The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Published Online:

F rontotemporal lobar degeneration presents as one of three major clinical syndromes 1 : frontal-variant frontotemporal dementia (fvFTD), 2 semantic dementia (sometimes referred to as primary progressive fluent aphasia or temporal-variant FTD), and progressive nonfluent aphasia. Despite frontotemporal lobar degeneration being the cause of 12.5–16.5% of all degenerative-type dementias, 3 there is a shortage of research to elucidate its underlying mechanisms, and there are few studies that have evaluated the effectiveness of pharmacological treatments to improve the symptoms of frontotemporal lobar degeneration. There is currently no standard pharmacological intervention for the treatment of frontotemporal lobar degeneration and no known treatment to delay its progression; however, there are various prescribed medications that are commonly used to help alleviate some symptoms.

Due to the heterogeneity in the clinical presentation of the syndromes comprising frontotemporal lobar degeneration, this article will focus on fvFTD. We will first characterize the clinical presentation of fvFTD, followed by an overview of the underlying neuropathology. We shall then review the research literature on the pharmacological agents that have been studied to ameliorate the behavioral and cognitive changes associated with FTD, as well as explore future directions.

Clinical Presentation

Each of the major syndromes constituting frontotemporal lobar degeneration has its own common clinical presentation. 4 Although semantic dementia and progressive nonfluent aphasia present with prominent language abnormalities at the onset of the disease course, fvFTD initially presents with a personality change and behavioral abnormalities. The behavioral features, which are seen early in fvFTD, may include the loss of personal and social awareness, disinhibition, poor insight, impulsivity, stereotypies, and hyperorality. Furthermore, the inability to recognize emotion—in particular anger and disgust—the inability to empathize with others, and the impaired moral reasoning that are observed in fvFTD patients closely resemble the profile of persons with antisocial personality disorder. 5 These patients may exhibit psychosis, 3 increased risk-taking behavior, 6 compulsions, 712 and apathy. 13 Although the behavioral abnormalities worsen with disease progression, many of the behavioral deficits lessen with increased apathy and withdrawal. Moreover, as the disease progresses, these patients are at risk for developing changes to the motor system, such as motor neuron disease, parkinsonism, and dysphagia, although motor neuron disease can precede the manifestations of fvFTD. 14

In addition to behavioral abnormalities, fvFTD is associated with cognitive deficits. For instance, patients may display great impairment in executive function 15 , 16 and at varying points in the disease course may 15 , 17 , 18 or may not 19 present with memory impairments. Furthermore, prominent attentional deficits may be present. 15 , 16

Importantly, the symptoms of fvFTD are more related to the affected areas of the brain than to specific neuropathological factors. 4 Consequently, the degree of frontal versus temporal compromise can account for much of the variability in the clinical presentations of the disorder. 20 In addition, patients with greater right hemispheric compromise show more pronounced impairment in social cognition. Moreover, although patients may initially present with features that are consistent with one particular frontotemporal lobar degeneration subtype, as the disease progresses, their clinical profile may be more consistent with a different subtype. 21 This is not to say that the profile switches from one subtype to another; rather, the new profile is an add-on to the initial one. However, the new profile may still be more prominent than the initial one. For example, a patient may initially be diagnosed with fvFTD but may later present with semantic dementia as his or her condition progresses. Therefore, the clinical presentation of fvFTD is heterogeneous within a population of patients and may vary within a particular patient.

Neurotransmitter and Receptor Alterations

Research evidence suggests that there are significant neurotransmitter system abnormalities in fvFTD, and the cholinergic, serotonergic, dopaminergic, noradrenergic, and glutamatergic systems are thought to be implicated. The following discussion will provide a general overview of the restricted findings of the involvement of neurotransmitter system abnormalities in fvFTD. However, although the scope of the research is moderate at present, significant venues for pharmacological intervention are implied.

Cholinergic System

Given the significance of an impaired cholinergic system in Alzheimer’s disease, the most common form of dementia, it is understandable why investigators explored this system in FTD patients. The literature is mixed in terms of the integrity of the cholinergic system in FTD. Most studies show normal or higher levels of cortical choline acetyltransferase, 2224 as well as normal levels of acetylcholinesterase. 25 One study reports that postsynaptic muscarinic receptor binding is intact in the brains of FTD patients, 23 yet others report a decrease in binding. 26 , 27 Lastly, studies have demonstrated that the neurons in the nucleus basalis of Meynert are preserved, 22 , 28 except for one study that found a decrease in the density of these neurons at autopsy. 29

Serotonergic System

Investigators have often found a dysfunctional serotonergic system in FTD patients. This is further supported by the clinical presentation of this illness, which closely corresponds to serotonergic dysfunction. 4 Researchers have identified a significant reduction in postsynaptic 5-HT1 and 5-HT2 serotonin receptors, particularly in the frontal cortex, including orbitofrontal and medial frontal regions, as well as in the temporal, cingulate, and hypothalamic regions. 23 , 24 , 30 Others have observed presynaptic serotonergic deficits 24 , 31 and neuronal loss by 40% in the raphe nuclei in FTD. 32 This evidence of serotonergic dysfunction in FTD lends support for therapeutic trials with medications that act to increase serotonergic tone in the brain.

Dopaminergic System

There is some evidence supporting a dysfunctional dopaminergic system in FTD. This includes observations of reduced D2 ligand uptake in the frontal cortex 33 and fewer presynaptic dopamine transporters in the striatum, 34 and in the syndrome frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), there are also reduced presynaptic striatal dopaminergic nerve terminals. 35 However, other studies did not find evidence of dopaminergic dysfunction in FTD. 36 , 37

Noradrenergic System

Similarly, there is conflicting evidence for a dysfunctional noradrenergic system in FTD. For instance, one group found that the density of neurons in the locus coeruleus is preserved, 32 whereas another group found that it is decreased. 38 Further research is required to ascertain the involvement of the noradrenergic system in FTD.

Glutamatergic System

Finally, the glutamatergic system may play a role in FTD. There are indications that the frontal and temporal cortices of FTD patients have reduced levels of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) and N- methyl- D -aspartate (NMDA) receptors, 23 as well as reduced numbers of glutamatergic pyramidal cells. 39 Further examination of the role of the glutamatergic system in FTD should be undertaken.

