1 s2.0 S1878747923019116 Main
1 s2.0 S1878747923019116 Main
1 s2.0 S1878747923019116 Main
https://doi.org/10.1007/s13311-023-01380-6
CURRENT PERSPECTIVES
Abstract
Frontotemporal dementia (FTD) comprises a diverse group of clinical neurodegenerative syndromes characterized by progres-
sive changes in behavior, personality, executive function, language, and motor function. Approximately 20% of FTD cases
have a known genetic cause. The three most common genetic mutations causing FTD are discussed. Frontotemporal lobar
degeneration refers to the heterogeneous group of neuropathology underlying FTD clinical syndromes. While there are no
current disease-modifying treatments for FTD, management includes off-label pharmacotherapy and non-pharmacological
approaches to target symptoms. The utility of several different drug classes is discussed. Medications used in the treatment of
Alzheimer’s disease have no benefit in FTD and can worsen neuropsychiatric symptoms. Non-pharmacological approaches
to management include lifestyle modifications, speech-, occupational-, and physical therapy, peer and caregiver support,
and safety considerations. Recent developments in the understanding of the genetics, pathophysiology, neuropathology, and
neuroimmunology underlying FTD clinical syndromes have expanded possibilities for disease-modifying and symptom-
targeted treatments. Different pathogenetic mechanisms are targeted in several active clinical trials, opening up exciting
possibilities for breakthrough advances in treatment and management of FTD spectrum disorders.
13
Vol.:(0123456789)
1056 K. D. Neylan, B. L. Miller
with relative sparing of visuospatial abilities [4]. Pathology More than twenty genetic mutations have been discovered
is close to evenly divided between the tau or TAR DNA- to cause FTD spectrum disorders, but the three most com-
binding protein 43 (TDP-43) subtypes [5]. In approximately mon genetic mutations are discussed below.
5% of cases, abnormal aggregates of the fused-in-sarcoma
(FUS) protein are present. C9orf72 The most common cause of familial FTD and ALS
Semantic variant PPA presents with anomia and impaired is a genetic mutation with a hexanucleotide repeat expan-
single-word comprehension if the left temporal lobe is more sion GGGGCCin the non-coding region of the chromosome
severely damaged, and prominent emotional and behavioral 9 open reading frame 72 (C9orf72) gene. This mutation
dysfunction including irritability, loss of empathy, libido accounts for 13–26% of familial FTD cases [14, 15, 17–19].
changes, and inability to interpret social cues if right tem- It is inherited in an autosomal dominant fashion which exhib-
poral lobe damage predominates [6]. Approximately 7% of its increased penetrance with age (near 100% by age 80) and
patients with left svPPA develop new artistic abilities early possible anticipation phenomenon (earlier age of onset and
in the course of their illness [7–9]. Most cases of svPPA greater severity in subsequent generations) [20]. The most
show underlying TDP-43 pathology, although Pick’s disease common clinical presentation of C9orf72 forms of FTD is
pathology and FTD-MND changes are also seen. bvFTD, ALS, and FTD-ALS. CBS, nfvPPA, and svPPA
Non-fluent variant PPA presents with effortful non-fluent occur less frequently. It commonly presents with psychiatric
speech, apraxia of speech, and agrammatism [6]. Other defi- symptoms including hallucinations, delusions, and obsessive
cits may emerge with disease progression, including execu- compulsive or psychotic features [15, 16]. Age of onset is
tive dysfunction and motor features similar to those seen 20–91, though typically between 50 and 64 [20]. Survival is
in PSP or CBS [10, 11]. In those patients, the underlying variable, ranging from 1 to 22 years with a worse prognosis in
pathology is often congruent with those clinical syndromes ALS patients (on average 1.8 years) [16, 20]. C9orf72 forms
showing PSP or corticobasal degeneration (see below). of FTD are most associated with TDP-43 type B pathology,
Progressive supranuclear palsy (PSP) is characterized though TDP-43 type A and type U, while less common, do
by axial dystonia and rigidity, bradykinesia, supranuclear occur. On neuroimaging, patients tend to exhibit mild and
gaze palsy, and falls [12]. Neuropsychiatric syndromes are symmetric dorsal frontal, temporal, parietal, cerebellar, and
common and patients with PSP pathology often present as dorsomedial thalamic atrophy [20]. The pathogenesis of
a bvFTD or nfvPPA syndrome. Nearly all people with this C9orf72 repeat expansion is not fully understood, but pro-
clinical phenotype show a 4R tauopathy consistent with PSP. posed mechanisms include toxic gain of function, alteration
Corticobasal syndrome (CBS) is a clinical entity charac- of RNA expression, and haploinsufficiency [21].
