Sie sind auf Seite 1von 10

Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449

Review Article

Neuropsychiatric signs and symptoms of Alzheimer’s disease:


New treatment paradigms
Krista L. Lanct^ota,*, Joan Amatniekb, Sonia Ancoli-Israelc,d, Steven E. Arnolde, Clive Ballardf,g,
Jiska Cohen-Mansfieldh, Zahinoor Ismaili, Constantine Lyketsosj, David S. Millerk, Erik Musiekl,
Ricardo S. Osoriom, Paul B. Rosenbergn, Andrew Satlino, David Steffensp, Pierre Tariotq,
Lisa J. Bainr, Maria C. Carrillos, James A. Hendrixs, Heidi Jurgenss, Brendon Boott,u
a
Hurvitz Brain Sciences Program, Sunnybrook Research Institute and Departments of Psychiatry and Pharmacology, University of Toronto, Toronto, Canada
b
Otsuka Pharmaceutical Development & Commercialization, Inc., Princeton, NJ, USA
c
Department of Psychiatry, University of California, San Diego, CA, USA
d
Department of Medicine, University of California, San Diego, CA, USA
e
Interdisciplinary Brain Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
f
Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
g
University of Exeter, Exeter, UK
h
Department of Health Promotion, School of Public Health, Sackler Faculty of Medicine and Minerva Center for the Interdisciplinary Study of End of Life, Tel
Aviv University, Tel Aviv, Israel
i
Department of Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
j
Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medicine Institutes, Baltimore, MD, USA
k
Bracket Global, Wayne, PA, USA
l
Department of Neurology, Hope Center for Neurological Disorders, and Knight Alzheimer Disease Research Center, Washington University School of
Medicine, St. Louis, MO, USA
m
Center for Brain Health, NYU Langone Medical Center, New York, NY, USA
n
Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
o
Eisai, Inc., Woodcliff Lake, NJ, USA
p
Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT, USA
q
Banner Alzheimer’s Institute, Phoenix, AZ, USA
r
Elverson, PA, USA
s
Alzheimer’s Association, Chicago, IL, USA
t
Department of Neurology, Brigham and Women’s Hospital, Harvard University School of Medicine, Boston, MA, USA
u
Voyager Therapeutics, Cambridge, MA, USA

Abstract Neuropsychiatric symptoms (NPSs) are hallmarks of Alzheimer’s disease (AD), causing substan-
tial distress for both people with dementia and their caregivers, and contributing to early institution-
alization. They are among the earliest signs and symptoms of neurocognitive disorders and incipient
cognitive decline, yet are under-recognized and often challenging to treat. With this in mind, the Alz-
heimer’s Association convened a Research Roundtable in May 2016, bringing together experts from
academia, industry, and regulatory agencies to discuss the latest understanding of NPSs and review
the development of therapeutics and biomarkers of NPSs in AD. This review will explore the neuro-
biology of NPSs in AD and specific symptoms common in AD such as psychosis, agitation, apathy,
depression, and sleep disturbances. In addition, clinical trial designs for NPSs in AD and regulatory
considerations will be discussed.
Ó 2017 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an
open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/
4.0/).

*Corresponding author. Tel.: 11-4164806100; Fax: 11-4164806022.


E-mail address: krista.lanctot@sunnybrook.ca

http://dx.doi.org/10.1016/j.trci.2017.07.001
2352-8737/ Ó 2017 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449 441

Keywords: Alzheimer’s disease; Neuropsychiatric symptoms; Trial design; Delusions; Hallucinations; Agitation; Apathy;
Depression; Sleep disturbance

