Skip to main content
Therapeutic Advances in Psychopharmacology logoLink to Therapeutic Advances in Psychopharmacology
. 2017 Nov 10;8(1):33–48. doi: 10.1177/2045125317739818

Frontotemporal dementia: latest evidence and clinical implications

Juan Joseph Young 1, Mallika Lavakumar 2, Deena Tampi 3, Silpa Balachandran 4, Rajesh R Tampi 5,
PMCID: PMC5761910  PMID: 29344342

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 pharmacological interventions, as there are currently no US Food and Drug administration approved treatments to manage FTD syndromes.

Keywords: classification, frontotemporal dementia, frontotemporal lobar degeneration, genetics, neurobiology, treatment

Introduction

Frontotemporal dementia (FTD) is a term used to describe a group of neurocognitive disorders that encompass progressive dysfunction in executive functioning, behavior, and language. It is considered the third most common form of dementia following Alzheimer’s disease (AD) and dementia with Lewy bodies.1 As per its namesake, it is a cluster of syndromes that result from degeneration of the frontal and temporal lobes, and is subdivided into two categories that are unique in respect to their predominating presentations; namely, the behavioral subtype that accounts for about half of FTD cases, and the language subtype.2 The language subtype is further subdivided into nonfluent and semantic variants of primary progressive aphasia (PPA), which are characterized by diverging localizations and underlying cerebral dysfunction.2,3 The semantic and nonfluent variants of PPA are both characterized by prominent language impairment, but also differ in neurological localization, severity of specific symptoms, and the overall course of their presentations. Illustrative of this, the semantic variant exhibits bilateral anterior temporal lobe atrophy and is associated with dysfunction of emotional processing, compulsions, and decline in language skills. In contrast, the nonfluent variant commonly presents with greater left hemisphere atrophy, and is associated with speech problems earlier while behavioral disturbances develop later in the disease course.3 As the population of geriatric patients grow and neurocognitive disorders become more prevalent, there will be an increased need for physicians with an expert understanding of the diverse clinical findings that define the heterogeneous FTD subtypes. This review will focus on the most recent findings concerning FTD neurobiology, current classification and assessment systems, and the most up-to-date expert consensus on the treatment of this unique collection of syndromes.

Epidemiology

According to the World Health Organization (WHO), there are an estimated 47.5 million people who have dementia with another 7.7 million new cases every year.4 Although AD is the most common form of dementia, contributing to 60–70% of cases, mixed types exist, and the overlap between different forms of dementia is sizeable. FTD is the second most common cause of dementia in patients aged < 65 years,5 with a further 25% of dementia cases in patients older than 65 years also attributed to FTD disorders.6 However, determining the total population with underlying FTD pathology is difficult due to the low disease frequency in a relatively large at-risk population. Additionally, studies estimating incidence and prevalence rates of FTD are limited by the inherent difficulty identifying FTD disorders.6,7 In the USA, Knopman and Roberts7 estimated that the prevalence of FTD between the ages of 45–65 years ranged from 15 to 22 per 100,000 people, with incidence estimates ranging from 2.7 to 4.1 per 100,000 members in the same age range. Overall, they estimated approximately 20,000 to 30,000 cases of FTD in the USA alone. Yet, these numbers may actually be underestimating the occurrence of FTD syndromes. This is especially evident, as older cases of FTD begin to echo symptoms of AD further confounding the delineation between the neurocognitive disorders.6 Further emphasizing the limitations of FTD epidemiologic studies, Lambert and colleagues8 conducted a systematic review with the goal of estimating prevalence rates of early-onset dementia. They found a significant disparity between studies estimating the prevalence of early-onset FTD with estimates ranging from 1.0 to 15.4 per 100,000 members of the population. Adding to this, it appears that the data presented was principally collected from populations studied in the UK, mainland Europe, and Japan, suggesting that the information gathered may not be generalizable to the world-wide population as a whole. For this reason, standardization of study designs will likely aid in more precisely defining FTD epidemiology globally, a focus that future studies should pursue in the future.

Classification and assessment systems

Researchers have developed multiple methodologies to classify FTD disorders using biological and molecular signatures. However, diagnosis is still obtained clinically due to the absence of definitive biomarkers. The international consensus criteria for the behavioral variant of FTD (bvFTD) was published by the International Behavioral Variant FTD Criteria Consortium9 to develop a sensitive standard for early screening and management of bvFTD. To be diagnosed, a patient must show progressive deterioration of behavior or cognition by observation or history provided by a reliable caretaker or informant in order to be diagnosed. Possible bvFTD is diagnosed if three or more of the following are present: (a) early behavioral disinhibition described as socially inappropriate behavior, loss of manners/decorum, impulsivity and rash actions; (b) early apathy or inertia; (c) early loss of empathy or sympathy, including diminished response to other people’s need or sympathies, and diminished social interest; (d) early perseverative or compulsive/ritualistic behavior (i.e. simple repetitive movements, complex, compulsive, or ritualistic behavior, stereotypy of speech); (e) hyperorality and dietary changes, including altered food preferences, binge eating, increased consumption of alcohol and cigarettes, oral exploration or consumption of inedible objects; (f) neuropsychological profile demonstrates deficits in executive functioning and relative sparing of episodic memory and visuospatial functioning. Probable bvFTD diagnosis would need to meet criteria for possible bvFTD, but also includes imaging suggestive of frontal or anterior atrophy on magnetic resonance imaging (MRI) or computed tomography (CT). Alternatively, positron emission tomography (PET) or single-photon emission computed tomography (SPECT) imaging demonstrating hypoperfusion or hypometabolism in the frontal or anterior temporal regions could be used to diagnose probable bvFTD. Definitive bvFTD with definite FTD pathology is diagnosed when a patient meets criteria for possible bvFTD and has one or both of the following: histopathological evidence of FTD or if there is evidence of a known pathogenic mutation. bvFTD is excluded if a patient’s presentation is better accounted for by other nondegenerative nervous system or medical disorders, a psychiatric diagnosis, or biomarkers are strongly indicative of other neurocognitive disorders or neurodegenerative processes such as AD.9

Gorno-Tempini and colleagues developed a common framework for the classification of PPA subtypes to provide consistency in clinical diagnosis and standardize PPA assessments for future clinical studies.10 According to this framework, PPA is diagnosed when a patient exhibits prominent difficulty with language wherein the deficits are the principal cause of impairment of daily activities, with aphasia being the most prominent deficit at symptom onset and during the initial phase of the disease. PPA diagnosis is excluded if the presenting symptoms are more consistent with other neurocognitive disorders, medical conditions, neurodegenerative processes, or a psychiatric diagnosis. PPA is also excluded if there are prominent initial episodic memory, visual memory, visuospatial impairments, and initial behavioral disturbance. Patients may be diagnosed with the semantic variant of PPA (svPPA) if they exhibit impaired confrontation naming (i.e. difficulty naming or recognizing objects or drawings) and impaired single-word comprehension, and have at least three of the following: (a) impaired object knowledge; (b) surface dyslexia (i.e. inability to recognize words as a whole) or dysgraphia; (c) spared repetition; (d) spared speech production. For an imaging-supported diagnosis, there must either be predominant anterior temporal lobe atrophy or predominant anterior temporal hypoperfusion/hypometabolism on SPECT or PET. For a definite pathology diagnosis, there must be histopathologic evidence or the presence of a known pathogenic mutation.

The nonfluent variant of PPA (nfvPPA) diagnosis requires at least one of two core features (agrammatism in language production or effortful, halting speech not consistent with apraxia of speech), and two out of three of the following features: (a) impaired comprehension of complex sentences; (b) spared single-word comprehension; (c) spared object knowledge. Imaging-supported nfvPPA variant diagnosis is obtained when there is imaging that demonstrates predominant left posterior-fronto-insular atrophy on MRI or predominantly left posterior fronto-insular hypoperfusion or hypometabolism on SPECT or PET. Alternatively, nfvPPA with definite pathology diagnosis is present when there is histopathologic evidence or the presence of a known pathogenic mutation.