Symptomatic Treatments of fvFTD

There is a dearth of clinical trials testing the efficacy of medications to treat the symptoms of FTD. The clinical trials that have been conducted include evaluations of cholinesterase inhibitors, antidepressants, an NMDA receptor antagonist, antipsychotics, and a stimulant in the treatment of FTD symptoms.

Cholinesterase Inhibitors

Cholinesterase inhibitors, with their relative efficacy in treating Alzheimer’s disease, were an intuitive pharmacologic intervention to undergo evaluation in the treatment of fvFTD. However, cholinesterase inhibitors do not consistently improve cognitive and behavioral symptoms of this disease. According to a review, such treatment is associated with increased aggressiveness and agitation. 31 Similarly, donepezil treatment was associated with an exacerbation of disinhibition and compulsions. 40

Some studies suggest positive effects from treatment with cholinesterase inhibitors while others do not. An open-label study compared a group treated with rivastigmine, an inhibitor of both acetylcholinesterase and butyrylcholinesterase, with comparison groups receiving an antipsychotic, a benzodiazepine, or selegiline. Rivastigmine treatment was associated with less behavioral impairment and less caregiver burden, though it did not improve cognition. 41 However, another study consisted of an open-label phase followed by a double-blind, randomized placebo-controlled phase comparing the effects of galantamine in patients with FTD versus those with progressive nonfluent aphasia and semantic dementia. 42 Galantamine improved word production in the progressive nonfluent aphasia and semantic dementia group, but it was ineffective at improving scores on the Clinical Global Impression Severity Scale in the FTD group. Furthermore, the Frontal Behavioral Inventory (the FBI) displayed a worsening , albeit nonsignificant, effect in the treatment group compared to the stable placebo group. Therefore, due to the strength in the design of this study, it may be suggested that galantamine is ineffective in treating FTD, although the strength of this conclusion is limited by the smaller size of this study (36 and 34 participants, respectively, in the open-label and placebo-controlled phases).

In a randomized comparison trial, nine FTD patients were treated immediately after diagnosis with either donepezil or rivastigmine. 43 All four men in the study showed clinically significant cognitive improvement, as evaluated by the Mini-Mental State Examination (MMSE) and the Clock Drawing Test, and three of the four men showed positive changes in their single photon emission computed tomography (SPECT) scans after 6 months. Three of the five women only improved slightly after 3 months, and two of the five women continued to deteriorate. It appears from this study that donepezil and rivastigmine may be effective in treating cognitive impairments in FTD, particularly in afflicted men, but it is worth noting that only nine participants were tested and that the males were younger than the females (64.2±2.2 years old versus 71.6±2.9 years old, respectively). This potential age effect may underlie the apparent gender difference.

Antidepressants

The medications most commonly studied to treat FTD are the class of antidepressants known as selective serotonin reuptake inhibitors (SSRIs). SSRIs such as sertraline, fluvoxamine, fluoxetine, and paroxetine are commonly used as the first line of treatment for the management of disinhibition, stereotypies, sexually inappropriate behaviors, and dietary changes (i.e., food cravings) in fvFTD. 4446 Unfortunately, most evaluations of SSRIs in the treatment of fvFTD have used open-label or case series designs, with only a few randomized, placebo-controlled trials. 14 The latter have produced mixed results for the efficacy of SSRIs in the treatment of fvFTD.

In an open-label study, sertraline appeared to decrease the stereotypical movements in a group of 18 FTD patients. 47 Similarly, fluvoxamine appeared to reduce stereotypies and compulsive complaints of pain in two cases of FTD. 48

In an uncontrolled study, 46 11 FTD patients were randomly assigned to receive fluoxetine, sertraline, or paroxetine for 3 months. The patients showed improved behavioral symptoms, including disinhibition, depressive symptoms, carbohydrate craving, and compulsions; however, cognition did not improve.

A meta-analysis 49 examined the combined effects of four different antidepressants (trazodone, selegiline, fluvoxamine, and paroxetine). The results show a considerable combined mean reduction (improvement) of 15.4 points on the Neuropsychiatric Inventory (NPI), a caregiver interview that assesses recent changes in particular behaviors. It should be noted that the authors of this study admit that the effect of these medications may be an overestimation as these studies had small sample sizes, only two of the six studies were placebo-controlled, and there may have been a publication bias toward positive trials. Additionally, the fluvoxamine trial included in the meta-analysis 50 tested 10 patients with fvFTD as well as five patients with semantic dementia. Because these two subgroups were not separated in the analysis, we cannot conclude how well the fvFTD subgroup truly responded to the treatment, yet the cohort as a whole, relative to baseline, showed significant improvement on total NPI scores and on each of the subscales of the Stereotypy Rating Inventory.

It should also be noted that the two studies evaluating paroxetine 45 , 51 found opposing results in terms of its efficacy. Both are randomized, controlled studies, yet they differ greatly in their methodologies, namely in study length, treatment dosage and inclusion criteria. Specifically, the investigators in the randomized, double-blind, placebo-controlled study 51 treated FTD and semantic dementia patients for 9 weeks and found that relative to placebo, paroxetine, 40 mg/day, failed to improve behavioral symptoms and impaired cognition. However, the investigators in the other study, 46 which used an open-label design, treated both FTD and semantic dementia patients with paroxetine, 20 mg/day (treatment), or piracetam, 1,200 mg/day (comparison), for 14 months. They found a significant behavioral improvement with no effect on cognition in the treatment group relative to the comparison group. Because these two studies were small, testing 10 and 16 patients, respectively, and due to the methodological differences described above, we cannot accurately compare the results of these studies, although the studies do suggest that paroxetine may only be efficacious in the treatment of FTD when given over a longer period of time. Trazodone, a low affinity serotonin reuptake inhibitor that increases extracellular levels of 5-HT in frontal cortex, also has a high affinity for 5-HT2 A receptors and acts as an antagonist at alpha-1 and alpha-2 adrenergic receptors. Considering the aforementioned neurotransmitter aberrations in FTD, trazodone may be a useful treatment option. A randomized, double-blind, placebo-controlled cross-over trial 52 and an open-label trial 53 both had similar findings: trazodone is well-tolerated and can significantly improve behavioral disturbances as measured by the NPI but does not improve cognition as measured by the MMSE.