1. Introduction NPSs are mapped to domains such as memory, visuospatial,


executive function, or others.
Neuropsychiatric symptoms (NPSs) are hallmarks of Alz-
The Neuropsychiatric Inventory (NPI) [14] is commonly
heimer’s disease (AD), causing substantial distress for both
used to assess NPSs in clinical trials. A meta-analysis of studies
people with dementia and their caregivers, and contributing
using this scale found that apathy was the most common NPS,
to early institutionalization. They are among the earliest signs
followed by depression, aggression, anxiety, sleep distur-
and symptoms of neurocognitive disorders and incipient cogni-
bances, irritability, appetite disturbances, motor problems, de-
tive decline, yet are under-recognized and often challenging to
lusions, disinhibition, hallucinations, and euphoria [15]. The
treat. The Alzheimer’s Association’s Research Roundtable, a NPI has some limitations: it multiplies severity and frequency
consortium of experts from academia, industry, and regulatory
scores resulting in noncontinuous and non-normal total scores
agencies first addressed these challenges in 2010 [1].
[16], it fails to capture ruminative, repetitive, compulsive, and
Since then, there is renewed interest in mechanisms of
somatoform behaviors, and is dependent on caregiver report.
NPSs and identification of effective treatments for these
The Clinician Rating of the NPI (NPI-C) expands the range
symptoms, in part through the efforts of the NPS professional
of the symptoms assessed by the NPI [17].
interest area within the International Society to Advance
NPSs can present in advance of dementia. Among
Alzheimer’s Research and Treatment [2]. In May 2016, the
cognitively normal individuals, symptoms of depression,
Research Roundtable revisited the topic, bringing together irritability, agitation, and apathy predict more rapid cogni-
experts in the field to advance therapy development through
tive decline [18–20]; some NPSs are more powerful
a more comprehensive understanding of underlying mecha-
predictors of incident mild cognitive impairment (MCI)
nisms and application of novel clinical trial approaches.
than hippocampal atrophy [21]. One MCI study found
NPSs in 59% of participants and were associated with poorer
2. History and overview
cognitive and psychosocial function [22]. Their presence
NPSs affect almost all individuals with dementia (97%) predicted faster progression from MCI to dementia in two
over the course of the disease [3], and although they fluctuate very large cohort studies [9,23].
[4], they rarely disappear [5]. The effects on both patients NPSs typically emerge in three phases: (1) irritability,
and caregivers are severe: they are associated with impair- depression, and nighttime behavior changes; (2) anxiety,
ment in activities of daily living [6], poor quality of life appetite changes, agitation, and apathy; and (3) elation, motor
[7], earlier institutionalization [8], accelerated disease disturbances, hallucinations, delusions, and disinhibition [24].
progression, increased mortality [9], caregiver stress [10], This emergence of NPSs in later life is core to the construct of
and increased cost of care [11]. mild behavioral impairment (MBI), which describes later life
Medical, environmental, and caregiver issues can impact acquired, sustained, and impactful NPS as an “at-risk” state
the expression of NPSs, making the categorization of the for cognitive decline and dementia. Operationalized in
symptoms challenging [12]. Each of these approaches has the recently published Alzheimer’s Association International
limitations because nosology does not always reflect patho- Society to Advance Alzheimer’s Research and Treatment
physiology. The top-down classification system of the Diag- (ISTAART) MBI criteria [25], NPSs can emerge in advance
nostic and Statistical Manual, 5th edition, provides various of, or in concert with MCI, must be of at least 6-month dura-
syndrome categories, yet NPSs overlap substantially among tion, and not better accounted for by conventional psychiatric
these syndromes. For example, agitation may accompany nosology. MBI symptoms are categorized into five domains:
depression, anxiety, or psychosis. The Research Domain decreased drive and motivation, affective and emotional
Criteria project classifies NPSs based on neurobiological di- dysregulation, impulsivity, social inappropriateness, and
mensions and behaviors rather than clinical syndromes, abnormal perceptions or thoughts (e.g., delusions and halluci-
grouping them into five domains: (1) negative valence; (2) nations). Many rating scales used in dementia do not differen-
positive valence; (3) cognitive systems; (4) processes for so- tiate between new symptoms and chronic psychiatric illness,
cial systems; and (5) arousal or regulatory systems [13]. Yet and they have short reference times that may not allow time
here, too, there is tremendous overlap. For example, impair- to rule out reactive states. The MBI checklist was developed
ment of cognitive systems may manifest in delusions, hallu- to remedy these issues [26] (available at www.MBItest.org).
cinations, agitation, aggression, depression or dysphoria, The MBI criteria and MBI checklist will provide a screening
anxiety, elation or euphoria, apathy, disinhibition, irritabil- method to capture neurodegenerative disease early, foster sys-
ity, motor disturbance, sleep disorder, appetite disorder, tematic research into the effect or predictive value of NPSs on
aberrant vocalization, and ruminative, repetitive, and soma- cognitive and functional outcomes, and improve clinical trial
toform behaviors. Extensive overlap is also evident when enrollment and design [25,27].
442 K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449