terized by progressive asymmetric cognitive syndrome with
executive loss, parkinsonism, myoclonus, apraxia, dysto- GRN Progranulin is a highly conserved protein that serves
nia, and sometimes in the later stages, alien limb syndrome as a growth factor and plays a role in the regulation of cell
[12]. Clinicopathologic studies have found that CBS is a growth and survival, inflammation, and microglial and lyso-
neuropathologically heterogenous syndrome with a range somal function in the central nervous system [22]. Progranu-
of underlying pathology including corticobasal degenera- lin is encoded by the progranulin gene (GRN) on chromo-
tion (CBD)—a 4R tauopathy within the FTLD spectrum—as some 17. Homozygous GRN mutations lead to absence of
well as Alzheimer’s disease (AD) [12]. progranulin and neuronal ceroid lipofuscinosis, a lysosomal
Finally, there is syndromic and genetic overlap with storage disorder characterized by young-onset neurodegener-
bvFTD and motor neuron disease or ALS, with FTD, MND, ation [22]. Heterozygous GRN mutations lead to progranulin
and FTD-MND (also commonly referred to as FTD-ALS) deficiency and constitute about 8% of genetic forms of FTD
[13]. Nearly all patients with FTD-MND show TDP-43 [15]. GRN mutations are inherited in an autosomal dominant
neuropathology. fashion with more variable penetrance (about 90% by age
75) than MAPT and C9orf72 [23]. Clinically, patients with
GRN most commonly present with a bvFTD phenotype, but
Genetics nfvPPA, CBS, svPPA, or an atypical parkinsonism pheno-
type all occur [24, 25]. Symptom onset tends to be in the
Approximately 20% of FTD cases are genetic and 40% of 50s–80s, with a mean of about 65 years [23]. Survival on
patients with FTD have a family history of dementia or neu- average is 3–12 years [23]. GRN mutations lead to accu-
ropsychiatric illness [14–16]. Of the FTLD spectrum dis- mulation of TDP-43 type A in the brain. On neuroimaging,
orders, FTD-ALS is the most heritable while svPPA is the patients commonly have asymmetric atrophy of the dorsal
least genetic. frontoparietal regions [15, 24].
13
New Approaches to the Treatment of Frontotemporal Dementia 1057
MAPT Tau is a multifunctional protein encoded by the FTLD‑Tau Accumulation of hyperphosphorylated tau can
microtubule-associated protein tau (MAPT) gene on chro- cause neurodegenerative diseases known as tauopathies,
mosome 17 that interacts with microtubules to stabilize the which include FTLD and AD [27]. Accumulation of hyper-
neuronal cytoskeleton and plays a role in axonal transport phosphorylated 3R tau filaments known as “Pick bodies”
[26]. Tau is predominantly expressed in six isoforms via is seen in 20% of FTLD cases [14, 16]. Clinical syndromes
alternative mRNA splicing [27]. In tauopathies, hyperphos- associated with 3R tauopathies include bvFTD, CBS,
phorylation of tau protein disrupts cell function, leading to nfvPPA, and svPPA. With classical Pick’s disease neuropa-
deposition of aggregates of neurotoxic hyperphosphorylated thology, neuroimaging may show “knife-edge” cortical atro-
tau [26]. Genetic mutations in MAPT lead to an imbalance phy (Fig. 1) [14, 33]. Accumulation of hyperphosphorylated
of 3R/4R tau isoforms and comprise 17–32% of cases of 4R tau inclusions is seen in CBD, PSP, globular glial tauopa-
bvFTD with a positive family history [15]. MAPT mutations thy, and argyrophilic grain disease, with a subset of those
are inherited in an autosomal dominant fashion with a high with 4R tauopathies presenting with FTD clinical syndromes
degree of penetrance [28]. Clinical presentation is variable [12, 33]. For example, approximately 50% of the nfvPPA
and may span the range of FTD phenotypes: bvFTD, svPPA, patients seen at the University of California San Francisco
nfvPPA, CBS, PSP, or AD-like presentation [12]. Patients Memory and Aging Center show CBD neuropathology and
often have a psychiatric prodrome and psychotic features. 85% have an underlying tauopathy [34].
Age of onset ranges from the 20s to 80s, with a mean around
49–50 [28]. Different mutations in MAPT can be associated FTLD‑FUS The fused-in-sarcoma (FUS) protein binds to
with either 3R or 4R tau pathology and vary as to their ages DNA and RNA and regulates expression of more than 100
at onset. On neuroimaging, patients with MAPT mutations proteins [35]. Cytoplasmic aggregations of FUS protein can
tend to have symmetric anterior temporal lobe and basal cause FTD or FTD-MND, typically with very early onset in
ganglia atrophy [15]. the third to fifth decades (20–40) [16]. Patients with FTLD-
FUS often have compulsive behavior, prominent psychotic
Rarer genetic causes of FTD include mutations in FUS, symptoms, and hyperorality [14].
TARDBP, VCP, and UBQLN2. For a more comprehensive
review, please refer to Deleon and Miller [29].
Neuropathology
13
1058 K. D. Neylan, B. L. Miller
In FTD, there is no benefit for the pharmacotherapies found Pharmacotherapy Targeting Motor Symptoms
useful for Alzheimer’s disease. Cholinesterase inhibitors,
one of the primary classes of medications used in the treat- Patients with PSP and CBD show atypical parkinsonian symp-
ment of AD which include donepezil, rivastigmine, and toms, with predominant rigidity and infrequent tremor. Unlike
galantamine, have been studied in FTD and show no effect patients with Parkinson’s disease (PD), patients with FTD-
on cognitive performance but can actually worsen neuropsy- related parkinsonism tend not to be responsive to dopamine
chiatric symptoms [15, 36]. One small study demonstrated replacement therapy including carbidopa-levodopa [46]. Still,
an improvement in patient behavior and caregiver reports some patients experience transient and usually mild motoric
of burden after donepezil was discontinued in patients with improvement, so physicians can consider a trial of dopamine
FTD who were previously given a diagnosis of AD [37]. replacement therapy in patients with functional impairments
Donepezil can worsen both gait and dysphagia in PSP [38]. due to tremor, bradykinesia, or rigidity [12, 41]. Side effects to
The NMDA receptor inhibitor memantine had no benefit monitor include nausea, hypotension, and psychosis.
on either cognition or behavior in FTD patients and was In patients with ALS, the FDA has approved riluzole,
associated with worsening of neuropsychiatric symptoms edaravone, and Relyvrio (combination of sodium phenylb-
in some patients [39, 40]. The NMDA-receptor antagonist utyrate and taurursodiol) as disease-modifying treatments
amantadine may improve gait and freezing in some patients [48, 49]. Riluzole was not found to affect rates of functional
with PSP but adverse events include visual hallucinations decline or survival in patients with PSP [50].
and delirium [41].
