol 2004; 30: 369–373.
inclusion body myopathy with Paget disease
of bone and/or frontotemporal dementia. Clin 91. Mackenzie IR, Rademakers R and Neumann
Genet 2013; 83: 422–431. M. TDP-43 and FUS in amyotrophic lateral
sclerosis and frontotemporal dementia. Lancet
79. Neumann M, Sampathu DM, Kwong LK, et al. Neurol 2010; 9: 995–1007.
Ubiquitinated TDP-43 in frontotemporal lobar
degeneration and amyotrophic lateral sclerosis. 92. Walker AK, Spiller KJ, Ge G, et al. Functional
Science 2006; 314: 130–133. recovery in new mouse models of ALS/FTLD
after clearance of pathological cytoplasmic
80. Benajiba L, Le Ber I, Camuzat A, et al. TDP-43. Acta Neuropathol 2015; 130: 643–660.
TARDBP mutations in motoneuron disease
with frontotemporal lobar degeneration. Ann 93. Mackenzie IR, Neumann M, Baborie A, et al. A
Neurol 2009; 65: 470–473. harmonized classification system for FTLD-TDP
pathology. Acta Neuropathol 2011; 122: 111–113.
81. Synofzik M, Born C, Rominger A, et al.
Targeted high-throughput sequencing identifies 94. Neumann M, Rademakers R, Roeber S,
a TARDBP mutation as a cause of early-onset et al. A new subtype of frontotemporal lobar
FTD without motor neuron disease. Neurobiol degeneration with FUS pathology. Brain 2009;
Aging 2014; 35: 1212. e1–e5. 132: 2922–2931.

82. Borroni B, Bonvicini C, Alberici A, et al. 95. Roeber S, Mackenzie IR, Kretzschmar HA,
Mutation within TARDBP leads to et al. TDP-43-negative FTLD-U is a significant
frontotemporal dementia without motor neuron new clinico-pathological subtype of FTLD. Acta
disease. Hum Mutat 2009; 30: E974–E983. Neuropathol 2008; 116: 147–157.

83. Borroni B, Archetti S, Del Bo R, et al. 96. Mackenzie IR, Foti D, Woulfe J, et al. Atypical
TARDBP mutations in frontotemporal lobar frontotemporal lobar degeneration with
degeneration: frequency, clinical features, and ubiquitin-positive, TDP-43-negative neuronal
disease course. Rejuvenation Res 2010; 13: inclusions. Brain 2008; 131: 1282–1293.
509–517.
97. Holm IE, Isaacs AM and Mackenzie IR.
84. Mutihac R, Alegre-Abarrategui J, Gordon D, Absence of FUS-immunoreactive pathology in
et al. TARDBP pathogenic mutations increase frontotemporal dementia linked to chromosome
cytoplasmic translocation of TDP-43 and cause 3 (FTD-3) caused by mutation in the CHMP2B
reduction of endoplasmic reticulum Ca(2)(+) gene. Acta Neuropathol 2009; 118: 719–720.
signaling in motor neurons. Neurobiol Dis 2015;
98. Holm IE, Englund E, Mackenzie IR, et al.
75: 64–77.
A reassessment of the neuropathology of
85. Lashley T, Rohrer JD, Mead S, et al. Review: frontotemporal dementia linked to chromosome
an update on clinical, genetic and pathological 3. J Neuropathol Exp Neurol 2007; 66: 884–891.
aspects of frontotemporal lobar degenerations.
99. Lindquist SG, Braedgaard H, Svenstrup K,
Neuropathol Appl Neurobiol 2015; 41: 858–881.
et al. Frontotemporal dementia linked to
86. Hock EM and Polymenidou M. Prion-like chromosome 3 (FTD-3)–current concepts and
propagation as a pathogenic principle in the detection of a previously unknown branch
frontotemporal dementia. J Neurochem 2016; of the Danish FTD-3 family. Eur J Neurol 2008;
138(Suppl. 1): 163–183. 15: 667–670.