Like other PPA subtypes, individuals with the logopenic variant of PPA exhibit language impairment but with more specific difficulties in confrontation naming and syntax. These individuals are more likely to have speech interrupted with filling sounds, frequent pauses, phonological errors, and impaired sentence repetition.11 The diagnosis of logopenic variant PPA requires the following core features to be present: impaired single-word retrieval in spontaneous speech and naming, and impaired repetition of sentences and phrases. Additionally, three of four of the following features must also be present for a diagnosis: (a) speech (phonologic) errors in spontaneous speech and naming; (b) spared single-word comprehension and object knowledge; (c) spared motor speech; (d) absence of frank agrammatism. As with other PPA-variant diagnoses, imaging-supported diagnosis is made with corresponding abnormalities in imaging. In the logopenic variant, MRI, SPECT, or PET scans must demonstrate predominant left posterior perisylvian or parietal atrophy/hypoperfusion/hypometabolism. Definite histopathologic evidence or the presence of a known pathogenic mutation would allow for a definite pathology specifier.

Familial frontotemporal dementia caused by frontotemporal dementia-associated mutations

FTD is a highly heritable disorder despite varying heritability among different clinical syndromes and subtypes due to a range of gene mutations.12 Up to half of FTD cases with autosomal-dominant inheritance report a family history of FTD.13 Familial FTD also accounts for approximately one third to half of all FTD cases and presents more commonly as bvFTD than other FTD subtypes.6,14 Mutations in microtubule-associated protein tau (MAPT),1525 progranulin (PGRN),2638 and chromosome 9 open reading frame 72 (C9orf72) expansion mutations32,3950 have been found to be the most common causes of familial FTD.51,52 Although FTD presentations are relatively homogenous early in the disease course, different biological correlates and varying genetic mutations ultimately result in diverging clinical courses.53 MAPT-associated familial FTD typically presents younger than FTD associated with other mutations, and presents as bvFTD with or without Parkinsonian symptoms, as well as with or without language decline. MAPT mutations occur on average at a frequency of 6–11% of all FTD subjects, is inherited in an autosomal-dominant manner, has high penetrance, and may have a shorter duration of illness relative to other mutations.17,54,55 The most prominent behavioral features with MAPT mutations include disinhibition, stereotyped repetitive behaviors, and obsessions. Notably, apathy is less common than in PGRN and C9orf72 cases. Semantic impairment is more common later during the disease course. Episodic memory loss may occur and mimic early onset AD.56 MAPT mutations have been implicated in the formation of hyperphosphorylated tau in cortical and subcortical grey and white matter suggesting a neuroanatomical correlation in the development of clinical symptoms.55

PGRN mutations present with more variable clinical presentations, but most commonly present as bvFTD and less commonly as a nfvPPA presentation.57 Mutations occur at a frequency of 5–20% in the FTD population, are inherited in an autosomal-dominant manner, and have relatively low penetrance until the age of 70.17,58,59 Apathy and social withdrawal are prominent in their presentation, and 10–30% of cases may present with episodic memory impairment.60 Neuropsychiatric symptoms are common and patients may report delusions, hallucinations, or exhibit ritualistic behaviors. There is also a predominance of early language involvement compared with MAPT or C9orf72 mutations. Extrapyramidal symptoms occur between 40% and 60% of PGRN-associated FTD cases.57,60 Interestingly, PGRN mutations, though thought to be pathogenic, do not appear to increase the risk of developing FTLD.54

FTD patients with the C9orf72 expansion mutation present more commonly as bvFTD, motor neuron disease, or a combination of the two, although clinical presentations tend to be heterogeneous early in the disease process.41,61 Apathy, disinhibition, and loss of empathy are prominent behavioral features in this type of familial FTD.62 Complex stereotyped behaviors are common, but language decline is rarer compared with patients with MAPT or PGRN mutations. Patients typically have profound disruption of executive functioning, may have reduced spontaneous speech, echolalia, perseveration, impaired visual and verbal episodic memory, apraxia, anomia, and dyscalculia.63 C9orf72 expansion mutations exhibit an autosomal-dominant pattern of inheritance, is implicated in disease anticipation that affect successive generations,44 and is considered the most common cause of ALS-FTD (amyotrophic lateral sclerosis associated with frontotemporal dysfunction).42,43,49 However, the minimum repeat length required to indicate increased risk of disease is unknown.47 These expansion mutations are highly associated with transactive response DNA-binding protein (TDP-43) pathology, and the presence of neuronal cytoplasmic and intranuclear inclusions that are immunoreactive to ubiquitin proteasome system markers. A confounding factor to consider when diagnosing this subtype is the possibility of AD-like biomarkers present in the cerebrospinal fluid.60 Psychiatric presentations, including delusions, auditory/visual/tactile hallucinations, somatoform symptoms, agitation, and anxiety may be observed. Parkinson-like symptoms are common and may present as gait disturbance, tremor, akinesis, and rigidity. Most patients do not respond to levodopa when administered.60,62

Two other studied pathognomonic mutations found in a minority of cases include mutations in vasolin-containing protein (VCP) and transactive DNA-binding protein (TARDBP).64 VCP mutations have been implicated in ubiquitin–proteasome pathway dysfunction, disruption of TDP-43 localization, and inducing neuronal apoptosis.65 Individuals with VCP mutations historically present as the triad of autosomal-dominant hereditary inclusion body myopathy with Paget’s disease of the bone and frontotemporal dementia (IBMPFD).6669 FTD symptoms typically include behavioral changes, executive dysfunction, and aphasia.70 Consequently, a number of European and Asian cases detailing progressive myopathy with signs and symptoms consistent with FTD have been associated with VCP mutations.7177 Of particular note is one study78 that investigated how the location of VCP gene mutations in 27 families related to the progression and severity of symptoms. Critically, the investigators found that the onset of neurocognitive deterioration foreshadowed a rapid progression of the disease leading to an average life span of 6 years.

Alternatively, TARDBP was found to be a major component of ubiquitin-positive and tau-negative inclusions in patients that have been diagnosed with both amyotrophic lateral sclerosis (ALS) and FTD.79 TARDBP has also been implicated in both behavioral and semantic FTD cases with co-occurring motor neuron disease (MND).80 However, other studies have reported cases of individuals with TARDBP mutations that have developed FTD without the progression of ALS or MND symptoms.81,82 Clinical presentations of patients with TARDBP mutations are highly heterogeneous and may present with apathy, poor executive functioning, poor language comprehension, or disinhibition.83 It has been suggested that mutations of TARDBP have led to increased translocation of TDP-43 causing dysregulation of neuronal endoplasmic reticulum calcium signaling resulting in an increase in B-cell lymphoma 2 (Bcl-2) mediated neuronal apoptosis.84

Neurobiology

FTD is classified into distinct, major subtypes based on their respective protein-based inclusions and underlying molecular pathologies. Namely, they are separated into the tau-based frontotemporal lobar degeneration (FTLD)-tau subtype, the FTLD-TDP subtype associated with TDP-43, the FTLD-FET subtype that is associated with the fused in sarcoma (FUS) proteinopathy and other FET [i.e. FUS, EWSR1 (EWS RNA-binding protein 1), TAF15 (TATA-box binding protein associated factor 15)] family proteins, and FTLD-ubiquitin–proteasome system (UPS).59,85 (See Table 1 for a summary of FTD tauopathies) FTLD-tau accounts for approximately 40% of all FTD cases, and is characterized by misfolded tau tangles similar to those found in AD.86 The tauopathy present in this variant involves hyperphosphorylated tau protein that forms insoluble filamentous inclusions, which adversely affect axonal transport regulation, microtubule formation, and microtubule stabilization. These changes lead to a decrease in microtubule binding and an increase in tau aggregation, the latter of which induces neurotoxicity and nerve cell dysfunction.87 Tau aggregates in FTLD-tau subtypes vary in the degree of phosphorylation and the amount of tau isoforms they contain. This has prompted their subclassifications into predominantly 3R or 4R (e.g. corticobasal degeneration, progressive supranuclear palsy, argyrophilic grain disease, globular glial tauopathy) tau isoform disorders.59,88 These subtypes of FTLD tauopathies also have distinct anatomical and histopathological correlates. In general, FTD is associated with severe neuronal pathology, and spherical argyrophilic neuronal cytoplasmic inclusions composed primarily of 3R tau isoforms.64 Progressive supranuclear palsy (PSP), a movement disorder that presents with rigidity, bradykinesia, and ophthalmoplegia, is characterized by significant degeneration of subcortical regions including the subthalamic nucleus, midbrain, substantia nigra, globus pallidus, and striatum. Postmortem examinations of PSP patients demonstrate spherical and flame-shaped neurofibrillary tangles, tufted astrocytes, and inclusions composed predominantly of the 4R tau isoform.89 FTD tauopathies may also exhibit the corticobasal syndrome, which may present as rigidity, bradykinesia, dystonia, apraxia, and alien limb phenomenon, is characterized by substantia nigra depigmentation, globus pallidus atrophy, and asymmetric and focal cortical atrophy. This syndrome exhibits similar traits compared with PSP, but develop more white matter involvement and has unique ring-shaped collections called astrocytic plaques caused by tau accumulation in astrocytes.59,89

Table 1.