Pharmacotherapy Targeting Pseudobulbar Affect
Selective Serotonin Reuptake Inhibitors (SSRIs)
Pseudobulbar affect (PBA), characterized by sudden bouts of
The class of medications with the largest supportive body uncontrollable crying or laughter, is frequently seen in FTD-
of evidence in FTD is the SSRIs. Several clinical trials of MND and PSP [51]. Several medications can help reduce the
13
New Approaches to the Treatment of Frontotemporal Dementia 1059
severity and frequency of pseudobulbar affect. Dextromethorphan- decline with age and with improved outcomes in several
quinidine sulfate (brand name Nuedexta®) is the first FDA- neurodegenerative diseases including AD, PD, and vascu-
approved medication specifically targeting PBA [51]. Antide- lar dementia. A recent study found physical and cognitive
pressants including SSRIs and serotonin-norepinephrine reup- exercise to be inversely related to disease severity and rate
take inhibitors (SNRIs) are also used as off-label treatment for of annual clinical decline in familial FTD [59]. In addition
PBA and may additionally help with concurrent behavior and to improving cardiovascular health, aerobic exercise can
neuropsychiatric symptoms. Tricyclic antidepressants have also increase strength and balance and reduce falls [41].
shown efficacy in treating PBA but should be used with cau- Other lifestyle modifications that are associated with bet-
tion in patients with FTD spectrum disorders given significant ter outcomes in dementia include eating a balanced diet and
anticholinergic side effects. limiting alcohol intake although studies of FTD populations
are lacking. Dietary changes may pose a challenge to some
Antiepileptic Medications patients with FTD as changes in appetite and physical activ-
ity are often major features of the disease. Therefore, it may
There is limited evidence suggesting a role for antiepileptic or may not be a realistic goal for an individual patient.
medications in FTD. Mood stabilizers such as carbamaz-
epine and valproic acid have been used in the management Speech Therapy
of mania in bipolar disorder, which has symptomatic over-
lap with bvFTD including impulsivity and irritability [46]. Speech therapy can be effective in patients with PPA or
There are limited studies suggesting the utility of mood sta- other FTLD spectrum disorders who have apraxia of speech,
bilizers in FTD, and there are only a few case reports that dysarthria, or hypophonia [60]. It is particularly effective
document improved behavior and agitation [52–54]. There if the speech therapist is trained in neurodegenerative apha-
have been case reports of the use of topiramate in reduc- sias. Sessions may be conducted effectively in person or
ing hyperorality in FTD patients [55, 56]. As antiepileptic via telehealth [61]. Speech therapy in PPA may include ele-
medications have significant side effects including gastro- ments of script training, the development of strategies for
intestinal upset, electrolyte imbalance, kidney stones, and improving communication, training on non-verbal commu-
possible adverse drug reactions, the risks and benefits must nication, and the use of augmentative or alternate modes of
be carefully considered. Currently, there is not enough qual- communication (AAC) including communication books or
ity evidence to recommend the use of antiepileptic medica- assistive technology. Patients with svPPA can often learn
tions in FTD. approximately 25 words that they need for day-to-day com-
munication. Speech therapists can also provide resources for
Medications to Avoid in FTD cognitive rehabilitation including organization strategies for
executive dysfunction [62]. Patients with dysphagia should
In general, patients should avoid or use with caution medica- undergo a swallowing evaluation.
tions with strong cognitive side effects, including benzodi-
azepines, antihistamines, and tricyclic antidepressants [41]. Physical and Occupational Therapy
Activating medications such as methylphenidate and dexa-
mphetamine have mixed reports in the literature but should Many patients with motor symptoms may benefit from
be used with great caution, if at all [57]. Adverse effects physical therapy to work on strength, gait, range of motion,
such as increased behavioral disturbances and hallucinations and balance training [63]. Occupational therapists can assess
have been noted in patients with PD taking dexamphetamine patients’ cognitive and physical status and provide strate-
[46]. There have been small studies in FTD patients demon- gies and interventions to maximize function—for example,
strating improved apathy and disinhibition, though no large the use of step-by-step “activity templates” or visual aids
placebo-controlled studies have been performed [58]. Dopa- for patients with apraxia or executive dysfunction. Occupa-
mine agonists and the NMDA-receptor antagonist amanta- tional therapy can also perform home safety evaluations to
dine do not improve symptoms in PSP [41]. prevent falls and provide assistive devices for activities of
daily living [41].
Non‑pharmacological Approaches to the Treatment
of FTD Peer Support Groups
13
1060 K. D. Neylan, B. L. Miller
patients may find use in support groups designed for patients to the point of choking, attempt to consume non-food items,
with younger onset dementia or PPA [65]. Research on the or risk burning their mouth on hot food given disrupted pain
impact of support groups in bvFTD is limited, but patients responses.
with PPA have reported improved quality of life, confidence, Another safety consideration is the removal of firearms
hope, and practical coping strategies from peer support and from the home.
education programs [66].