Researchers have also evaluated treating fvFTD with monoamine oxidase inhibitors (MAOIs), a class of antidepressants with a different pharmacodynamic action than SSRIs. Unlike SSRIs, MAOIs increase the availability of norepinephrine, serotonin, and dopamine, which is of particular interest because there is evidence supporting the disruption of each of these three neurotransmitter systems in FTD (refer to previous section). Furthermore, MAO-A and MAO-B, enzymes targeted by MAOIs, show regional specificity to neuroanatomical areas that are associated with fvFTD. For example, MAO-A and MAO-B displayed significantly increased activity in the hypothalamus and decreased activity in the nucleus basalis of Meynert, while MAO-A also displayed decreased activity in the temporal pole. 54 Moclobemide, a well-tolerated MAO-A inhibitor, appeared to improve behavior, affect, and speech in six FTD patients after a 4-week treatment period. 55 Additionally, selegiline, a well-tolerated MAO-B inhibitor, may be useful in the treatment of FTD as it may have neuroprotective effects by reducing free radical production and by saving dying neurons. 56 , 57 A 3-month treatment with selegiline was associated with significantly improved neuropsychiatric symptoms as well as attention in three FTD patients. 58 Despite the positive results obtained in these case report studies, MAOIs need to be evaluated in larger randomized, double-blind, placebo-controlled studies in order to ascertain their efficacy in treating FTD.

Memantine

At present, only a few studies have examined the effects of memantine, an NMDA-receptor antagonist. In a case series report, 59 three FTD patients (ages 52, 57, and 64 years old) were treated for 3 months with the therapeutic dose of 20 mg/day of memantine. All three showed significant improvement in total NPI score, specifically in the subscales of apathy, agitation, and anxiety, and all showed good tolerance for memantine. However, a case study found that a 73-year-old man’s clinical state continually deteriorated while his treatment with sertraline was augmented with memantine for a period of 6 months. 60 The latter case study not only used memantine as an adjunct treatment but also used a lower dose of the medication at 10 mg/day. Thus, there may have been a dose effect as well as a potential interaction between the two medications.

Recently, a 26-week Phase IV nonrandomized, uncontrolled, open-label trial evaluating the efficacy of memantine in treating frontotemporal lobar degeneration was conducted on 21 patients with FTD, 13 with semantic dementia, and nine with progressive nonfluent aphasia. 61 The target dose of 20 mg daily was well-tolerated by most patients. Treatment was associated with a transient improvement in total NPI scores primarily in the FTD group, though variable declines were observed on numerous cognitive and behavioral measures, specifically, the Alzheimer’s Disease Assessment Scale-Cognitive (ADAS-cog), Executive Interview (EXIT25), the FBI, the NPI, the Texas Functional Living Scale, and the Unified Parkinson's Disease Rating Scale-motor scale (UPDRS). The FTD and semantic dementia groups declined on most of these measures, but remained stable on the UPDRS, whereas the progressive nonfluent aphasia group remained relatively stable on the ADAS-cog, NPI, and Texas Functional Living Scale, but declined on the UPDRS. The researchers suggest that future placebo-controlled trials of memantine in frontotemporal lobar degeneration are warranted and may have greater power to detect behavioral and cognitive effects if focused on FTD and semantic dementia. It is clear from these reports that more research must be conducted to ascertain whether memantine is an effective treatment in frontotemporal lobar degeneration syndromes.

Antipsychotics

Numerous studies have investigated the efficacy of typical and atypical antipsychotics for the treatment of FTD. Considering the impairment observed in the dopaminergic and serotonergic systems in FTD patients, aripiprazole, an atypical antipsychotic that has dopaminergic and 5-HT 1A partial agonistic effects, is a theoretically viable option for treatment, and clinicians evaluated it in a case study examining the 73-year-old man discussed in the memantine section. 60 Under consecutive treatment with this medication, the patient’s clinical symptoms significantly improved. The patient showed improvement in cognition, as measured by the MMSE, and a marked clinical improvement in the negative symptom score of the Positive and Negative Syndrome Scale. Additionally, prior to aripiprazole administration the patient had a decrease in frontal glucose metabolism, which showed partial normalization after only 5 weeks of treatment. This case study provides very limited support for the use of aripiprazole to improve cognition and negative symptoms in FTD patients. Aripiprazole must be more rigorously tested before deeming it an appropriate treatment option.

In another case study, the treating physicians monitored the effects of risperidone on a 42-year-old woman thought to have a “frontal dementia of a non-Alzheimer’s type, possibly Pick’s disease.” 62 After 1 week her psychotic features began to be allayed, and after 3 to 4 months she was free from delusions and hallucinations, she had increased insight and motivation, and her declining cognition had stabilized. However, she became depressed and was treated with paroxetine. She also began to display extrapyramidal symptoms of akathisia and parkinsonism, requiring treatment with clonazepam.

Additional potential support for the use of antipsychotics to treat FTD is observed in a study that reports reduction in agitation in all of the three patients treated with quetiapine, and in one of the three patients treated with risperidone. 13 However, this study combined patients with FTD, semantic dementia and progressive nonfluent aphasia, and we do not know how many people in these two groups were FTD patients, if any.

Investigators examined the effects of olanzapine on the neuropsychiatric manifestations of FTD in a group of 17 patients with some success. 63 This 24-month follow-up study found that olanzapine was well-tolerated and significantly improved scores on all of the behavioral and mood outcomes measured: Clinical Insight Rating Scale, NPI, Behavioral Pathology in Alzheimer’s Disease Rating Scale, and the Cornell Scale for Depression in Dementia. Although cognitive test scores did not significantly differ from baseline, they displayed a worsening trend. This is a noteworthy result because it is plausible that the observed worsening trend could reach significance if a future study were to employ a larger sample size. Therefore, a medication such as olanzapine that improves behavior but potentially worsens or fails to stabilize cognition would not be an ideal FTD treatment option.