Although the presence of NPSs in the early stages of symptoms. They occurred two to six times per week, per-
dementia predict disease progression, it is not clear whether sisted for 12 weeks among 32% and recurred in 50% within
NPSs reflect a more aggressive form of disease [28]. In 12 months [43]. They were associated with accelerated
addition, there are few pharmacologic treatment options cognitive and functional decline and increased mortality
with proven efficacy and acceptable levels of risk that target [44]. These frequencies were higher still among patients
NPSs. Only one medication is approved in AD for treatment with dementia with Lewy bodies. Hallucinations occur less
of NPSs (Canada and Europe only): risperidone for frequently than delusions in AD and rarely occur in isola-
short-term treatment of aggression. Nonpharmacologic inter- tion; the opposite is true for dementia with Lewy bodies
ventions are considered by most clinicians to be first-line treat- and Parkinson’s disease dementia. The hallucinations and
ments, yet translation from clinical studies to the real world delusions in AD are phenomenologically different to those
has been limited [29], in part because these interventions are in schizophrenia, psychotic depression, or mania. Hallucina-
not reimbursed, and caregivers are not trained to provide tions in AD are usually visual, less commonly auditory, and
them. They are generally based on psychosocial models for rarely tactile or olfactory [45]. Persecutory delusions occur
understanding these conditions across the continuum of the earlier in AD than misidentification delusions; both kinds in-
disease and in the presence of comorbid conditions [30–33]. crease with dementia severity.
Psychotic symptoms are correlated with increased
neocortical neurofibrillary tangles [46], increased levels of
3. The neurobiology of NPSs in AD
intraneuronal hyperphosphorylated tau [47], disruption of
Understanding the neurobiology of NPSs in the context of serotoninergic signaling [48], decreased neuronal density,
the AD brain could greatly advance the development of new and increased myoinositol/creatinine ratio assessed using
therapies. Most efforts to map the progression of AD pathol- proton magnetic resonance spectroscopy [49]. Individuals
ogy have focused on the cortex (e.g. [34]). For NPSs, however, with MCI who develop delusions also have greater gray-
distinct syndromes have different neurobiological underpin- matter atrophy, particularly in the right frontal areas that
nings, so understanding the dysfunction or dysregulation of regulate inhibitory control [50,51]. Some psychotic
subcortical forebrain, diencephalic, and brainstem nuclei presentations are associated with distinct neuroanatomic
that generate or mediate visceral, emotional, motivational, substrates. For example, misidentifications, which might
and other psychiatric symptoms will be required. Early be understood as a disconnect between memory and
studies recognized significant involvement of AD pathology familiarity, are associated with lower cell counts in the
in key components of the limbic system, including the amyg- hippocampal CA1 region. Persecutory delusions are
dala, basal forebrain, hypothalamus, and brainstem. Neurofi- associated with cell loss in the dorsal raphe nucleus [52].
brillary tangles, for example, are abundant in the amygdala of Genetics may play a role in AD psychosis: the serotonin
the Alzheimer’s brain [35], as well as in the basal nucleus of 2A receptor single-nucleotide polymorphism 102 T allele is
Meynert [36], the locus coeruleus [37], substantia nigra [38], associated with the presence of delusions, whereas the C
dorsal raphe nucleus [39], and hypothalamus [40]. Neuronal allele is protective [53]. The apolipoprotein E (APOE) 34
loss in the basal nucleus of Meynert and locus coeruleus allele is associated with an increased risk for AD, but the as-
also correlates with progression of AD dementia [41]. The hy- sociation between APOE 34 and NPSs in AD is weak [54].
pothalamus is important for the regulation of appetite, sleep, In recent years, atypical antipsychotics such as risperi-
and circadian rhythms, and mediates many aspects of done, olanzapine, quetiapine, and aripiprazole have largely
emotional experience. However, the degree to which patho- replaced conventional antipsychotics such as haloperidol
logic involvement of these structures correlates with NPSs for the treatment of psychosis in dementia. They have mini-
has not been well studied. The relevance of neuropathology mal efficacy on psychotic symptoms [55] but are associated
is fundamental to drug development, where currently with many adverse effects, such as somnolence, cognitive
approved treatments for mood and psychotic symptoms, decline, movement disorders, infections, edema, weight
such as antidepressants and antipsychotics, may not work gain, metabolic syndrome, and hypotension, which leads to
because of lack of target engagement in a degenerating brain. an increased risk of falls and stroke [55]. The major concern
is their association with an increased risk of death [56]. Alter-
native treatments, such the 5-HT2A receptor antagonists/in-
4. Neuropsychiatric symptoms
verse agonists and 5-HT6 receptor antagonists, are
This section will review the neurobiology, epidemiology, undergoing clinical evaluation in AD psychosis. Part of the
and current treatments for psychosis, agitation, apathy, challenge in the development of new therapies is that psycho-
depression, and sleep disturbances. sis in AD is difficult to define and clinically heterogeneous,
and there is a paucity of clinically relevant outcome measures.
An alternative approach conceptualizes dementia psy-
4.1. Psychotic symptoms in AD
chosis as the combined impact of memory loss with other
Both delusions and hallucinations are reported in AD personal factors, such as post-traumatic stress disorder
[42]. In a study of 124 demented patients, 67% had psychotic [57,58]. For example, a dementia sufferer might forget
K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449 443