Caregivers of patients with FTD demonstrate high rates of Frontotemporal dementia encompasses a wide spectrum of
burden and distress [67]. Efforts to improve caregiver sup- diverse clinical presentations and underlying pathology, pos-
port, education, coping skills, and strategies for redirecting ing a challenge to therapeutic development. Recent advance-
or minimizing unwanted behaviors have all shown success ments in the understanding of the genetics, pathophysiology,
in reducing caregiver stress [68]. Organizations such as the neuropathology, and neuroimmunology of the frontotempo-
Association for Frontotemporal Degeneration and CurePSP ral dementia spectrum of disorders have expanded possibili-
offer information-rich resources and peer support groups for ties for disease-modifying and symptom-targeted precision
caregivers and families. Caregivers may benefit from respite medicine treatments.
care, short-term caregiving services, or enrolling patients in
longer-duration day care programs. Pharmacotherapy and Gene Therapy Approaches
The American Academy of Neurology recommends early The autosomal dominant forms of FTD represent an area
integration of palliative care in patients with progressive of blossoming research in drug development utilizing gene
neurodegenerative disease including ALS and FTD spec- therapy techniques and the use of antisense oligonucleotides
trum disorders, with particular attention paid to eliciting (ASOs) or other small molecules. One advantage of study-
patient preferences before disease progression precludes ing genetic populations is that early intervention is possible.
capacity and effective communication [69]. Referral to pal- Similarly, the therapies are known to be directed towards the
liative care demonstrates improved quality of life in PSP specific cause for FTD.
and ALS [41, 70]. Therefore, planning ahead with advanced Several studies have targeted progranulin replacement in
care directives and assigning a designated power of attor- patients with GRN mutations. Prior trials using nimodipine
ney are recommended early in the disease course whenever and histone deacetylase inhibitors did not successfully raise
possible [41]. Patients with motor neuron disease should progranulin concentrations [72, 73]. Newer techniques in
be connected with ALS centers for multidisciplinary care. development for GRN mutations include the use of adeno-
As FTD commonly affects patients of working age, many associated viral vector therapies (active trials include
families undergo significant financial stress. Patients may NCT04747431 and NCT04408625), recombinant progranu-
qualify for Social Security disability benefits. Patients are lin protein replacement, antibody delivery of progranulin to
at increased risk for financial victimization or exorbitant the brain (active trials include NCT05262023), and the use of
spending. Laws regarding reporting of dementia diagno- monoclonal antibodies targeting sortilin, a protein involved in
sis to the Department of Motor Vehicles differ by state, progranulin degradation, to investigate the effect of increased
but patients with frontotemporal dementia have difficulty progranulin concentration on patients with GRN mutations
driving safely. They should, at the very least, have regular (active trials include NCT03987295 and NCT04374136) [21,
driving assessment. Deficits in executive function, impulse 74–77]. Several different candidate ASOs have been devel-
control, and judgment, as well as motor and gaze abnormali- oped to try to suppress the aberrant RNA transcript expan-
ties in FTD spectrum disorders, can all impair safety while sion seen in C9orf72 mutation carriers with active trials for
driving. ALS and FTD patients (NCT04931862) [21].
Patients with FTD may require supervision given
impaired judgment, a lack of typical responses to pain stim-
uli, and risk of inadvertent self-harm related to compulsive Tau‑Targeted Approaches
behaviors [71]. For example, patients may have repetitive
pacing or roaming behavior to the point of developing blis- Clinical trials targeting tau protein have utilized different
ters. Patients may need to be monitored while eating to mini- strategies to reduce accumulation of hyperphosphorylated
mize choking, as they may eat quickly, overfill their mouths tau, including using gene therapy techniques to alter MAPT
13
New Approaches to the Treatment of Frontotemporal Dementia 1061
13
1062 K. D. Neylan, B. L. Miller
myeloperoxidase, with the aim of reducing oxidative stress Required Author Forms Disclosure forms provided by the authors are
and pathologic activation of microglia leading to cell death available with the online version of this article.
(NCT05184569). Verdiperstat has previously been stud- Funding This study was supported by NIH/NIA P30AG062422 and
ied in multiple system atrophy and ALS and demonstrated P01AG019724 (BM) and the Rainwater Charitable Foundation.
safety but not efficacy on measures of disease progression
(NCT03952806, NCT04436510, NCT02388295). Another
approach using a derivative of a nucleoside analog reverse References
transcriptase inhibitor originally developed for HIV is cur-
rently under investigation. This involves a Phase 2a trial of 1. Neary D, Snowden J, Mann D. Frontotemporal dementia. Lancet
patients with ALS or FTD with a C9orf72 mutation with the Neurol. 2005;4:771–80.
2. Seo SW, Thibodeau MP, Perry DC, et al. Early vs late age at
aim of preventing the neuroinflammatory response triggered onset frontotemporal dementia and frontotemporal lobar degen-
by TDP-43 inclusion accumulation (NCT04993755). eration. Neurology. 2018;90:e1047–56.
Lastly, preclinical laboratory studies are actively investi- 3. Onyike CU, Diehl-Schmid J. The epidemiology of frontotempo-
gating whether the recent advancements in pharmacotherapy ral dementia. Int Rev Psychiatry. 2013;25:130–7.
4. Rascovsky K, Hodges JR, Knopman D, et al. Sensitivity of
for ALS could have a role in the treatment of FTD. Edara- revised diagnostic criteria for the behavioural variant of fronto-
vone, a free radical scavenger, reduced tau phosphorylation temporal dementia. Brain. 2011;134:2456–77.
and neuroinflammation in a mouse model of FTD [99]. 5. Snowden J, Neary D, Mann D. Frontotemporal lobar degenera-
tion: clinical and pathological relationships. Acta Neuropathol.
2007;114:31–8.
Non‑pharmacological Clinical Trials 6. Gorno-Tempini ML, Hillis AE, Weintraub S, et al. Classifica-
tion of primary progressive aphasia and its variants. Neurology.