Furthermore, symptoms such as delusions, euphoric symptoms, and disinhibited outbursts, which are commonly refractory to treatment with SSRIs, usually can be controlled with the use of atypical antipsychotics. 14 However, great caution must be taken when prescribing antipsychotics to FTD patients. Unfortunately, both typical and atypical antipsychotics may cause extrapyramidal side effects 62 , 64 , 65 and FTD patients may be particularly susceptible to these effects. 64 In addition, treating behavioral symptoms in elderly dementia patients with typical and atypical antipsychotics may increase mortality compared with placebo. 64 , 65

Methylphenidate

Frontotemporal dementia patients may display an increased tendency toward compulsive and risk-taking behaviors, and in order to reduce these behaviors, a treatment should target the underlying neural substrates. Methylphenidate may be an effective treatment because deficits in the dopaminergic and noradrenergic systems exist in FTD, and methylphenidate likely affects risk-taking behavior observed in FTD through its effects on the orbitofrontal cortex and the striatum and the connections between these two regions. The dopamine transporters that methylphenidate is believed to target are primarily found in the striatum. The purpose of the dopaminergic projections from the midbrain to the striatum is to signal reward-related information. This circuit is likely hyperactive in FTD and, coupled with the hypoactivity found in the orbitofrontal cortex, leads to the expression of reward-dependent behavior. The increases in orbitofrontal cortex metabolism observed due to methylphenidate administration may ultimately modulate the activity of the dopaminergic projections from the midbrain to the striatum. 66 Moreover, disrupted circuitry in the orbitofrontal cortex correlates with increased disinhibition, 67 which may be ameliorated with the increase in orbitofrontal cortex metabolism that occurs from treatment with methylphenidate.

One group of investigators 66 administered methylphenidate to eight FTD patients. Despite not finding any significant changes in cognition, notably memory and executive function, as well as not finding significant improvements in mood, the medication significantly decreased risk-taking behavior. Similarly, one case study reports that methylphenidate was associated with returning the patient’s personality to near-normal levels. 68

Pathological Indicators of FTD and Disease-Modifying Interventions

To date, medications used to treat FTD do not delay the progression of the disease. However, several neuropathological features of the various forms of FTD are identified and implicate possible targets for disease-modifying therapies. These features include ubiquitin inclusions, progranulin mutations, and tau mutations.

FTD with ubiquinated inclusions (FTD-U, also referred to as FTLD-U), including FTD with motor neuron disease, constitutes the most common neuropathologies in frontotemporal lobar degeneration. Ubiquinated inclusions consist primarily of a protein called TAR DNA-binding protein 43 kDA (TDP-43). 69 Less common are tauopathies, such as Pick’s disease, corticobasal degeneration, and progressive supranuclear palsy. 21

Frontotemporal dementia with motor neuron disease is highly heritable 70 and appears to be associated with an unidentified gene on chromosome 9p. 71 , 72 Therefore, this gene and its protein product(s) are potential disease-modifying targets, as are the genetic and biochemical pathways involving TDP-43.

Another pathological correlate of FTD that occurs in most cases without motor neuron disease is the result of genetic mutations that cause a loss of function in the progranulin (PRGN) gene, 73 , 74 where progranulin is a pleiotropic protein involved in mediating neuronal development and inflammation. 75 Postmortem, these patients have FTD with ubiquinated inclusions/TDP-43 pathology, which implicates the potential efficacy of PRGN-altering therapies. Such treatments may act by normalizing PRGN gene expression, by enhancing progranulin posttranslational modification, or by introducing exogenous progranulin into the brains of FTD patients with PRGN mutations. These therapies may even be useful for patients without such mutations. 14

Lastly, according to a review paper on FTD, 76 the most common histological feature of the disease, in cases that meet the strict clinical criteria for fvFTD, 1 is tauopathy, where the microtubule-associated protein tau aggregates in a hyperphosphorylated form. Mutant forms of tau have been found to exist in 30%–42% of FTD cases at autopsy. 7779 These aggregates disrupt normal cellular function and lead to cell death and loss of synapses. The altered levels of tau and its deposition may result from alterations in splicing, tau expression levels, and posttranslational processing. 80

To study the mechanisms of neurodegeneration in diseases with abnormal changes in tau, a group of investigators generated several cell models of tauopathy. 81 They created neuroblastoma cell lines that could inducibly express different variants of the repeat domain of tau (tau RD ), including a repeat domain with the deletion mutation ΔK280. This mutation is known to exist in FTD and is very prone to spontaneously aggregate. The researchers confirmed that the tau RD is toxic to cells and observed that fragmentation of the tau RD precedes aggregation, suggesting that fragmentation, rather than phosphorylation, is important for aggregation. Fragmentation can be inhibited by treating the cells with various inhibitors, thereby preventing aggregation. Importantly, preventing tau aggregation, even without altering its expression, can reduce toxicity to cells.

The findings from this study 81 have important clinical implications, despite its limitations. The results suggest the viability of incorporating inhibitors in pharmacological agents to prevent the toxic aggregation of tau in FTD. However, before attempting to apply these results clinically, it is important to remember that the tauopathy that underlies fvFTD is heterogeneous, which may limit the utility of disease-modifying treatments directed at tau. 14 Additionally, this study was conducted on cells in vitro , therefore, these results may not necessarily be observed in vivo . Of course, one must also take into consideration obstacles such as the blood-brain barrier and selectivity and specificity for the target when designing a disease-modifying agent. Medications like the anticancer drug paclitaxel, which is designed to stabilize microtubules, are currently being evaluated. 82 A randomized, double-blind, placebo-controlled trial evaluated methylthioninium chloride, a tau aggregation inhibitor, and found that it stabilized the progression of Alzheimer’s disease in mild and moderate Alzheimer’s disease patients. 83 Perhaps FTD patients would benefit from methylthioninium chloride treatment as well. Other inhibitors and microtubule tau stabilizers are showing positive results in in vitro and animal studies. 8486

Conclusions, Limitations, and Future Directions

Frontotemporal dementia is a relatively common cause of dementia and accounts for most of the dementia cases in late middle-aged adults. At the time of disease onset, many people are in the middle of their working years, 87 and the rapid decline in their functioning puts great burden on both their families and the health care system, as many people with FTD are institutionalized 1 year following the diagnosis. 88 Despite the relative lack of cognitive treatments for FTD, there are several options for treating some of the behavioral manifestations of the disease. Given the evidence, SSRIs and trazodone appear to be the best first options for treatment. Nevertheless, more research must be completed to ensure that those with FTD are diagnosed and treated early in their disease course in the hopes of delaying the progression of the disease and to delay institutionalization.