Frontotemporal dementia tauopathies.59

Tauopathies Presentation Histopathology Tau isoform
Pick’s disease bvFTD or nfvPPA Pick bodies, argyrophilic neuronal cytoplasmic inclusions 3R
Progressive supranuclear palsy Rigidity, ophthalmoplegia, bradykinesia Spherical and flame-shaped neurofibrillary tangles, tufted astrocytes 4R
Corticobasal degeneration Rigidity, bradykinesia, dystonia, apraxia, alien limb phenomenon White matter degeneration with ring-shaped astrocytic plaques

FTD, frontotemporal dementia; bvFTD, behavioral variant FTD; nfvPPA, nonfluent variant primary progressive aphasia.

Alternatively, neuronal inclusions immunoreactive to ubiquitin are characteristic of the FTLD-TDP and FTLD-FET forms.85,90 In FTLD-TDP, these neuronal inclusions are primarily composed of TDP-43, which has been associated with multiple genetic markers thought to be pathognomonic of FTD. These include mutations of PGRN, C9orf72, and VCP.86,91 In mice, accumulation of the insoluble, phosphorylated cytoplasmic TDP-43 and loss of nuclear TDP-43 is associated with brain atrophy, muscle denervation, motor neuron loss, and progressive motor impairments that are commonly observed both in FTLD-TDP and ALS suggesting a common neuropathological pathway.92 FTLD-TDP is further subdivided into Types A, B, C, and D that differ in the constitution of pathological inclusions and their distribution within the cortices.93 Patients with FTLD-TDP Type A have predominantly neuronal cytoplasmic inclusions and dystrophic neurites with TDP-43 immunoreactive lesions in the second layer of the cortices. FTLD-TDP type B features more neuronal cytoplasmic inclusions in the superficial and deeper cortical layers. FTLD-TDP type C has a significant amount of dystrophic neurites demonstrating a ‘corkscrew appearance’ through the cortical layers. FTLD-TDP type D is associated with multiple neuronal intranuclear inclusions and dystrophic neurites with fewer neuronal cytoplasmic inclusions.85,93 Protein immunoreactivity to members of the FET protein family, with FUS being the more well-known FTD-associated proteinopathy, is the last major subgroup of FTD pathology, and is associated with co-aggregation of FET proteins into ubiquitinated inclusions found in severe cases of sporadic FTD.59,86 Of note, FUS protein involvement in the pathology of certain FTD subtypes is suggestive of a common neuropathologic mechanism and close relationship between the FTD disorders and ALS, since FUS has been described as a common cause of autosomal-dominant ALS.92,94 Interestingly, FTD-associated proteinopathies including tau, TDP-43, and FUS have demonstrated aggregation and seeding behavior in both in vivo and in vitro models, suggesting FTD pathologies have prion-like characteristics.86,87 Lastly, the FTLD-UPS subtype is characterized by ubiquitin-immunoreactive inclusions that do not label for tau, TDP-43, or FET proteins.12,9597 This subtype is commonly associated with a mutation in the charged multivesicular body protein 2B (CHMP2B) gene that leads to dysfunction in the endosomal ESCRTIII complex that causes disruption of endosomal traffic.98101 A decline in levels of CHMP2B results in a decrease of hippocampal dendritic branching and reduced excitatory synapse activity effectively diminishing synaptic plasticity and causing neurodegenerative deficits.102 This subtype typically presents with changes in behavior and personality (i.e. disinhibition, apathy, loss of emotion, dyscalculia, progressive aphasia that leads to mutism), as well extrapyramidal motor symptoms as the disease progresses.98 However, CHMP2B mutations are relatively rare,103 and do not appear to increase the risk of development of FTD.104

Neuroimaging

A recent review105 of neuroimaging studies have detailed advances in quantifying brain atrophy in FTD using volumetric MRI measurements and by observing hypometabolism in specific regions using [18F] fluorodeoxyglucose PET (18F-FDG PET). Volumetric MRI studies they reviewed focused mainly on cerebral loss of grey matter with the goal of improving accurate diagnosis and characterization of FTD variants. In these studies, MRI imaging demonstrated disruption of the salience network, default mode network, left hemisphere language and semantic networks, and executive control networks that imply pathoanatomical relationships in FTD syndromes. Notably, one study has demonstrated how baseline salience network resting state activity may predict behavioral changes in FTD.106 MRI studies of bvFTD patients in particular show losses in the prefrontal cortex, anterior temporal regions, insula, anterior cingulate, striatum, and thalamus, which differentiate bvFTD from AD with relatively high sensitivity and specificity. A recent retrospective observational study also suggests that there are specific anatomical phenotypes based on the salience and semantic appraisal networks that may contribute in classifying the otherwise heterogeneous bvFTD subgroups.107 The svPPA exhibits more asymmetric temporal lobe atrophy on imaging scans with primarily anterior and inferior regional atrophy more commonly seen in the left hemisphere. The earliest changes observed are grey matter losses in the inferior temporal and fusiform gyri, temporal pole, and parahippocampal entorhinal cortex. Notably, temporal pole atrophy could be seen on MRI imaging despite a patient’s continued ability to maintain activities of daily functioning. However, the severity of functional decline does later correlate with the extent of atrophy as it progresses to orbitofrontal, inferior frontal, insular, and anterior cingulate cortices anteriorly. While both variants of PPA share the trait of primarily left hemisphere atrophy, the nfvPPA differs from svPPA in distribution during the course of the disease. In nfvPPA, atrophic losses may begin in the inferior frontal gyrus (particularly in the pars opercularis), dorsolateral prefrontal cortex, superior temporal gyrus, and insula. Over time, these losses will include the prefrontal and temporal lobe structures in the right hemisphere. Ipsilateral anterior frontal, lateral temporal, and anterior parietal lobes are also affected. Lastly, the logopenic PPA subtype has a more posterior profile involving the left temporoparietal and posterior cingulate atrophy in the early stages of disease progression. In summary, bvFTD subtypes present with frontal and temporal lobe pathology that includes key tracts such as the corpus callosum and cingulum, while PPA subtypes are more focal and asymmetric presenting with white matter changes in specific networks associated with language processing. Other pathologically defined forms also exhibit specific atrophic patterns. PSP is associated with midbrain atrophy, and TDP-43 proteinopathies present with a variation of symmetric and asymmetric atrophic findings according to the underlying TDP type.

18F FDG-PET imaging studies also demonstrate patterns of hypometabolism that correlate with areas of atrophy, and are useful in differentiating AD from FTD. In FTD, PET studies typically demonstrate hypometabolism in the anterior frontotemporal regions including the cingulate gyri, uncus, insula, subcortical areas, basal ganglia, and medial thalamic regions. Hypometabolism is limited to frontal, parietal, and temporal cortices during the early stages of the disease but spreads outwards as the disorder progresses. Orbitofrontal, dorsolateral, medial prefrontal cortex, and anterior poles display hypometabolism in bvFTD patients’ PET scans. Likewise, svPPA patients exhibit more asymmetrical presentations with exclusively left temporal lobe hypometabolism, nfvPPA imaging shows hypometabolism in the left frontal and superior temporal regions, and logopenic variants demonstrate a left parietotemporal hypometabolism extending to anterior temporal and frontal regions.105,108 However, it is important to note that there is considerable individual variability when using imaging to classify FTD disorders as a consequence of heterogeneity in genetic associations and the underlying pathological causes that are yet to be fully understood. Consequently, neurobiological and imaging markers remain ancillary modalities in the diagnosis and classification of FTD subtypes in contrast to the gold standard of clinical evaluations. Please see Table 2 for a summary of neuroimaging findings commonly found in FTD variants.

Table 2.