For PPA, an active area of investigation involves the use 2011;76:1006–14.
of transcranial direct current stimulation (tDCS), a non- 7. Miller BL, Cummings J, Mishkin F, et al. Emergence of artistic
talent in frontotemporal dementia. Neurology. 1998;51:978–82.
invasive method of applying electrical current to stimulate 8. Erkkinen MG, Zuniga RG, Pardo CC, Miller BL, Miller ZA.
particular brain regions, with initial results suggesting mod- Artistic renaissance in frontotemporal dementia. JAMA.
erate enhancement in language outcomes [100, 101]. Vari- 2018;319:1304–6.
ous studies have adapted tDCS protocols to examine effects 9. Friedberg A, Pasquini L, Diggs R, et al. Prevalence, timing, and
network localization of emergent visual creativity in frontotem-
on language abilities in patients with PPA, with and without poral dementia. JAMA Neurol. 2023.
concurrent speech and language therapy [102, 103]. There 10. Montembeault M, Brambati SM, Gorno-Tempini ML, Migliaccio
are several studies of this type currently active (includ- R. Clinical, anatomical, and pathological features in the three
ing NCT05615922, NCT04486586, NCT05368350, and variants of primary progressive aphasia: a review. Front Neurol.
2018;9:692.
NCT04566731). Other studies are using comparable design 11. Grossman M. The non-fluent/agrammatic variant of primary
with transcranial magnetic stimulation (TMS) in patients progressive aphasia. Lancet Neurol. 2012;11:545–55.
with PPA (recently completed trials include NCT03728582 12. Hoglinger GU, Respondek G, Stamelou M, et al. Clinical diag-
and NCT03406429) [104]. nosis of progressive supranuclear palsy: the movement disorder
society criteria. Mov Disord. 2017;32:853–64.
Using a similar paradigm, the use of transcranial stimu- 13. Cividini C, Basaia S, Spinelli EG, et al. Amyotrophic lateral sclero-
lation in other FTD syndromes is actively under investiga- sis-frontotemporal dementia: shared and divergent neural correlates
tion. One study in patients with FTD found tDCS led to across the clinical spectrum. Neurology. 2021;98:e402-415.
improvement of clinical scores and behavioral symptoms 14. Bang J, Spina S, Miller BL. Frontotemporal dementia. Lancet.
2015;386:1672–82.
[105]. Another active trial is using transcranial alternating 15. Bott NT, Radke A, Stephens ML, Kramer JH. Frontotemporal
current stimulation (tACS) in a cohort of bvFTD patients dementia: diagnosis, deficits and management. Neurodegener Dis
(NCT04425148). Manag. 2014;4:439–54.
While there are currently no disease-modifying treat- 16. Younes K, Miller BL. Frontotemporal dementia: neuropathology,
genetics, neuroimaging, and treatments. Psychiatr Clin North
ments available for FTD, the rapid expansion of knowledge Am. 2020;43:331–44.
in pathogenesis, genetics, neuropathology, and neuroimmu- 17. Morita M, Al-Chalabi A, Andersen PM, et al. A locus on chro-
nology across the main variants of FTD and the advances mosome 9p confers susceptibility to ALS and frontotemporal
in gene therapies have opened up exciting possibilities for dementia. Neurology. 2006;66:839–44.
18. Vance C, Al-Chalabi A, Ruddy D, et al. Familial amyotrophic
breakthrough advances in this family of disorders. We are lateral sclerosis with frontotemporal dementia is linked to a locus
on the cusp of a new era of treatment and management for on chromosome 9p13.2-21.3. Brain. 2006;129:868–76.
frontotemporal dementia spectrum disorders. 19. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, et al. Expanded
GGGG CC hexanucleotide repeat in noncoding region of
Supplementary Information The online version contains supplemen- C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron.
tary material available at https://d oi.o rg/1 0.1 007/s 13311-0 23-0 1380-6. 2011;72:245–56.
13
New Approaches to the Treatment of Frontotemporal Dementia 1063
20. Gossye H, Engelborghs S, Van Broeckhoven C, van der Zee J. 41. Bluett B, Pantelyat AY, Litvan I, et al. Best practices in the
C9orf72 frontotemporal dementia and/or amyotrophic lateral clinical management of progressive supranuclear palsy and
sclerosis. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, corticobasal syndrome: a consensus statement of the CurePSP
Wallace SE, Bean LJH, et al., editors. GeneReviews((R)). Seattle Centers of Care. Front Neurol. 2021;12:694872.
(WA); 1993. 42. Nisar M, Abubaker ZJ, Nizam MA, et al. Behavioral and cog-
21. Boeve BF, Boxer AL, Kumfor F, Pijnenburg Y, Rohrer JD. nitive response to selective serotonin reuptake inhibitors in
Advances and controversies in frontotemporal dementia: diag- frontotemporal lobar degeneration: a systematic review and
nosis, biomarkers, and therapeutic considerations. Lancet Neurol. meta-analysis. Clin Neuropharmacol. 2021;44:175–83.
2022;21:258–72. 43. Herrmann N, Black SE, Chow T, et al. Serotonergic function and
22. Townley RA, Boeve BF, Benarroch EE. Progranulin: functions treatment of behavioral and psychological symptoms of fronto-
and neurologic correlations. Neurology. 2018;90:118–25. temporal dementia. Am J Geriatr Psychiatry. 2012;20:789–97.
23. Hsiung GYR, Feldman HH. GRN frontotemporal dementia. In: 44. Sanchez C, Reines EH, Montgomery SA. A comparative review
Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, of escitalopram, paroxetine, and sertraline: are they all alike?
Bean LJH, et al., editors. GeneReviews((R)). Seattle (WA); 1993. Int Clin Psychopharmacol. 2014;29:185–96.