An essential element for the early detection of FTD is the use of standardized assessment tools, as there are currently none specific to this disease. One study demonstrated that early in the course of the disease, the brains of FTD patients contain lesions in the orbitofrontal cortex, superior medial frontal cortex, and the hippocampus. 89 Dysfunctions in the orbitofrontal cortex and in the medial frontal cortex underlie social cognitive deficits, 90 , 91 yet most neuropsychological tests are primarily sensitive to dorsolateral frontal dysfunction and have low sensitivity to early FTD. This illustrates the need for behavioral and neuropsychological measures that are sensitive to lesions in the orbitofrontal and medial frontal cortices, as well as tests of social cognition such as measures of Theory of Mind. 92

Another factor involved in the early detection of FTD is the initial difficulty clinicians may have in distinguishing it from other forms of dementia. Fortunately, various forms of dementia may be reliably differentiated from one another when various behavioral rating scales are employed, such as the Cambridge Behavioral Inventory 93 and the FBI. 94 , 95 There are mixed results regarding the utility of the NPI in discriminating various forms of dementia. 94 , 95 Additionally, behavioral measures have been demonstrated to have superior sensitivity over cognitive measures in diagnosing FTD. 96

Furthermore, early diagnosis may be possible with the use of neuroimaging technology. For example, positron emission tomography (PET) research illustrates the ubiquitous dysfunction in the ventromedial frontal cortex of FTD patients, 97 a region highly implicated in the clinical presentation of FTD.

It is interesting to note that while patients with frontotemporal lobar degeneration are diagnosed based on clinical criteria that suggest which region of the brain is most affected, neuroimaging data may suggest primary dysfunction in a different region. This occurs due to lack of an exclusive relation between the syndrome and the distribution of pathological change, which is generally measured as atrophy. 98 For example, patients with semantic dementia invariably have temporal lobe atrophy, but such atrophy does not guarantee neuropsychological findings of language dysfunction. Similarly, patients with fvFTD invariably possess frontal-lobe atrophy and temporal-lobe atrophy, yet the atrophy in the temporal lobe may be greater than in the frontal lobe, and the patients may not display obvious semantic impairment. Therefore, these examples demonstrate the importance of recognizing that despite the usefulness of neuroimaging as a diagnostic aid, it could lead to an incorrect clinical diagnosis if the results were given undue weight in differential diagnosis.

An issue that has created inconsistencies in the FTD literature is the operational definition as to which patients classify as FTD. This varies from study to study. Some studies use the term FTD as the broad superordinate term to encompass the three major syndromes (FTD, semantic dementia, and progressive nonfluent aphasia), while others use the term frontotemporal lobar degeneration. Although the authors clearly define their classification system, it is not always mentioned how many patients comprise each subtype. This makes it difficult, for example, to ascertain the potential use of a particular treatment for a patient with a particular frontotemporal lobar degeneration syndrome.

Researchers choose from a plethora of outcome measures to determine whether a particular intervention is successful. This creates the challenge of accurately comparing various studies’ results to one another. Some of the measures used in FTD treatment studies are the MMSE, the NPI, the FBI, and the FTD Inventory (see Freedman 99 for a review). Interestingly, the studies discussed throughout this article rarely observed any changes in cognition and have made the argument that the cognitive impairment may be refractory to treatment. However, noting that the MMSE, a measure commonly utilized to evaluate cognitive change in FTD studies, is not sensitive to the prominent executive impairment found in FTD patients, any changes in cognition that may have occurred as a result of treatment may be undetected.

Another issue is that there is variability in what constitutes a significant change or improvement. For instance, a statistically significant improvement has been defined as being a total score reduction of 50% on the NPI, 52 whereas elsewhere, improvement on the NPI has been defined as posttreatment scores being significantly different from baseline. 41 Clearly, in order to properly compare the results of individual studies and, in turn, accurately evaluate therapeutic effectiveness, investigators should be consistent in their choices of outcome measures and should standardize what constitutes a statistically significant and clinically relevant improvement on any given measure.

Finally, given the considerable prevalence of FTD, and the relative paucity of pharmacological trials for it, funding agencies should give high priority to financing research aimed at improving early FTD detection and at evaluating the most appropriate forms of treatment. Future studies need to have rigorous experimental designs, include a sufficient sample size, and employ an intent-to-treat analysis to account for participants who fail to complete the protocol. This would strengthen the conclusions from these studies, thereby improving the ability of physicians and other caregivers to provide optimal care for patients living with FTD.

Received April 17, 2009; accepted May 26, 2009. Mr. Kaye and Dr. Verhoeff are affiliated with Kunin-Lunenfeld Applied Research Unit at Baycrest, Toronto, Ontario; Mr. Kaye is also affiliated with the Institute of Medical Science at the University of Toronto; Mrs. Petrovic-Poljak and Drs. Verhoeff and Freedman are affiliated with the Sam and Ida Ross Memory Clinic, Brain Health Centre Clinics, at Baycrest; Mrs. Petrovic-Poljak is also affiliated with the Mood and Related Disorders Clinic, Brain Health Centre Clinics, at Baycrest; Dr. Verhoeff is also affiliated with the Department of Psychiatry, Division of Geriatric Psychiatry, at the University of Toronto; Dr. Freedman is also affiliated with the Division of Neurology and Rotman Research Institute, at Baycrest, and with the Departments of Medicine, Division of Neurology, at Mount Sinai Hospital, at the University Health Network, and at the University of Toronto. Address correspondence to Mr. Edward Kaye, Baycrest, Kunin-Lunenfeld Applied Research Unit, Brain Health Complex, 7th Floor, Room 726, 3560 Bathurst Street, Toronto, Ontario M6A2E1, Canada; [email protected] (e-mail).

Copyright © 2010 American Psychiatric Publishing, Inc.