Frontotemporal dementia neuroimaging correlations.105

Variant MRI findings PET/SPECT findings
bvFTD Atrophy in prefrontal cortex, anterior temporal regions, anterior cingulate, the striatum, thalamus, the insula Decreased FA in uncinate fasciculus, genu of corpus callosum and cingulum, superior and inferior longitudinal fasciculi, inferior fronto-occipital fasciculus
nfvPPA Predominant left hemisphere cortical loss; atrophy predominantly affecting pars opercularis in the inferior frontal gyrus extending to prefrontal and temporal cortices, caudate and putamen atrophy bilaterally Decreased FA in left uncinate fasciculus, inferior longitudinal fasciculus tracts, corpus callosum, cingulum; notable sparing of occipital white matter, brainstem, cerebellum
svPPA Predominant left hemisphere cortical loss; grey matter loss predominantly in inferior temporal and fusiform gyri, temporal pole, parahippocampal cortex, entorhinal cortex; losses extend to anterior cingulate, orbitofrontal, inferior frontal, and insular cortices Decreased FA in left superior longitudinal fasciculus, corpus callosum, left cingulum, left orbitofrontal, inferior frontal, anterior temporal, inferior parietal white matter regions
Logopenic PPA Atrophy of left temporoparietal and posterior cingulate Notable white matter losses in left 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.

Management and treatment

There is a lack of quality studies and reports concerning the management of problematic behaviors in FTD with mainly sparse case reports and case series detailing their effectiveness. Nevertheless, nonpharmacological strategies are considered the preferred intervention before resorting to pharmacological therapies that may exacerbate medical comorbidities that affect more elderly patients. The main purpose of nonpharmacological interventions is to prevent disruptive behaviors, provide symptom relief, and lessen caregiver distress. Environmental approaches aim to decrease irritability, aggression, and anxiety that arise from patients’ difficulty processing information from the array of daily stimuli. These interventions include reducing noise, limiting stimulation, simplifying social parameters by limiting interaction to small groups of people, and facilitating the removal of complicated daily activities that may confuse, and therefore agitate, patients. There should be special care in implementing hearing aids, and optimizing sensory stimulation to limit patient discomfort. Introducing games or old hobbies may reduce disinhibition and inappropriate behavior. Exercise has been suggested to reduce behavioral symptoms as well. Confronting and challenging psychotic symptoms may lead to more agitation, so reassurance and distracting patients are considered to be more acceptable alternatives in decreasing behavioral symptoms. Clinicians have attempted to modify compulsions, apathy, and lack of motivation using reward systems, although there are limited data concerning this intervention. Cognitive behavioral therapy is currently being studied as a possible option to modulate behavior, but studies are still preliminary.109,110

There are currently no US Food and Drug Administration (FDA) approved pharmacological therapies for FTD.111 There is limited evidence that antidepressants such as selective serotonin reuptake inhibitors (SSRIs) may be useful for impulsivity, irritability, eating behavior, and disinhibition.112 For example, one small, randomized-controlled, open-label 14-month trial113 reported improvement of behavioral symptoms in subjects with ages ranging from 64 to 68 years after being prescribed paroxetine at dosages of up to 20 mg/day. Notably, subjects tolerated paroxetine well and exhibited improvement in Neuropsychiatric Inventory (NPI) scoring. However, a follow-up study114 using placebo controls with a similarly small subject population determined that paroxetine administration at dosages up to 40 mg/day provided little objective improvement and may have caused impairment in several neuropsychological tests including visual discrimination tasks, errors in paired-associates learning task, and delayed pattern recognition memory accuracy. Discrepancies between study results were attributed to subjects responding more favorably to lower doses of paroxetine suggesting a higher rate of tolerability at 20 mg daily compared with higher dosages. Additionally, paroxetine may be more effective when prescribed earlier in the disease process, although it is difficult to determine without more in-depth and higher-powered studies.

Citalopram is another SSRI that has been recently shown to provide some improvement in FTD patient behavior. Herrmann and colleagues found improvement with a citalopram challenge at 40 mg daily dosages that led to a decrease in behavioral symptoms including irritability, depression, apathy, and disinhibition, while also improving overall NPI scores.115 More recently, in a randomized double-blinded placebo-controlled crossover study, bvFTD patients who were prescribed 30 mg daily dosages of citalopram demonstrated improved disinhibition symptoms in addition to a partial restoration of serotonergic neurotransmission in dysfunctional prefrontal cortical systems as measured by magnetoencephalography and electroencephalography.116

Studies of sertraline efficacy in treating FTD symptoms are limited to mainly observational studies. One study comparing stereotypical behavior in FTD and AD subjects suggested that stereotypical movements in FTD could be decreased with sertraline administration at a dosage range of 50–100 mg/day.117 A case study of a 53-year-old ALS-FTD patient indicated that sertraline treatment could also decrease inappropriate sexual behavior in addition to decreasing aggressive behavior which was helpful in minimizing caregiver burden.118

Alternatively, treatment with trazodone at dosages of at least 300 mg/day over 12 weeks has been reported to be helpful in decreasing symptoms of problematic eating, agitation, irritability, dysphoria, and depression, although adverse events were more prominent in subjects being prescribed the psychotropic. Typical emergent side effects included fatigue, dizziness, and hypotension, but the authors reported that these symptoms were mild in severity.119 At least one open-label 12-week trial showed a treatment response with fluvoxamine administration from a dosage range of 50–150 mg/day (mean dose was 110 mg/day) improving stereotypic behavior, eating behavior, and roaming behavior.120

Stimulants have been proposed as alternative treatments due to their mechanism of action in elevating extracellular catecholamine concentrations to improve cognitive functioning and lessen frontostriatal and orbitofrontal dysfunction reported to affect risk-taking behavior in FTD patients.121123 Exemplifying this, one study123 found that eight patients who were prescribed methylphenidate at 40 mg daily dosages demonstrated decreased risk-taking behavior with overall improvement in general behavior. Additionally, a randomized double-blinded crossover trial comparing dextroamphetamine and quetiapine use in FTD patients found that dextroamphetamine administration decreased disinhibition and apathy symptoms, while being more tolerable than quetiapine, which caused sedation in their subject population.124 Despite these studies suggesting the safety and efficacy of stimulants in treating specific symptoms common in the FTD population, a clinician’s medical decision-making process should take into account the limited data provided by these stimulant studies due to their low power and questionable generalizability, as well as possible side effects and tolerability issues of stimulants in more elderly patients.

Memantine and cholinesterase inhibitors have not demonstrated clinically significant efficacy in treating FTD patients, and may also hasten cognitive decline or worsen behavioral symptoms.125,126 This is likely due to a lack of consistent evidence of a cholinergic deficit in FTD without concomitant pathology of other major neurocognitive disorders.127 However, at least one study reported some efficacy in improving behavioral symptoms with galantamine treatment in an aphasic subgroup of FTD without a preponderance of serious adverse events, although no effect was found in overall treatment of the general FTD subject population.128

Complaints of insomnia may be due to behavioral dysfunction or neuronal degeneration causing impairment of the sleep/wake cycle. However, primary sleep disorders such as restless leg syndrome may also contribute to insomnia and can be treated with alpha-2-delta calcium channel ligands (e.g. gabapentin, pregabalin) or dopaminergic agents. Agitation may be controlled by atypical antipsychotics such as risperidone, olanzapine, quetiapine, and clozapine, but they are not FDA-approved and could be detrimental in a dementia-associated agitation due to increased risk of mortality and unwanted side effects.111,129,130 Of note, antipsychotics have been suggested to be more helpful in treating psychotic symptoms in patients with the C9orf72 subtype of FTD, but once again, data is limited with increased risk of extrapyramidal side effects and morbidity in dementia patients preventing safe utilization of these neuroleptics.131 Consequently, due to the limited data and power of pharmacologic treatment studies in FTD populations; nonpharmacological interventions and caregiver support are the preferred management options.125

Due to the limited contemporary psychopharmacologic interventions available to physicians, several novel approaches are being researched to develop more effective treatments for FTD symptoms. One such approach is the use of the neuropeptide, oxytocin, to aid in mediating behavior and improving apathy and empathy in svPPA patients.132 Another study suggested agomelatine, a melatonin receptor agonist, may improve apathy in FTD patients.133 A preclinical trial reported active immunization against tau aggregates using anti-tau antibodies may improve cognition through decreasing tauopathy.134 A study investigating the effectiveness of inhibiting tau aggregation in bvFTD patients using leuco-methylthioninium was recently completed, but detailed results are pending release and publication.135 Other targets for therapy include disrupting the downstream effects of PGRN and C9orf72 mutations.111 For example, antisense oligonucleotides are being used to target the transcription elongation factor SUPT4H1/SUPT5H to bi-directionally suppress C9orf72 RNA transcription.136 Other proposed methods involve elevating PGRN levels through molecular manipulation of biological pathways using suberoylanilide hydroxamic acid, androgen treatment, and bafilomycin A1.135