24. Beck J, Rohrer JD, Campbell T, et al. A distinct clinical, neu- 45. Pijnenburg YA, Sampson EL, Harvey RJ, Fox NC, Rossor MN.
ropsychological and radiological phenotype is associated Vulnerability to neuroleptic side effects in frontotemporal lobar
with progranulin gene mutations in a large UK series. Brain. degeneration. Int J Geriatr Psychiatry. 2003;18:67–72.
2008;131:706–20. 46. Tsai RM, Boxer AL. Treatment of frontotemporal dementia.
25. Le Ber I, Camuzat A, Hannequin D, et al. Phenotype variability Curr Treat Options Neurol. 2014;16:319.
in progranulin mutation carriers: a clinical, neuropsychological, 47. Shin HW, Chung SJ. Drug-induced parkinsonism. J Clin Neu-
imaging and genetic study. Brain. 2008;131:732–46. rol. 2012;8:15–21.
26. Boyarko B, Hook V. Human tau isoforms and proteolysis for 48. Cho H, Shukla S. Role of edaravone as a treatment option for patients
production of toxic tau fragments in neurodegeneration. Front with amyotrophic lateral sclerosis. Pharmaceuticals (Basel).
Neurosci. 2021;15:702788. 2020;14.
27. Spillantini MG, Goedert M. Tau pathology and neurodegeneration. 49. Paganoni S, Hendrix S, Dickson SP, et al. Effect of sodium
Lancet Neurol. 2013;12:609–22. phenylbutyrate/taurursodiol on tracheostomy/ventilation-free
28. Moore KM, Nicholas J, Grossman M, et al. Age at symptom survival and hospitalisation in amyotrophic lateral sclerosis:
onset and death and disease duration in genetic frontotemporal long-term results from the CENTAUR trial. J Neurol Neuro-
dementia: an international retrospective cohort study. Lancet surg Psychiatry. 2022;93:871–5.
Neurol. 2020;19:145–56. 50. Bensimon G, Ludolph A, Agid Y, et al. Riluzole treatment,
29. Deleon J, Miller BL. Frontotemporal dementia. Handb Clin Neu- survival and diagnostic criteria in Parkinson plus disorders:
rol. 2018;148:409–30. the NNIPPS study. Brain. 2009;132:156–71.
30. Pennington C, Marini L, Coulthard E, Love S. Mixed neuropa- 51. Hakimi M, Maurer CW. Pseudobulbar affect in parkinsonian
thology in frontotemporal lobar degeneration. Amyotroph Lateral disorders: a review. J Mov Disord. 2019;12:14–21.
Scler Frontotemporal Degener. 2020;21:301–8. 52. Poetter CE, Stewart JT. Treatment of indiscriminate, inappropri-
31. Yousef A, Robinson JL, Irwin DJ, et al. Neuron loss and degen- ate sexual behavior in frontotemporal dementia with carbamaz-
eration in the progression of TDP-43 in frontotemporal lobar epine. J Clin Psychopharmacol. 2012;32:137–8.
degeneration. Acta Neuropathol Commun. 2017;5:68. 53. Devanand DP, Pelton GH, D’Antonio K, et al. Low-dose lith-
32. Lee EB, Porta S, Michael Baer G, et al. Expansion of the clas- ium treatment for agitation and psychosis in Alzheimer dis-
sification of FTLD-TDP: distinct pathology associated with rap- ease and frontotemporal dementia: a case series. Alzheimer
idly progressive frontotemporal degeneration. Acta Neuropathol. Dis Assoc Disord. 2017;31:73–5.
2017;134:65–78. 54. Chow TW, Mendez MF. Goals in symptomatic pharmacologic
33. Coughlin DG, Dickson DW, Josephs KA, Litvan I. Progressive management of frontotemporal lobar degeneration. Am J Alz-
supranuclear palsy and corticobasal degeneration. Adv Exp Med heimers Dis Other Demen. 2002;17:267–72.
Biol. 2021;1281:151–76. 55. Singam C, Walterfang M, Mocellin R, Evans A, Velakoulis D.
34. Lee SE, Rabinovici GD, Mayo MC, et al. Clinicopathological correla- Topiramate for abnormal eating behaviour in frontotemporal
tions in corticobasal degeneration. Ann Neurol. 2011;70:327–40. dementia. Behav Neurol. 2013;27:285–6.
35. Deng H, Gao K, Jankovic J. The role of FUS gene variants in 56. Shinagawa S, Tsuno N, Nakayama K. Managing abnormal eating
neurodegenerative diseases. Nat Rev Neurol. 2014;10:337–48. behaviours in frontotemporal lobar degeneration patients with
36. Mendez MF, Shapira JS, McMurtray A, Licht E. Preliminary topiramate. Psychogeriatrics. 2013;13:58–61.
findings: behavioral worsening on donepezil in patients with 57. Kaye ED, Petrovic-Poljak A, Verhoeff NP, Freedman M. Fron-
frontotemporal dementia. Am J Geriatr Psychiatry. 2007;15:84–7. totemporal dementia and pharmacologic interventions. J Neu-
37. Kimura T, Takamatsu J. Pilot study of pharmacological treat- ropsychiatry Clin Neurosci. 2010;22:19–29.
ment for frontotemporal dementia: risk of donepezil treatment 58. Huey ED, Garcia C, Wassermann EM, Tierney MC, Grafman J.
for behavioral and psychological symptoms. Geriatr Gerontol Stimulant treatment of frontotemporal dementia in 8 patients. J
Int. 2013;13:506–7. Clin Psychiatry. 2008;69:1981–2.
38. Litvan I, Phipps M, Pharr VL, et al. Randomized placebo- 59. Casaletto KB, Staffaroni AM, Wolf A, et al. Active lifestyles mod-
controlled trial of donepezil in patients with progressive supra- erate clinical outcomes in autosomal dominant frontotemporal
nuclear palsy. Neurology. 2001;57:467–73. degeneration. Alzheimers Dement. 2020;16:91–105.