References

1. Neary D, Snowden JS, Gustafson L, et al: Frontotemporal lobar degeneration: a consensus on clinical diagnostic criteria. Neurology 1998; 51:1546–1554Google Scholar

2. Hodges JR, Miller B: The classification, genetics, and neuropathology of frontotemporal dementia introduction to the special topic papers, part I. Neurocase 2001; 7:31–35Google Scholar

3. Gustafson L: Frontal lobe degeneration of non-Alzheimer type II: clinical picture and differential diagnosis. Arch Gerontol Geriatr 1987; 6:209–223Google Scholar

4. Weder ND, Aziz R, Wilkins K, et al: Frontotemporal dementias: a review. Ann Gen Psychiatry 2007; 6:15Google Scholar

5. Lough S, Kipps CM, Treise C, et al: Social reasoning, emotion, and empathy in frontotemporal dementia. Neuropsychologia 2006; 44:950–958Google Scholar

6. Rahman S, Sahakian BJ, Hodges JR, et al: Specific cognitive deficits in early frontal variant frontotemporal dementia. Brain 1999; 122:1469–1493Google Scholar

7. Ames D, Cumming JL, Wirshing WC, et al: Repetitive and compulsive behavior in frontal lobe degenerations. J Neuropsychiatry Clin Neurosci 1994; 6:100–113Google Scholar

8. Chow TW, Mendez MF: Goals in symptomatic pharmacologic management of frontotemporal lobar degeneration. Am J Alzheimers Dis Other Demen 2002; 17:267–272Google Scholar

9. Gregory CA, Hodges JR: Clinical features of frontal lobe dementia in comparison to Alzheimer’s disease. J Neural Transm 1996; 47:103–123Google Scholar

10. Mendez MF, Perryman KM, Miller BL, et al: Compulsive behaviors as presenting symptoms of frontotemporal dementia. J Geriatr Psychiatry Neurol 1997; 10:154–157Google Scholar

11. Miller BL, Darby AL, Swartz JR, et al: Dietary changes, compulsions, and sexual behavior in frontotemporal degeneration. Dementia 1995; 6:195–199Google Scholar

12. Swartz JR, Miller BL, Lesser IM, et al: Frontotemporal dementia: treatment response to serotonin selective reuptake inhibitors. J Clin Psychiatry 1997; 58:212–216Google Scholar

13. Liu W, Miller BL, Kramer JH, et al: Behavioral disorders in the frontal and temporal variants of frontotemporal dementia. Neurology 2004; 62:742–748Google Scholar

14. Boxer AL, Boeve BF: Frontotemporal dementia treatment: current symptomatic therapies and implications of recent genetic, biochemical, and neuroimaging studies. Alzheimer Dis Assoc Disord 2007; 21:S79–S87Google Scholar

15. Walker AJ, Meares S, Sachdev PS, et al: The differentiation of mild frontotemporal dementia from Alzheimer’s disease and healthy aging by neuropsychological tests. Int Psychogeriatr 2005; 17:57–68Google Scholar

16. Elderkin-Thompson V, Boone KB, Hwang S, et al: Neurocognitive profiles in elderly patients with frontotemporal degeneration or major depressive disorder. J Int Neuropsychol Soc 2004; 10:753–771Google Scholar

17. Graham A, Davies R, Xuereb J, et al: Pathologically proven frontotemporal dementia presenting with severe amnesia. Brain 2005; 128:597–605Google Scholar

18. Pasquier F, Grymonprez L, Lebert F, et al: Memory impairment differs in frontotemporal dementia and Alzheimer’s disease. Neurocase 2001; 7:161–171Google Scholar

19. Glosser G, Gallo JL, Clark CM, et al: Memory encoding and retrieval in frontotemporal dementia and Alzheimer’s disease. Neuropsychology 2002; 16:190–196Google Scholar

20. McMurtray AM, Chen AK, Shapira JS, et al: Variations in regional SPECT hypoperfusion and clinical features in frontotemporal dementia. Neurology 2006; 66:517–522Google Scholar

21. Kertesz A, McMonagle P, Blair M, et al: The evolution and pathology of frontotemporal dementia. Brain 2005; 128:1996–2005Google Scholar

22. Hansen LA, Deteresa R, Tobias H, et al: Neocortical morphometry and cholinergic neurochemistry in Pick’s disease. Am J Pathol 1998; 131:507–518Google Scholar

23. Procter AW, Qurne M, Francis PT: Neurochemical features of frontotemporal dementia. Dement Geriatr Cogn Disord 1999; 10(suppl 1):80–84Google Scholar

24. Sparks DL, Markesberry WR: Altered serotonergic and cholinergic synaptic markers in Pick’s disease. Arch Neurol 1991; 48:796–799Google Scholar

25. Meier-Ruge W, Iwangoff P, Reichlmeier K: Neurochemical enzyme changes in Alzheimer’s and Pick’s disease. Arch Gerontol Geriatr 1984; 3:161–165Google Scholar

26. Odawara T, Shiozaki K, Iseki E, et al: Alterations of muscarinic acetylcholine receptors in atypical Pick’s disease without Pick bodies. J Neurol Sci 2003; 74:965–967Google Scholar

27. Weinberger DR, Gibson R, Coppola R, et al: The distribution of cerebral muscarinic acetylcholine receptors in vivo in patients with dementia: a controlled study with 123IQNB and single photon emission computed tomography. Arch Neurol 1991; 48:169–176Google Scholar

28. Tagliavini F, Pilleri G: Basal nucleus of Meynert: a neuropathological study in Alzheimer’s disease, simple senile dementia, Pick’s disease, and Huntington’s chorea. J Neurol Sci 1983; 63:243–260Google Scholar

29. Uhl GR, Hilt DC, Hedreen JC, et al: Pick’s disease (lobar sclerosis): depletion of neurons in the nucleus basalis of Meynert. Neurology 1983; 33:1470–1473Google Scholar

30. Franceschi M, Anchisi D, Pelati O, et al: Glucose metabolism and serotonin receptors in the frontotemporal lobe degeneration. Ann Neurol 2005; 57:216–225Google Scholar

31. Miller BL: Pick’s Disease and Frontotemporal Dementias: Clinical and Biological Overview. Philadelphia, American Academy of Neurology, 2001Google Scholar

32. Yang Y, Schmitt HP: Frontotemporal dementia: evidence for impairment of ascending serotoninergic but not noradrenergic innervations: immunocytochemical and quantitative study using a graph method. Acta Neuropathol 2001; 101:256–270Google Scholar

33. Frisoni GB, Pizzolato G, Bianchetti A, et al: Single photon emission computed tomography with [99Tc]-HM-PAO and [123I]-IBZM in Alzheimer’s disease and dementia of frontal type: preliminary results. Acta Neurol Scand 1994; 89:199–203Google Scholar

34. Rinne JO, Laine M, Kaasinen V, et al: Striatal dopamine transporter and extrapyramidal symptoms in frontotemporal dementia. Neurology 2002; 58:1489–1493Google Scholar