Conclusion

The entirety of the FTD clinical syndrome is now known to be composed of complex and heterogeneous variants that vary in presentation, genetic alterations, and pathological processes. Consequently, there is a need for physicians to become well versed in the diagnostic criteria set forth by consortiums and expert consensus in order to properly assess and manage FTD patients. According to recent epidemiologic studies, FTD is likely to be underdiagnosed due to the inherent difficulty accurately diagnosing these disorders. These findings underline the need to standardize and update current criteria to increase diagnostic sensitivity, and allow for better management of these variants at an earlier stage of the disease course. There have been several breakthroughs in genetic and neuroimaging studies that have elucidated the pathophysiological characteristics of these FTD subtypes in the past decade. However, FTD pathology is still relatively unknown and requires further investigations to produce more effective diagnostic biomarkers and therapies, especially as later course presentations begin to mimic AD symptomatology. Current management guidelines recommend a focus on nonpharmacological interventions, as these have been found to be the most studied and effective practices at the disposal of clinicians. Contemporary pharmacological treatment options are still lacking due to a deficit of comprehensive randomized controlled trials to determine efficacy and safety, although there is limited evidence of SSRI efficacy in decreasing FTD-specific symptomatology and difficult behavior. Additionally, the extensive list of symptoms that are being targeted for intervention typically necessitates a variety of therapies for clinical improvement, which fundamentally complicates treatment planning. However, this renewed interest in FTD research gives hope for future advancements that aim to increase diagnostic capabilities, and expand treatment options through novel therapeutic targets.

Footnotes

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of interest statement: Mallika Lavakumar received honorarium from Oakstone and receives research funding from the Health Research Services Administration. Rajesh R Tampi receives honorarium from Oakstone and royalties from Lippincott Williams & Wilkins.

Contributor Information

Juan Joseph Young, Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA.

Mallika Lavakumar, Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA.

Deena Tampi, Mercy Regional Medical Center, 3700 Kolbe Rd, Lorain, OH 44053, USA.

Silpa Balachandran, Department of Psychiatry, MetroHealth Medical Center, Cleveland, OH, USA Case Western Reserve University, Cleveland, OH, USA.

Rajesh R. Tampi, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Drive, Cleveland, OH 44109, USA.