39. Boxer AL, Knopman DS, Kaufer DI, et al. Memantine in 60. Henry ML, Meese MV, Truong S, et al. Treatment for apraxia of
patients with frontotemporal lobar degeneration: a multicen- speech in nonfluent variant primary progressive aphasia. Behav
tre, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2013;26:77–88.
Neurol. 2013;12:149–56. 61. Dial HR, Hinshelwood HA, Grasso SM, et al. Investigating the
40. Vercelletto M, Boutoleau-Bretonniere C, Volteau C, et al. utility of teletherapy in individuals with primary progressive
Memantine in behavioral variant frontotemporal dementia: aphasia. Clin Interv Aging. 2019;14:453–71.
negative results. J Alzheimers Dis. 2011;23:749–59.
13
1064 K. D. Neylan, B. L. Miller
62. Khayum B, Wieneke C, Rogalski E, Robinson J, O’Hara M. supranuclear palsy: a phase 2, randomized, placebo-controlled
Thinking outside the stroke: treating primary progressive aphasia trial. Nat Med. 2021;27:1451–7.
(PPA). Perspect Gerontol. 2012;17:37–49. 82. Shiells H, Schelter BO, Bentham P, et al. Concentration-
63. Steffen TM, Boeve BF, Mollinger-Riemann LA, Petersen CM. dependent activity of hydromethylthionine on clinical decline
Long-term locomotor training for gait and balance in a patient and brain atrophy in a randomized controlled trial in behav-
with mixed progressive supranuclear palsy and corticobasal ioral variant frontotemporal dementia. J Alzheimers Dis.
degeneration. Phys Ther. 2007;87:1078–87. 2020;75:501–19.
64. Medina J, Weintraub S. Depression in primary progressive apha- 83. Vivash L, Malpas CB, Meletis C, et al. A phase 1b open-label
sia. J Geriatr Psychiatry Neurol. 2007;20:153–60. study of sodium selenate as a disease-modifying treatment for
65. Taylor-Rubin C, Azizi L, Croot K, Nickels L. Primary progressive possible behavioral variant frontotemporal dementia. Alzhei-
aphasia education and support groups: a clinical evaluation. Am mers Dement (N Y). 2022;8:e12299.
J Alzheimers Dis Other Demen. 2020;35:1533317519895638. 84. Dai CL, Chen X, Kazim SF, et al. Passive immunization tar-
66. Morhardt DJ, O’Hara MC, Zachrich K, Wieneke C, Rogalski EJ. geting the N-terminal projection domain of tau decreases
Development of a psycho-educational support program for indi- tau pathology and improves cognition in a transgenic mouse
viduals with primary progressive aphasia and their care-partners. model of Alzheimer disease and tauopathies. J Neural Transm
Dementia (London). 2019;18:1310–27. (Vienna). 2015;122:607–17.
67. Wong C, Merrilees J, Ketelle R, et al. The experience of car- 85. Hoglinger GU, Huppertz HJ, Wagenpfeil S, et al. Tideglusib
egiving: differences between behavioral variant of frontotem- reduces progression of brain atrophy in progressive supra-
poral dementia and Alzheimer disease. Am J Geriatr Psychiatry. nuclear palsy in a randomized trial. Mov Disord. 2014;29:
2012;20:724–8. 479–87.
68. Vandepitte S, Van Den Noortgate N, Putman K, et al. Effective- 86. Tsai RM, Miller Z, Koestler M, et al. Reactions to multiple
ness of respite care in supporting informal caregivers of persons ascending doses of the microtubule stabilizer TPI-287 in patients
with dementia: a systematic review. Int J Geriatr Psychiatry. with Alzheimer disease, progressive supranuclear palsy, and cor-
2016;31:1277–88. ticobasal syndrome: a randomized clinical trial. JAMA Neurol.
69. Taylor LP, Besbris JM, Graf WD, et al. Clinical guidance in 2020;77:215–24.
neuropalliative care: an AAN position statement. Neurology. 87. Sud R, Geller ET, Schellenberg GD. Antisense-mediated exon
2022;98:409–16. skipping decreases tau protein expression: a potential therapy for
70. Bede P, Oliver D, Stodart J, et al. Palliative care in amyotrophic tauopathies. Mol Ther Nucleic Acids. 2014;3:e180.
lateral sclerosis: a review of current international guidelines and 88. DeVos SL, Miller RL, Schoch KM, et al. Tau reduction prevents
initiatives. J Neurol Neurosurg Psychiatry. 2011;82:413–8. neuronal loss and reverses pathological tau deposition and seed-
71. Bathgate D, Snowden JS, Varma A, Blackshaw A, Neary D. ing in mice with tauopathy. Sci Transl Med. 2017;9.
Behaviour in frontotemporal dementia, Alzheimer’s disease and 89. Hoglinger GU, Litvan I, Mendonca N, et al. Safety and effi-
vascular dementia. Acta Neurol Scand. 2001;103:367–78. cacy of tilavonemab in progressive supranuclear palsy: a
72. Sha SJ, Miller ZA, Min SW, et al. An 8-week, open-label, dose- phase 2, randomised, placebo-controlled trial. Lancet Neurol.
finding study of nimodipine for the treatment of progranulin 2021;20:182–92.
insufficiency from GRN gene mutations. Alzheimers Dement 90. VandeVrede L, Dale ML, Fields S, et al. Open-label phase 1 futil-
(N Y). 2017;3:507–12. ity studies of salsalate and young plasma in progressive supranu-
73. Ljubenkov PA, Edwards L, Iaccarino L, et al. Effect of the his- clear palsy. Mov Disord Clin Pract. 2020;7:440–7.
tone deacetylase inhibitor FRM-0334 on progranulin levels in 91. Boxer AL, Lang AE, Grossman M, et al. Davunetide in patients
patients with progranulin gene haploinsufficiency: a randomized with progressive supranuclear palsy: a randomised, double-
clinical trial. JAMA Netw Open. 2021;4:e2125584. blind, placebo-controlled phase 2/3 trial. Lancet Neurol.