87. Bodea LG, Eckert A, Ittner LM, et al. 100. Skibinski G, Parkinson NJ, Brown JM, et al.
Tau physiology and pathomechanisms in Mutations in the endosomal ESCRTIII-
frontotemporal lobar degeneration. J Neurochem complex subunit CHMP2B in frontotemporal
2016; 138(Suppl. 1): 71–94. dementia. Nat Genet 2005; 37: 806–808.

46 journals.sagepub.com/home/tpp
J Young, M Lavakumar et al.

101. Urwin H, Authier A, Nielsen JE, et al. 114. Deakin JB, Rahman S, Nestor PJ, et al.
Disruption of endocytic trafficking in Paroxetine does not improve symptoms and
frontotemporal dementia with CHMP2B impairs cognition in frontotemporal dementia:
mutations. Hum Mol Genet 2010; 19: 2228– a double-blind randomized controlled trial.
2238. Psychopharmacology (Berl) 2004; 172: 400–408.
102. Chassefeyre R, Martinez-Hernandez J, Bertaso 115. Herrmann N, Black SE, Chow T, et al.
F, et al. Regulation of postsynaptic function Serotonergic function and treatment of
by the dementia-related ESCRT-III subunit behavioral and psychological symptoms
CHMP2B. J Neurosci 2015; 35: 3155–3173. of frontotemporal dementia. Am J Geriatr
Psychiatry 2012; 20: 789–797.
103. Cannon A, Baker M, Boeve B, et al. CHMP2B
mutations are not a common cause of 116. Hughes LE, Rittman T, Regenthal R, et al.
frontotemporal lobar degeneration. Neurosci Lett Improving response inhibition systems in
2006; 398: 83–84. frontotemporal dementia with citalopram. Brain
2015; 138: 1961–1975.
104. Schumacher A, Friedrich P, Diehl-Schmid J,
et al. No association of chromatin-modifying 117. Mendez MF, Shapira JS and Miller BL.
protein 2B with sporadic frontotemporal Stereotypical movements and frontotemporal
dementia. Neurobiol Aging 2007; 28: 1789–1790. dementia. Mov Disord 2005; 20: 742–745.
105. Gordon E, Rohrer JD and Fox NC. Advances 118. Anneser JM, Jox RJ and Borasio GD.
in neuroimaging in frontotemporal dementia. J Inappropriate sexual behaviour in a case of ALS
Neurochem 2016; 138(Suppl. 1): 193–210. and FTD: successful treatment with sertraline.
Amyotroph Lateral Scler 2007; 8: 189–190.
106. Day GS, Farb NA, Tang-Wai DF, et al. Salience
network resting-state activity: prediction of 119. Lebert F, Stekke W, Hasenbroekx C, et al.
frontotemporal dementia progression. JAMA Frontotemporal dementia: a randomised,
Neurol 2013; 70: 1249–1253. controlled trial with trazodone. Dement Geriatr
Cogn Disord 2004; 17: 355–359.
107. Ranasinghe KG, Rankin KP, Pressman PS,
et al. Distinct subtypes of behavioral variant 120. Ikeda M, Shigenobu K, Fukuhara R, et al.
frontotemporal dementia based on patterns of Efficacy of fluvoxamine as a treatment for
network degeneration. JAMA Neurol 2016; 73: behavioral symptoms in frontotemporal lobar
1078–1088. degeneration patients. Dement Geriatr Cogn
Disord 2004; 17: 117–121.
108. Kato T, Inui Y, Nakamura A, et al. Brain
fluorodeoxyglucose (FDG) PET in dementia. 121. Mega MS and Cummings JL. Frontal-
Ageing Res Rev 2016; 30: 73–84. subcortical circuits and neuropsychiatric
disorders. J Neuropsychiatry Clin Neurosci 1994;
109. Barton C, Ketelle R, Merrilees J, et al. Non-
6: 358–370.
pharmacological management of behavioral
symptoms in frontotemporal and other 122. Rahman S, Sahakian BJ, Hodges JR, et al.
dementias. Curr Neurol Neurosci Rep Specific cognitive deficits in mild frontal variant
2016; 16: 14. frontotemporal dementia. Brain 1999; 122:
1469–1493.
110. Shinagawa S. Neuropsychiatric management of
young-onset dementias. Psychiatr Clin North Am 123. Rahman S, Robbins TW, Hodges JR, et al.
2015; 38: 323–331. Methylphenidate (‘Ritalin’) can ameliorate
abnormal risk-taking behavior in the
111. Tsai RM and Boxer AL. Therapy and clinical
frontal variant of frontotemporal dementia.
trials in frontotemporal dementia: past, present,
Neuropsychopharmacology 2006; 31: 651–658.
and future. J Neurochem 2016; 138(Suppl. 1):
211–221. 124. Huey ED, Garcia C, Wassermann EM, et al.
Stimulant treatment of frontotemporal dementia
112. Swartz JR, Miller BL, Lesser IM, et al.
in 8 patients. J Clin Psychiatry 2008; 69:
Frontotemporal dementia: treatment response
1981–1982.
to serotonin selective reuptake inhibitors. J Clin
Psychiatry 1997; 58: 212–216. 125. Mocellin R, Scholes A, Walterfang M, et al.
Clinical update on frontotemporal dementia:
113. Moretti R, Torre P, Antonello RM, et al.
diagnosis and treatment. Australas Psychiatry
Frontotemporal dementia: paroxetine as a
2015; 23: 481–487.
possible treatment of behavior symptoms. A
randomized, controlled, open 14-month study. 126. Vercelletto M, Boutoleau-Bretonniere C,
Eur Neurol 2003; 49: 13–19. Volteau C, et al. Memantine in behavioral