References

  • 1. Bang J, Spina S, Miller BL. Frontotemporal dementia. Lancet 2015; 386: 1672–1682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Kurz A, Kurz C, Ellis K, et al. What is frontotemporal dementia? Maturitas 2014; 79: 216–219. [DOI] [PubMed] [Google Scholar]
  • 3. Bott NT, Radke A, Stephens ML, et al. Frontotemporal dementia: diagnosis, deficits and management. Neurodegener Dis Manag 2014; 4: 439–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. World Health Organization. Dementia fact sheet, http://www.who.int/mediacentre/factsheets/fs362/en/ (2016). Accessed January 22, 2017.
  • 5. Rosso SM, Donker Kaat L, Baks T, et al. Frontotemporal dementia in The Netherlands: patient characteristics and prevalence estimates from a population-based study. Brain 2003; 126: 2016–2022. [DOI] [PubMed] [Google Scholar]
  • 6. Onyike CU, Diehl-Schmid J. The epidemiology of frontotemporal dementia. Int Rev Psychiatry 2013; 25: 130–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Knopman DS, Roberts RO. Estimating the number of persons with frontotemporal lobar degeneration in the US population. J Mol Neurosci 2011; 45: 330–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Lambert MA, Bickel H, Prince M, et al. Estimating the burden of early onset dementia; systematic review of disease prevalence. Eur J Neurol 2014; 21: 563–569. [DOI] [PubMed] [Google Scholar]
  • 9. Rascovsky K, Hodges JR, Knopman D, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain 2011; 134: 2456–2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Gorno-Tempini ML, Hillis AE, Weintraub S, et al. Classification of primary progressive aphasia and its variants. Neurology 2011; 76: 1006–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Leyton CE, Hodges JR. Towards a clearer definition of logopenic progressive aphasia. Curr Neurol Neurosci Rep 2013; 13: 396. [DOI] [PubMed] [Google Scholar]
  • 12. Rohrer JD, Guerreiro R, Vandrovcova J, et al. The heritability and genetics of frontotemporal lobar degeneration. Neurology 2009; 73: 1451–1456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Rademakers R, Neumann M, Mackenzie IR. Advances in understanding the molecular basis of frontotemporal dementia. Nat Rev Neurol 2012; 8: 423–434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Capozzo R, Sassi C, Hammer MB, et al. Clinical and genetic analyses of familial and sporadic frontotemporal dementia patients in Southern Italy. Alzheimers Dement 2017; 13: 858–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Moreno F, Indakoetxea B, Barandiaran M, et al. The unexpected co-occurrence of GRN and MAPT p.A152T in Basque families: clinical and pathological characteristics. PLoS One 2017; 12: e0178093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Rademakers R, Cruts M, van Broeckhoven C. The role of tau (MAPT) in frontotemporal dementia and related tauopathies. Hum Mutat 2004; 24: 277–295. [DOI] [PubMed] [Google Scholar]
  • 17. Boeve BF, Hutton M. Refining frontotemporal dementia with parkinsonism linked to chromosome 17: introducing FTDP-17 (MAPT) and FTDP-17 (PGRN). Arch Neurol 2008; 65: 460–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Tacik P, Sanchez-Contreras M, DeTure M, et al. Clinicopathologic heterogeneity in frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) due to microtubule-associated protein tau (MAPT) p.P301L mutation, including a patient with globular glial tauopathy. Neuropathol Appl Neurobiol 2017; 43: 200–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Biswas MH, Almeida S, Lopez-Gonzalez R, et al. MMP-9 and MMP-2 contribute to neuronal cell death in iPSC models of frontotemporal dementia with MAPT mutations. Stem Cell Reports 2016; 7: 316–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Ghetti B, Oblak AL, Boeve BF, et al. Invited review: frontotemporal dementia caused by microtubule-associated protein tau gene (MAPT) mutations: a chameleon for neuropathology and neuroimaging. Neuropathol Appl Neurobiol 2015; 41: 24–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Ehrlich M, Hallmann AL, Reinhardt P, et al. Distinct neurodegenerative changes in an induced pluripotent stem cell model of frontotemporal dementia linked to mutant TAU protein. Stem Cell Reports 2015; 5: 83–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Lee SE, Tartaglia MC, Yener G, et al. Neurodegenerative disease phenotypes in carriers of MAPT p.A152T, a risk factor for frontotemporal dementia spectrum disorders and Alzheimer disease. Alzheimer Dis Assoc Disord 2013; 27: 302–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Coppola G, Chinnathambi S, Lee JJ, et al. Evidence for a role of the rare p.A152T variant in MAPT in increasing the risk for FTD-spectrum and Alzheimer’s diseases. Hum Mol Genet 2012; 21: 3500–3512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Rovelet-Lecrux A, Hannequin D, Guillin O, et al. Frontotemporal dementia phenotype associated with MAPT gene duplication. J Alzheimers Dis 2010; 21: 897–902. [DOI] [PubMed] [Google Scholar]
  • 25. Hutton M, Lendon CL, Rizzu P, et al. Association of missense and 5’-splice-site mutations in tau with the inherited dementia FTDP-17. Nature 1998; 393: 702–705. [DOI] [PubMed] [Google Scholar]
  • 26. Sudre CH, Bocchetta M, Cash D, et al. White matter hyperintensities are seen only in GRN mutation carriers in the GENFI cohort. Neuroimage Clin 2017; 15: 171–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Kao AW, McKay A, Singh PP, et al. Progranulin, lysosomal regulation and neurodegenerative disease. Nat Rev Neurosci 2017; 18: 325–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Jackman K, Kahles T, Lane D, et al. Progranulin deficiency promotes post-ischemic blood-brain barrier disruption. J Neurosci 2013; 33: 19579–19589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Paternico D, Premi E, Gazzina S, et al. White matter hyperintensities characterize monogenic frontotemporal dementia with granulin mutations. Neurobiol Aging 2016; 38: 176–180. [DOI] [PubMed] [Google Scholar]
  • 30. Baker M, Mackenzie IR, Pickering-Brown SM, et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006; 442: 916–919. [DOI] [PubMed] [Google Scholar]
  • 31. Caroppo P, Le Ber I, Camuzat A, et al. Extensive white matter involvement in patients with frontotemporal lobar degeneration: think progranulin. JAMA Neurol 2014; 71: 1562–1566. [DOI] [PubMed] [Google Scholar]
  • 32. Lashley T, Rohrer JD, Mahoney C, et al. A pathogenic progranulin mutation and C9orf72 repeat expansion in a family with frontotemporal dementia. Neuropathol Appl Neurobiol 2014; 40: 502–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Chen-Plotkin AS, Unger TL, Gallagher MD, et al. TMEM106B, the risk gene for frontotemporal dementia, is regulated by the microRNA-132/212 cluster and affects progranulin pathways. J Neurosci 2012; 32: 11213–11227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Tapia L, Milnerwood A, Guo A, et al. Progranulin deficiency decreases gross neural connectivity but enhances transmission at individual synapses. J Neurosci 2011; 31: 11126–11132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Gijselinck I, Van Broeckhoven C, Cruts M. Granulin mutations associated with frontotemporal lobar degeneration and related disorders: an update. Hum Mutat 2008; 29: 1373–1386. [DOI] [PubMed] [Google Scholar]
  • 36. van Swieten JC, Heutink P. Mutations in progranulin (GRN) within the spectrum of clinical and pathological phenotypes of frontotemporal dementia. Lancet Neurol 2008; 7: 965–974. [DOI] [PubMed] [Google Scholar]
  • 37. Cruts M, Gijselinck I, van der Zee J, et al. Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21. Nature 2006; 442: 920–924. [DOI] [PubMed] [Google Scholar]
  • 38. Snowden JS, Pickering-Brown SM, Mackenzie IR, et al. Progranulin gene mutations associated with frontotemporal dementia and progressive non-fluent aphasia. Brain 2006; 129: 3091–3102. [DOI] [PubMed] [Google Scholar]
  • 39. Mahoney CJ, Beck J, Rohrer JD, et al. Frontotemporal dementia with the C9ORF72 hexanucleotide repeat expansion: clinical, neuroanatomical and neuropathological features. Brain 2012; 135: 736–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Majounie E, Renton AE, Mok K, et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol 2012; 11: 323–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Hsiung GY, DeJesus-Hernandez M, Feldman HH, et al. Clinical and pathological features of familial frontotemporal dementia caused by C9ORF72 mutation on chromosome 9p. Brain 2012; 135: 709–722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 2011; 72: 245–256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Renton AE, Majounie E, Waite A, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 2011; 72: 257–268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Van Mossevelde S, van der Zee J, Gijselinck I, et al. Clinical evidence of disease anticipation in families segregating a C9orf72 repeat expansion. JAMA Neurol 2017; 74: 445–452. [DOI] [PubMed] [Google Scholar]
  • 45. Devenney E, Hornberger M, Irish M, et al. Frontotemporal dementia associated with the C9ORF72 mutation: a unique clinical profile. JAMA Neurol 2014; 71: 331–339. [DOI] [PubMed] [Google Scholar]
  • 46. Farg MA, Sundaramoorthy V, Sultana JM, et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum Mol Genet 2014; 23: 3579–3595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Rohrer JD, Isaacs AM, Mizielinska S, et al. C9orf72 expansions in frontotemporal dementia and amyotrophic lateral sclerosis. Lancet Neurol 2015; 14: 291–301. [DOI] [PubMed] [Google Scholar]