74. Miyakawa S, Sakuma H, Warude D, et al. Anti-sortilin1 antibody 2014;13:676–85.
up-regulates progranulin via sortilin1 down-regulation. Front 92. Oliver LD, Stewart C, Coleman K, et al. Neural effects of oxy-
Neurosci. 2020;14:586107. tocin and mimicry in frontotemporal dementia: a randomized
75. Hinderer C, Miller R, Dyer C, et al. Adeno-associated virus crossover study. Neurology. 2020;95:e2635–47.
serotype 1-based gene therapy for FTD caused by GRN muta- 93. Finger E, Berry S, Cummings J, et al. Adaptive crossover designs
tions. Ann Clin Transl Neurol. 2020;7:1843–53. for assessment of symptomatic treatments targeting behaviour in
76. Hu F, Padukkavidana T, Vaegter CB, et al. Sortilin-mediated neurodegenerative disease: a phase 2 clinical trial of intranasal
endocytosis determines levels of the frontotemporal dementia oxytocin for frontotemporal dementia (FOXY). Alzheimers Res
protein, progranulin. Neuron. 2010;68:654–67. Ther. 2018;10:102.
77. Arrant AE, Onyilo VC, Unger DE, Roberson ED. Progranulin 94. Sawyer RP, Hill EJ, Yokoyama J, et al. Differences in peripheral
gene therapy improves lysosomal dysfunction and microglial immune system gene expression in frontotemporal degeneration.
pathology associated with frontotemporal dementia and neu- Medicine (Baltimore). 2022;101:e28645.
ronal ceroid lipofuscinosis. J Neurosci. 2018;38:2341–58. 95. Miller ZA, Rankin KP, Graff-Radford NR, et al. TDP-43 fronto-
78. VandeVrede L, Boxer AL, Polydoro M. Targeting tau: clini- temporal lobar degeneration and autoimmune disease. J Neurol
cal trials and novel therapeutic approaches. Neurosci Lett. Neurosurg Psychiatry. 2013;84:956–62.
2020;731: 134919. 96. Hartnell IJ, Blum D, Nicoll JAR, Dorothee G, Boche D. Glial
79. Olney NT, Spina S, Miller BL. Frontotemporal dementia. Neu- cells and adaptive immunity in frontotemporal dementia with tau
rol Clin. 2017;35:339–74. pathology. Brain. 2021;144:724–45.
80. Boxer AL, Qureshi I, Ahlijanian M, et al. Safety of the tau- 97. Lui H, Zhang J, Makinson SR, et al. Progranulin deficiency pro-
directed monoclonal antibody BIIB092 in progressive supranu- motes circuit-specific synaptic pruning by microglia via comple-
clear palsy: a randomised, placebo-controlled, multiple ascend- ment activation. Cell. 2016;165:921–35.
ing dose phase 1b trial. Lancet Neurol. 2019;18:549–58. 98. Boland S, Swarup S, Ambaw YA, et al. Deficiency of the fron-
81. Dam T, Boxer AL, Golbe LI, et al. Safety and efficacy of totemporal dementia gene GRN results in gangliosidosis. Nat
anti-tau monoclonal antibody gosuranemab in progressive Commun. 2022;13:5924.
13
New Approaches to the Treatment of Frontotemporal Dementia 1065
99. Kelliny S, Xiong J, Bobrovskaya L, Zhou XF. Preclinical vali- 104. Pytel V, Cabrera-Martin MN, Delgado-Alvarez A, et al. Person-
dation of a novel oral Edaravone formulation for treatment of alized repetitive transcranial magnetic stimulation for primary
frontotemporal dementia. Neurotox Res. 2021;39:1689–707. progressive aphasia. J Alzheimers Dis. 2021;84:151–67.
100. Coemans S, Struys E, Vandenborre D, et al. A systematic review 105. Benussi A, Dell’Era V, Cosseddu M, et al. Transcranial stimula-
of transcranial direct current stimulation in primary progressive tion in frontotemporal dementia: a randomized, double-blind,
aphasia: methodological considerations. Front Aging Neurosci. sham-controlled trial. Alzheimers Dement (N Y). 2020;6:e12033.
2021;13:710818.
101. Nissim NR, Moberg PJ, Hamilton RH. Efficacy of noninvasive Publisher's Note Springer Nature remains neutral with regard to
brain stimulation (tDCS or TMS) paired with language therapy jurisdictional claims in published maps and institutional affiliations.
in the treatment of primary progressive aphasia: an exploratory
meta-analysis. Brain Sci. 2020;10. Springer Nature or its licensor (e.g. a society or other partner) holds
102. McConathey EM, White NC, Gervits F, et al. Baseline performance exclusive rights to this article under a publishing agreement with the
predicts tDCS-mediated improvements in language symptoms in author(s) or other rightsholder(s); author self-archiving of the accepted
primary progressive aphasia. Front Hum Neurosci. 2017;11:347. manuscript version of this article is solely governed by the terms of
103. Gervits F, Ash S, Coslett HB, et al. Transcranial direct current such publishing agreement and applicable law.
stimulation for the treatment of primary progressive aphasia: an
open-label pilot study. Brain Lang. 2016;162:35–41.
13