35. Sperfeld AD, Collatz MB, Baier H, et al: Ftdp-17: an early-onset phenotype with parkinsonism and epileptic seizures caused by a novel mutation. Ann Neurol 1999; 46:708–715Google Scholar

36. Francis PT, Holmes C, Webster MT, et al: Preliminary neurochemical findings in non-Alzheimer dementia due to lobar atrophy. Dementia 1993; 4:172–177Google Scholar

37. Sjogren M, Wikkelso D, Ostling S, et al: Biological correlates of clinical subgroups of Alzheimer’s disease. Dement Geriatr Cogn Disord 2002; 14:191–197Google Scholar

38. Luque JM, Chan-Palay V: Alterations in tyrosine hydroxylase immunoreactive neurons of the locus ceruleus in Pick’s disease. Dementia 1991; 2:291–296Google Scholar

39. Ferrer I: Neurons and their dendrites in frontotemporal dementia. Dement Geriatr Cogn Disord 1999; 10(suppl 1):55–60Google Scholar

40. Mendez MF, Shapira JS: Preliminary findings: behavioral worsening on donepezil in patients with frontotemporal dementia. Am J Geriatr Psychiatry 2007; 15:84–87Google Scholar

41. Moretti R, Torre P, Antonello RM, et al: Rivastigmine in frontotemporal dementia: an open-label study. Drugs Aging 2004; 21:931–937Google Scholar

42. Kertesz A, Morlog D, Light M, et al: Galantamine in frontotemporal dementia and primary progressive aphasia. Dement Geriatr Cogn Disord 2008; 25:178–185Google Scholar

43. Lampl Y, Sadeh M, Lorberboym M: Efficacy of acetylcholinesterase inhibitors in frontotemporal dementia. Ann Pharmacother 2004; 38:1967–1968Google Scholar

44. Chow TW: Treatment approaches to symptoms associated with frontotemporal degeneration. Curr Psychiatry Rep 2005; 7:376–380Google Scholar

45. 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 [MA]Google Scholar

46. Swartz JR, Miller BL, Lesser IM, et al: Frontotemporal dementia: treatment response to serotonin selective reuptake inhibitors. J Clin Psychiatry 1997; 58:212–216Google Scholar

47. Mendez MF, Shapira JS, Miller BL: Stereotypical movements and frontotemporal dementia. Mov Disord 2005; 20:742–745Google Scholar

48. Ishikawa H, Shimomura T, Shimizu T: [Stereotyped behaviors and compulsive complaints of pain improved by fluvoxamine in two cases of frontotemporal dementia.] Seishin Shinkeigaku Zasshi 2006; 108:1029–1035 (Japanese)Google Scholar

49. Huey ED, Putnam KT, Grafman J: A systematic review of neurotransmitter deficits and treatments in frontotemporal dementia. Neurology 2006; 66:17–22Google Scholar

50. 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 [MA]Google Scholar

51. 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 2003; 172:400–408 [MA]Google Scholar

52. Lebert F, Stekke W, Hasenbroekx C, et al: Frontotemporal dementia: a randomized, controlled trial with trazodone. Dement Geriatr Cogn Disord 2004; 17:355–359 [MA]Google Scholar

53. Lebert F, Pasquier F: Trazodone in the treatment of behavior in frontotemporal dementia. Hum Psychopharmacol 1999; 14:279–281 [MA]Google Scholar

54. Sparks DL, Woeltz VM, Markesberry WR: Alterations in brain monoamine oxidase activity in aging, Alzheimer’s disease, and Pick’s disease. Arch Neurol 1991; 48:718–721Google Scholar

55. Adler G, Martin T, Drach LM: Pharmacological treatment of frontotemporal dementia: treatment response to the MAO-A inhibitor moclobemide. Int J Geriatr Psychiatry 2003; 18:653–655Google Scholar

56. Alafuzoff I, Helisalmi S, Heinonen EH, et al: Selegiline treatment and the extent of degenerative changes in brain tissue of patients with Alzheimer’s disease. Eur J Clin Pharmacol 2000; 55:815–819Google Scholar

57. Thomas T: Monoamine oxidase-B inhibitors in the treatment of Alzheimer’s disease. Neurobiol Aging 2000; 21:343–348Google Scholar

58. Moretti R, Torre P, Antonello RM, et al: Effects of selegiline on fronto-temporal dementia: a neuropsychological evaluation. Int J Geriatr Psychiatry 2002; 17:391–392 [MA]Google Scholar

59. Swanberg MM: Memantine for behavioral disturbances in frontotemporal dementia: a case series. Alzheimer Dis Assoc Disord 2007; 21:164–166Google Scholar

60. Fellgiebel A, Müller MJ, Hiemke C, et al: Clinical improvement in a case of frontotemporal dementia under aripiprazole treatment corresponds to partial recovery of disturbed frontal glucose metabolism. World J Biol Psychiatry 2007; 8:123–126Google Scholar

61. Boxer AL, Lipton AM, Womack K, et al: An open-label study of memantine treatment in 3 subtypes of frontotemporal lobar degeneration. Alzheimer Dis Assoc Disord 2009; 23:211–217Google Scholar

62. Curtis TC, Resch DS: Case of Pick’s central lobar atrophy with apparent stabilization of cognitive decline after treatment with risperidone. J Clin Psychopharmacol 2000; 20:384–385Google Scholar

63. Moretti R, Torre P, Antonello RM, et al: Olanzapine as a treatment of neuropsychiatric disorders of Alzheimer’s disease and other dementias: a 24-month follow-up of 68 patients. Am J Alzheimers Dis Other Demen 2003; 18:205–214Google Scholar

64. Kerrsens CJ, Pijnenburg YAL: Vulnerability to neuroleptic side effects in frontotemporal dementia. Eur J Neurol 2008; 15:111–112Google Scholar

65. Herrmann N, Lactot KL: Atypical antipsychotics for neuropsychiatric symptoms of dementia—malignant or maligned? Drug Saf 2006; 29:833–843Google Scholar

66. Rahman S, Robbins TW, Hodges R, et al: Methylphenidate (“Ritalin”) can ameliorate abnormal risk-taking behavior in the frontal variant of frontotemporal dementia. Neuropsychopharmacology 2006; 31:651–688Google Scholar

67. Peters F, Perani D, Herholz K, et al: Orbitofrontal dysfunction related to both apathy and disinhibition in frontotemporal dementia. Dement Geriatr Cogn Disord 2006; 21:373–379Google Scholar