  • 48. Lee SE, Khazenzon AM, Trujillo AJ, et al. Altered network connectivity in frontotemporal dementia with C9orf72 hexanucleotide repeat expansion. Brain 2014; 137: 3047–3060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Irwin DJ, McMillan CT, Brettschneider J, et al. Cognitive decline and reduced survival in C9orf72 expansion frontotemporal degeneration and amyotrophic lateral sclerosis. J Neurol Neurosurg Psychiatry 2013; 84: 163–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Khan BK, Yokoyama JS, Takada LT, et al. Atypical, slowly progressive behavioural variant frontotemporal dementia associated with C9ORF72 hexanucleotide expansion. J Neurol Neurosurg Psychiatry 2012; 83: 358–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Rohrer JD, Nicholas JM, Cash DM, et al. Presymptomatic cognitive and neuroanatomical changes in genetic frontotemporal dementia in the Genetic Frontotemporal dementia Initiative (GENFI) study: a cross-sectional analysis. Lancet Neurol 2015; 14: 253–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Ferrari R, Hernandez DG, Nalls MA, et al. Frontotemporal dementia and its subtypes: a genome-wide association study. Lancet Neurol 2014; 13: 686–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Piguet O, Brooks WS, Halliday GM, et al. Similar early clinical presentations in familial and non-familial frontotemporal dementia. J Neurol Neurosurg Psychiatry 2004; 75: 1743–1745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Pickering-Brown SM, Rollinson S, Du Plessis D, et al. Frequency and clinical characteristics of progranulin mutation carriers in the Manchester frontotemporal lobar degeneration cohort: comparison with patients with MAPT and no known mutations. Brain 2008; 131: 721–731. [DOI] [PubMed] [Google Scholar]
  • 55. Ng AS. Genetics of frontotemporal dementia in Asia: advancing knowledge through collaboration. Neurology 2015; 85: 2060–2062. [DOI] [PubMed] [Google Scholar]
  • 56. Snowden JS, Adams J, Harris J, et al. Distinct clinical and pathological phenotypes in frontotemporal dementia associated with MAPT, PGRN and C9orf72 mutations. Amyotroph Lateral Scler Frontotemporal Degener 2015; 16: 497–505. [DOI] [PubMed] [Google Scholar]
  • 57. Moreno F, Indakoetxea B, Barandiaran M, et al. “Frontotemporoparietal” dementia: clinical phenotype associated with the c.709–1G>A PGRN mutation. Neurology 2009; 73: 1367–1374. [DOI] [PubMed] [Google Scholar]
  • 58. Gass J, Cannon A, Mackenzie IR, et al. Mutations in progranulin are a major cause of ubiquitin-positive frontotemporal lobar degeneration. Hum Mol Genet 2006; 15: 2988–3001. [DOI] [PubMed] [Google Scholar]
  • 59. Mackenzie IR, Neumann M. Molecular neuropathology of frontotemporal dementia: insights into disease mechanisms from postmortem studies. J Neurochem 2016; 138(Suppl. 1): 54–70. [DOI] [PubMed] [Google Scholar]
  • 60. Woollacott IO, Rohrer JD. The clinical spectrum of sporadic and familial forms of frontotemporal dementia. J Neurochem 2016; 138(Suppl. 1): 6–31. [DOI] [PubMed] [Google Scholar]
  • 61. Patel AN, Sampson JB. Cognitive profile of C9orf72 in frontotemporal dementia and amyotrophic lateral sclerosis. Curr Neurol Neurosci Rep 2015; 15: 59. [DOI] [PubMed] [Google Scholar]
  • 62. Takada LT, Sha SJ. Neuropsychiatric features of C9orf72-associated behavioral variant frontotemporal dementia and frontotemporal dementia with motor neuron disease. Alzheimers Res Ther 2012; 4: 38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Suhonen NM, Haanpaa RM, Korhonen V, et al. Neuropsychological profile in the C9ORF72 associated behavioral variant frontotemporal dementia. J Alzheimers Dis 2017; 58: 479–489. [DOI] [PubMed] [Google Scholar]
  • 64. Rohrer JD, Lashley T, Schott JM, et al. Clinical and neuroanatomical signatures of tissue pathology in frontotemporal lobar degeneration. Brain 2011; 134: 2565–2581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Gitcho MA, Strider J, Carter D, et al. VCP mutations causing frontotemporal lobar degeneration disrupt localization of TDP-43 and induce cell death. J Biol Chem 2009; 284: 12384–12398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Watts GD, Wymer J, Kovach MJ, et al. Inclusion body myopathy associated with Paget disease of bone and frontotemporal dementia is caused by mutant valosin-containing protein. Nat Genet 2004; 36: 377–381. [DOI] [PubMed] [Google Scholar]
  • 67. Neumann M, Mackenzie IR, Cairns NJ, et al. TDP-43 in the ubiquitin pathology of frontotemporal dementia with VCP gene mutations. J Neuropathol Exp Neurol 2007; 66: 152–157. [DOI] [PubMed] [Google Scholar]
  • 68. Forman MS, Mackenzie IR, Cairns NJ, et al. Novel ubiquitin neuropathology in frontotemporal dementia with valosin-containing protein gene mutations. J Neuropathol Exp Neurol 2006; 65: 571–581. [DOI] [PubMed] [Google Scholar]
  • 69. Bersano A, Del Bo R, Lamperti C, et al. Inclusion body myopathy and frontotemporal dementia caused by a novel VCP mutation. Neurobiol Aging 2009; 30: 752–758. [DOI] [PubMed] [Google Scholar]
  • 70. Jacquin A, Rouaud O, Soichot P, et al. Psychiatric presentation of frontotemporal dementia associated with inclusion body myopathy due to the VCP mutation (R155H) in a French family. Case Rep Neurol 2013; 5: 187–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Kazamel M, Sorenson EJ, McEvoy KM, et al. Clinical spectrum of valosin containing protein (VCP)-opathy. Muscle Nerve 2016; 54: 94–99. [DOI] [PubMed] [Google Scholar]
  • 72. Gidaro T, Modoni A, Sabatelli M, et al. An Italian family with inclusion-body myopathy and frontotemporal dementia due to mutation in the VCP gene. Muscle Nerve 2008; 37: 111–114. [DOI] [PubMed] [Google Scholar]
  • 73. Komatsu J, Iwasa K, Yanase D, et al. Inclusion body myopathy with Paget disease of the bone and frontotemporal dementia associated with a novel G156S mutation in the VCP gene. Muscle Nerve. Epub ahead of print 19 July 2013. DOI: 10.1002/mus.23960. [DOI] [PubMed] [Google Scholar]
  • 74. Kimonis VE, Fulchiero E, Vesa J, et al. VCP disease associated with myopathy, Paget disease of bone and frontotemporal dementia: review of a unique disorder. Biochim Biophys Acta 2008; 1782: 744–748. [DOI] [PubMed] [Google Scholar]
  • 75. Shi Z, Hayashi YK, Mitsuhashi S, et al. Characterization of the Asian myopathy patients with VCP mutations. Eur J Neurol 2012; 19: 501–509. [DOI] [PubMed] [Google Scholar]
  • 76. Haubenberger D, Bittner RE, Rauch-Shorny S, et al. Inclusion body myopathy and Paget disease is linked to a novel mutation in the VCP gene. Neurology 2005; 65: 1304–1305. [DOI] [PubMed] [Google Scholar]
  • 77. Kim EJ, Park YE, Kim DS, et al. Inclusion body myopathy with Paget disease of bone and frontotemporal dementia linked to VCP p.Arg155Cys in a Korean family. Arch Neurol 2011; 68: 787–796. [DOI] [PubMed] [Google Scholar]
  • 78. Mehta SG, Khare M, Ramani R, et al. Genotype-phenotype studies of VCP-associated inclusion body myopathy with Paget disease of bone and/or frontotemporal dementia. Clin Genet 2013; 83: 422–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Neumann M, Sampathu DM, Kwong LK, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006; 314: 130–133. [DOI] [PubMed] [Google Scholar]
  • 80. Benajiba L, Le Ber I, Camuzat A, et al. TARDBP mutations in motoneuron disease with frontotemporal lobar degeneration. Ann Neurol 2009; 65: 470–473. [DOI] [PubMed] [Google Scholar]
  • 81. Synofzik M, Born C, Rominger A, et al. Targeted high-throughput sequencing identifies a TARDBP mutation as a cause of early-onset FTD without motor neuron disease. Neurobiol Aging 2014; 35: 1212 e1–e5. [DOI] [PubMed] [Google Scholar]
  • 82. Borroni B, Bonvicini C, Alberici A, et al. Mutation within TARDBP leads to frontotemporal dementia without motor neuron disease. Hum Mutat 2009; 30: E974–E983. [DOI] [PubMed] [Google Scholar]
  • 83. Borroni B, Archetti S, Del Bo R, et al. TARDBP mutations in frontotemporal lobar degeneration: frequency, clinical features, and disease course. Rejuvenation Res 2010; 13: 509–517. [DOI] [PubMed] [Google Scholar]
  • 84. Mutihac R, Alegre-Abarrategui J, Gordon D, et al. TARDBP pathogenic mutations increase cytoplasmic translocation of TDP-43 and cause reduction of endoplasmic reticulum Ca(2)(+) signaling in motor neurons. Neurobiol Dis 2015; 75: 64–77. [DOI] [PubMed] [Google Scholar]
  • 85. Lashley T, Rohrer JD, Mead S, et al. Review: an update on clinical, genetic and pathological aspects of frontotemporal lobar degenerations. Neuropathol Appl Neurobiol 2015; 41: 858–881. [DOI] [PubMed] [Google Scholar]
  • 86. Hock EM, Polymenidou M. Prion-like propagation as a pathogenic principle in frontotemporal dementia. J Neurochem 2016; 138(Suppl. 1): 163–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Bodea LG, Eckert A, Ittner LM, et al. Tau physiology and pathomechanisms in frontotemporal lobar degeneration. J Neurochem 2016; 138(Suppl. 1): 71–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci 2001; 24: 1121–1159. [DOI] [PubMed] [Google Scholar]
  • 89. Josephs KA, Hodges JR, Snowden JS, et al. Neuropathological background of phenotypical variability in frontotemporal dementia. Acta Neuropathol 2011; 122: 137–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Josephs KA, Holton JL, Rossor MN, et al. Frontotemporal lobar degeneration and ubiquitin immunohistochemistry. Neuropathol Appl Neurobiol 2004; 30: 369–373. [DOI] [PubMed] [Google Scholar]