journals.sagepub.com/home/tpp 47
Therapeutic Advances in Psychopharmacology 8(1)

variant frontotemporal dementia: negative perspectives. Neuropsychiatr Dis Treat 2014; 10:
results. J Alzheimers Dis 2011; 23: 297–310.
749–759.
132. Finger EC, MacKinley J, Blair M, et al.
127. Chow TW. Treatment approaches to symptoms Oxytocin for frontotemporal dementia: a
associated with frontotemporal degeneration. randomized dose-finding study of safety and
Curr Psychiatry Rep 2005; 7: 376–380. tolerability. Neurology 2015; 84: 174–181.
128. Kertesz A, Morlog D, Light M, et al. 133. Callegari I, Mattei C, Benassi F, et al.
Galantamine in frontotemporal dementia and Agomelatine improves apathy in frontotemporal
primary progressive aphasia. Dement Geriatr dementia. Neurodegener Dis 2016; 16: 352–356.
Cogn Disord 2008; 25: 178–185.
134. Golde TE, Lewis J and McFarland NR. Anti-
129. McCarter SJ, St Louis EK and Boeve BF. Sleep tau antibodies: hitting the target. Neuron 2013;
disturbances in frontotemporal dementia. Curr 80: 254–256.
Neurol Neurosci Rep 2016; 16: 85.
135. Li YQ, Tan MS, Yu JT, et al. Frontotemporal
130. Manoochehri M and Huey ED. Diagnosis lobar degeneration: mechanisms and therapeutic
and management of behavioral issues in strategies. Mol Neurobiol 2016; 53: 6091–6105.
frontotemporal dementia. Curr Neurol Neurosci
Visit SAGE journals online 136. Jiang J and Cleveland DW. Bidirectional
Rep 2012; 12: 528–536.
journals.sagepub.com/ transcriptional inhibition as therapy for ALS/
home/tpp
131. Riedl L, Mackenzie IR, Forstl H, et al. FTD caused by repeat expansion in C9orf72.
SAGE journals Frontotemporal lobar degeneration: current Neuron 2016; 92: 1160–1163.

48 journals.sagepub.com/home/tpp

You might also like