68. Goforth HW, Konopka L, Primeau M, et al: Quantitative electroencephalography in frontotemporal dementia with methylphenidate response: a case study. Clin EEG Neurosci 2004; 35:108–111Google Scholar

69. Neumann M, Sampathu DM, Kwong JL, et al: Ubiquinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006; 314:130–133Google Scholar

70. Goldman JS, Farmer JM, Johnson JK, et al: Comparison of healthy histories in FTLD subtypes and related tauopathies. Neurology 2005; 65:1817–1819Google Scholar

71. Morita M, Al-Chalabi A, Andersen PM, et al: A locus on chromosome 9p confers susceptibility to ALS and frontotemporal dementia. Neurology 2006; 66:839–844Google Scholar

72. Vance C, Al-Chalabi A, Ruddy D, et al: Familial amyotrophic lateral sclerosis with frontotemporal dementia is linked to a locus on chromosome 9p13.2–21.3. Brain 2006; 129:868–876Google Scholar

73. Baker M, Mackenzie IR, Pickering-Brown SM, et al: Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17. Nature 2006; 442:916–919Google Scholar

74. Cruts M, Gijselinck I, van der Zee J, et al: Null mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17q21. Nature 2006; 442:920–924Google Scholar

75. Ahmed Z, Mackenzie IR, Hutton ML, et al: Progranulin in frontotemporal lobar degeneration and neuroinflammation. J Neuroinflammation 2007; 4:7Google Scholar

76. Neary D, Snowden J, Mann D: Frontotemporal dementia. Lancet Neurol 2005; 4:771–780Google Scholar

77. Foster NL, Wilhelmsen K, Sima AA, et al: Frontotemporal dementia and parkinsonism linked to chromosome 17: a consensus conference. Ann Neurol 1997; 41:706–715Google Scholar

78. Josephs KA, Petersen RC, Knopman DS, et al: Clinicopathic analysis of frontotemporal and corticobasal degenerations and PSP. Neurology 2006; 66:41–48Google Scholar

79. 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; 126:2016–2022Google Scholar

80. McKhann GM, Albert MS, Grossman M, et al: Clinical and pathological diagnosis of frontotemporal dementia: report of the work group on frontotemporal dementia and Pick’s disease. Arch Neurol 2001; 58:1803–1809Google Scholar

81. Khlistunova I, Biernat J, Wang Y, et al: Inducible expression of tau repeat domain in cell models of tauopathy: aggregation is toxic to cells but can be reversed by inhibitor drugs. J Biol Chem 2006; 281:1205–1214Google Scholar

82. Götz J, Ittner LM, Schonrock N: Alzheimer’s disease and frontotemporal dementia: prospects of a tailored therapy? Med J Aust 2006; 185:381–384Google Scholar

83. Wischik C, Bentham P, Wischik D, et al: Tau aggregation inhibitor (TAI) therapy with rember (MTC) arrests disease progression in mild and moderate Alzheimer’s disease over (50) 84 weeks. ICAD 2008: Alzheimer’s Association International Conference on Alzheimer’s Disease: Abstract 08-A-2332-ALZ. Presented July 29, 2008Google Scholar

84. Asuni AA, Boutajangout A, Quartermain D, et al: Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J Neurosci 2007; 27:9115–9129Google Scholar

85. Noble W, Planel E, Zehr C, et al: Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci USA 2005; 102:6990–6995Google Scholar

86. Zhang B, Maiti A, Shively S, et al: Microtubule-binding drugs offset tau sequestration by stabilizing microtubules and reversing fast axonal transport deficits in a tauopathy model. Proc Natl Acad Sci USA 2005; 102:227–231Google Scholar

87. Piguet O, Brooks WS, Halliday GM, et al: Similar early clinical presentations in familial and non-familial frontotemporal dementia. J Neurol Neurosurg Psychiatry 2004; 75:1743–1745Google Scholar

88. Hodges JR, Davies R, Xuereb J, et al: Survival in frontotemporal dementia. Neurology 2003; 61:349–354Google Scholar

89. Broe M, Hodges JR, Schofield E, et al: Staging disease severity in pathologically confirmed cases of frontotemporal dementia. Neurology 2003; 60:1005–1011Google Scholar

90. Frith U, Frith CD: Development and neurophysiology of mentalizing. Phil Trans R Soc Lond B: 2003; 358:459–473Google Scholar

91. D’Argembeau A, Ruby P, Collette F, et al: Distinct regions of the medial prefrontal cortex are associated with self-referential processing and perspective taking. J Cogn Neuroscience 2007; 19:935–944Google Scholar

92. Stuss DT, Gallup GG, Alexander MP: The frontal lobes are necessary for “theory of mind.” Brain 2001; 124:279–286Google Scholar

93. Wedderburn C, Wear H, Brown J, et al: The utility of the Cambridge Behavioral Inventory in neurodegenerative disease. J Neurol Neurosurg Psychiatry 2008; 79:500–503Google Scholar

94. Blair M, Kertesz A, Davis-Faroque N, et al: Behavioral measures in frontotemporal lobar dementia and other dementias: the utility of the Frontal Behavioral Inventory and the Neuropsychiatric Inventory in a national cohort study. Dement Geriatr Cogn Disord 2007; 23:406–415Google Scholar

95. Milan G, Lamenza F, Iavarone A, et al: Frontal Behavioral Inventory in the differential diagnosis of dementia. Acta Neurol Scand 2008; 117:260–265Google Scholar

96. Kertesz A, Davidson W, McCabe P, et al: Behavioral quantitation is more sensitive than cognitive testing in frontotemporal dementia. Alzheimer Dis Assoc Disord 2003; 17:223–239Google Scholar

97. Salmon E, Garaux G, Delbeuck X, et al: Predominant ventromedial frontopolar metabolic impairment in frontotemporal dementia. Neuroimage 2003; 20:435–440Google Scholar

98. Snowden JS, Neary D, Mann DMA: Frontotemporal Lobar Degeneration: Frontotemporal Dementia, Progressive Aphasia, Semantic Dementia. London, Churchill Livingstone, 1996Google Scholar

99. Freedman M: Frontotemporal dementia: recommendations for therapeutic studies, designs, and approaches. Canadian Journal of Neurology 2007; 34(suppl 1):S18–24Google Scholar