  • 91. Mackenzie IR, Rademakers R, Neumann M. TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol 2010; 9: 995–1007. [DOI] [PubMed] [Google Scholar]
  • 92. Walker AK, Spiller KJ, Ge G, et al. Functional recovery in new mouse models of ALS/FTLD after clearance of pathological cytoplasmic TDP-43. Acta Neuropathol 2015; 130: 643–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Mackenzie IR, Neumann M, Baborie A, et al. A harmonized classification system for FTLD-TDP pathology. Acta Neuropathol 2011; 122: 111–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Neumann M, Rademakers R, Roeber S, et al. A new subtype of frontotemporal lobar degeneration with FUS pathology. Brain 2009; 132: 2922–2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Roeber S, Mackenzie IR, Kretzschmar HA, et al. TDP-43-negative FTLD-U is a significant new clinico-pathological subtype of FTLD. Acta Neuropathol 2008; 116: 147–157. [DOI] [PubMed] [Google Scholar]
  • 96. Mackenzie IR, Foti D, Woulfe J, et al. Atypical frontotemporal lobar degeneration with ubiquitin-positive, TDP-43-negative neuronal inclusions. Brain 2008; 131: 1282–1293. [DOI] [PubMed] [Google Scholar]
  • 97. Holm IE, Isaacs AM, Mackenzie IR. Absence of FUS-immunoreactive pathology in frontotemporal dementia linked to chromosome 3 (FTD-3) caused by mutation in the CHMP2B gene. Acta Neuropathol 2009; 118: 719–720. [DOI] [PubMed] [Google Scholar]
  • 98. Holm IE, Englund E, Mackenzie IR, et al. A reassessment of the neuropathology of frontotemporal dementia linked to chromosome 3. J Neuropathol Exp Neurol 2007; 66: 884–891. [DOI] [PubMed] [Google Scholar]
  • 99. Lindquist SG, Braedgaard H, Svenstrup K, et al. Frontotemporal dementia linked to chromosome 3 (FTD-3)–current concepts and the detection of a previously unknown branch of the Danish FTD-3 family. Eur J Neurol 2008; 15: 667–670. [DOI] [PubMed] [Google Scholar]
  • 100. Skibinski G, Parkinson NJ, Brown JM, et al. Mutations in the endosomal ESCRTIII-complex subunit CHMP2B in frontotemporal dementia. Nat Genet 2005; 37: 806–808. [DOI] [PubMed] [Google Scholar]
  • 101. Urwin H, Authier A, Nielsen JE, et al. Disruption of endocytic trafficking in frontotemporal dementia with CHMP2B mutations. Hum Mol Genet 2010; 19: 2228–2238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Chassefeyre R, Martinez-Hernandez J, Bertaso F, et al. Regulation of postsynaptic function by the dementia-related ESCRT-III subunit CHMP2B. J Neurosci 2015; 35: 3155–3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Cannon A, Baker M, Boeve B, et al. CHMP2B mutations are not a common cause of frontotemporal lobar degeneration. Neurosci Lett 2006; 398: 83–84. [DOI] [PubMed] [Google Scholar]
  • 104. Schumacher A, Friedrich P, Diehl-Schmid J, et al. No association of chromatin-modifying protein 2B with sporadic frontotemporal dementia. Neurobiol Aging 2007; 28: 1789–1790. [DOI] [PubMed] [Google Scholar]
  • 105. Gordon E, Rohrer JD, Fox NC. Advances in neuroimaging in frontotemporal dementia. J Neurochem 2016; 138(Suppl. 1): 193–210. [DOI] [PubMed] [Google Scholar]
  • 106. Day GS, Farb NA, Tang-Wai DF, et al. Salience network resting-state activity: prediction of frontotemporal dementia progression. JAMA Neurol 2013; 70: 1249–1253. [DOI] [PubMed] [Google Scholar]
  • 107. Ranasinghe KG, Rankin KP, Pressman PS, et al. Distinct subtypes of behavioral variant frontotemporal dementia based on patterns of network degeneration. JAMA Neurol 2016; 73: 1078–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Kato T, Inui Y, Nakamura A, et al. Brain fluorodeoxyglucose (FDG) PET in dementia. Ageing Res Rev 2016; 30: 73–84. [DOI] [PubMed] [Google Scholar]
  • 109. Barton C, Ketelle R, Merrilees J, et al. Non-pharmacological management of behavioral symptoms in frontotemporal and other dementias. Curr Neurol Neurosci Rep 2016; 16: 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Shinagawa S. Neuropsychiatric management of young-onset dementias. Psychiatr Clin North Am 2015; 38: 323–331. [DOI] [PubMed] [Google Scholar]
  • 111. Tsai RM, Boxer AL. Therapy and clinical trials in frontotemporal dementia: past, present, and future. J Neurochem 2016; 138(Suppl. 1): 211–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Swartz JR, Miller BL, Lesser IM, et al. Frontotemporal dementia: treatment response to serotonin selective reuptake inhibitors. J Clin Psychiatry 1997; 58: 212–216. [PubMed] [Google Scholar]
  • 113. Moretti R, Torre P, Antonello RM, et al. Frontotemporal dementia: paroxetine as a possible treatment of behavior symptoms. A randomized, controlled, open 14-month study. Eur Neurol 2003; 49: 13–19. [DOI] [PubMed] [Google Scholar]
  • 114. Deakin JB, Rahman S, Nestor PJ, et al. Paroxetine does not improve symptoms and impairs cognition in frontotemporal dementia: a double-blind randomized controlled trial. Psychopharmacology (Berl) 2004; 172: 400–408. [DOI] [PubMed] [Google Scholar]
  • 115. Herrmann N, Black SE, Chow T, et al. Serotonergic function and treatment of behavioral and psychological symptoms of frontotemporal dementia. Am J Geriatr Psychiatry 2012; 20: 789–797. [DOI] [PubMed] [Google Scholar]
  • 116. Hughes LE, Rittman T, Regenthal R, et al. Improving response inhibition systems in frontotemporal dementia with citalopram. Brain 2015; 138: 1961–1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Mendez MF, Shapira JS, Miller BL. Stereotypical movements and frontotemporal dementia. Mov Disord 2005; 20: 742–745. [DOI] [PubMed] [Google Scholar]
  • 118. Anneser JM, Jox RJ, Borasio GD. Inappropriate sexual behaviour in a case of ALS and FTD: successful treatment with sertraline. Amyotroph Lateral Scler 2007; 8: 189–190. [DOI] [PubMed] [Google Scholar]
  • 119. Lebert F, Stekke W, Hasenbroekx C, et al. Frontotemporal dementia: a randomised, controlled trial with trazodone. Dement Geriatr Cogn Disord 2004; 17: 355–359. [DOI] [PubMed] [Google Scholar]
  • 120. Ikeda M, Shigenobu K, Fukuhara R, et al. Efficacy of fluvoxamine as a treatment for behavioral symptoms in frontotemporal lobar degeneration patients. Dement Geriatr Cogn Disord 2004; 17: 117–121. [DOI] [PubMed] [Google Scholar]
  • 121. Mega MS, Cummings JL. Frontal-subcortical circuits and neuropsychiatric disorders. J Neuropsychiatry Clin Neurosci 1994; 6: 358–370. [DOI] [PubMed] [Google Scholar]
  • 122. Rahman S, Sahakian BJ, Hodges JR, et al. Specific cognitive deficits in mild frontal variant frontotemporal dementia. Brain 1999; 122: 1469–1493. [DOI] [PubMed] [Google Scholar]
  • 123. Rahman S, Robbins TW, Hodges JR, et al. Methylphenidate (‘Ritalin’) can ameliorate abnormal risk-taking behavior in the frontal variant of frontotemporal dementia. Neuropsychopharmacology 2006; 31: 651–658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Huey ED, Garcia C, Wassermann EM, et al. Stimulant treatment of frontotemporal dementia in 8 patients. J Clin Psychiatry 2008; 69: 1981–1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Mocellin R, Scholes A, Walterfang M, et al. Clinical update on frontotemporal dementia: diagnosis and treatment. Australas Psychiatry 2015; 23: 481–487. [DOI] [PubMed] [Google Scholar]
  • 126. Vercelletto M, Boutoleau-Bretonniere C, Volteau C, et al. Memantine in behavioral variant frontotemporal dementia: negative results. J Alzheimers Dis 2011; 23: 749–759. [DOI] [PubMed] [Google Scholar]
  • 127. Chow TW. Treatment approaches to symptoms associated with frontotemporal degeneration. Curr Psychiatry Rep 2005; 7: 376–380. [DOI] [PubMed] [Google Scholar]
  • 128. Kertesz A, Morlog D, Light M, et al. Galantamine in frontotemporal dementia and primary progressive aphasia. Dement Geriatr Cogn Disord 2008; 25: 178–185. [DOI] [PubMed] [Google Scholar]
  • 129. McCarter SJ, St Louis EK, Boeve BF. Sleep disturbances in frontotemporal dementia. Curr Neurol Neurosci Rep 2016; 16: 85. [DOI] [PubMed] [Google Scholar]
  • 130. Manoochehri M, Huey ED. Diagnosis and management of behavioral issues in frontotemporal dementia. Curr Neurol Neurosci Rep 2012; 12: 528–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Riedl L, Mackenzie IR, Forstl H, et al. Frontotemporal lobar degeneration: current perspectives. Neuropsychiatr Dis Treat 2014; 10: 297–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Finger EC, MacKinley J, Blair M, et al. Oxytocin for frontotemporal dementia: a randomized dose-finding study of safety and tolerability. Neurology 2015; 84: 174–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Callegari I, Mattei C, Benassi F, et al. Agomelatine improves apathy in frontotemporal dementia. Neurodegener Dis 2016; 16: 352–356. [DOI] [PubMed] [Google Scholar]
  • 134. Golde TE, Lewis J, McFarland NR. Anti-tau antibodies: hitting the target. Neuron 2013; 80: 254–256. [DOI] [PubMed] [Google Scholar]
  • 135. Li YQ, Tan MS, Yu JT, et al. Frontotemporal lobar degeneration: mechanisms and therapeutic strategies. Mol Neurobiol 2016; 53: 6091–6105. [DOI] [PubMed] [Google Scholar]
  • 136. Jiang J, Cleveland DW. Bidirectional transcriptional inhibition as therapy for ALS/FTD caused by repeat expansion in C9orf72. Neuron 2016; 92: 1160–1163. [DOI] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Psychopharmacology are provided here courtesy of SAGE Publications

RESOURCES