where she placed her makeup and presume someone has tive, motor, and sensory abilities. These approaches educate
stolen it, but will change this perception when the makeup caregivers, make changes to the physical environment, increase
is found. The core symptom is memory loss and not a social engagement, exercise and activities, and address sleep
psychotic episode. Thus, treatments targeting memory problems. They produce significant positive impacts on NPSs
rather than psychosis would be more appropriate. Effective in randomized controlled trials [65,66]. The comprehensive
psychological interventions for functional psychosis might assessment includes assessing pain, discomfort, and other
also be adapted for people with AD [59]. medical conditions, as sometimes treating other medical
conditions may relieve agitation. For example, many patients
with dementia have pain, which may increase agitation and
4.2. Agitation in AD
anxiety [67]. Indeed, treatment of pain has been shown to alle-
Agitation occurs frequently in AD. The prevalence varies viate agitation in persons with dementia [68].
greatly between studies due to the use of different defini- A range of medications—antipsychotics, benzodiaze-
tions. The International Psychogeriatric Association pines, cholinesterase inhibitors, memantine, anticonvul-
consensus statement defines agitation as excessive motor ac- sants, and selective serotonin reuptake inhibitor (SSRI)
tivity, or verbal or physical aggression that associated with antidepressants—have been used to treat agitation in
emotional distress: (1) severe enough to produce disability; patients with dementia but have shown minimal efficacy
(2) beyond what would be expected from cognitive impair- and/or substantial adverse effects. In a recent study, the
ment by itself; and (3) not solely attributable to another dis- SSRI citalopram was shown to reduce agitation and care-
order, environmental conditions, or the physiological effects giver distress but was associated with cognitive decline
of a substance [60]. Agitation tends to persist; it increases as and corrected QT interval prolongation [69]. A combination
disease severity increases and is often associated with psy- of dextromethorphan hydrobromide and quinidine sulfate,
chosis, anxiety, and disinhibition. Agitation and psychosis which is approved for the treatment of pseudobulbar affect
together are predictive of more rapid decline, increased insti- in amyotrophic lateral sclerosis, is being investigated as a
tutionalization, and earlier death. treatment for agitation in AD [70]. Several other drug devel-
As recently reviewed, agitation in AD is associated with opment efforts targeting N-methyl-D-aspartate, dopamine
structural and functional abnormalities of the brain regions D2, and serotonin receptors (5-HT1A and 5-HT2A), and
associated with emotional regulation and salience: the cannabinoids are also being assessed.
frontal, anterior cingulate, and posterior cingulate cortices,
amygdala, and hippocampus [61]. Degeneration of these cir-
4.3. Apathy in AD
cuits may result in the overestimation of threat and/or
affective dysregulation that induces hypervigilance. Apathy, characterized by lack of motivation, decreased
Agitation and aggression are associated with decreased cholin- initiative, akinesia, and emotional indifference, is the most
ergic and serotoninergic markers, increased tau and phospho- common NPS associated with AD and a primary cause of
tau, and regional decreases in the N-acetylaspartate/creatine caregiver distress [71]. It is common in predementia states,
ratio and increases in the myoinositol/creatine ratio [49,62]. increases in frequency as the disease progresses, and predicts
Psychological theoretical frameworks provide alternative conversion from normal cognition to MCI and MCI to
models with which to understand agitation in dementia [63]. dementia [72]. An international task force published
The behavioral model focuses on triggers to the agitated diagnostic criteria for apathy in 2009, which require that
behavior and reinforcements, which maintain the behavior. two of three dimensions of diminished motivation must be
Interventions focus on eliminating the triggers and changing present for at least 4 weeks with identifiable associated func-
the relationship between behavior and reinforcement. The tional impairment [73]. The Apathy Evaluation Scale is
reduced stress threshold model posits that persons with commonly used to assess apathy across the AD continuum
dementia have a reduced threshold for stress, necessitating [72,74]. The NPI also includes an apathy subscale, but it
a very calm and quiet environment. The unmet needs model has not yet been validated for use on its own. Apathy can
describes how people with dementia are unable to address occur alone or as a symptom of depression [75].
their own needs or communicate them to others. These needs In neuroimaging studies of preclinical or prodromal AD,
are unmet because their caregivers are unaware of the needs apathy is associated with cortical dysfunction in the posterior
or of ways to meet them [64]. cingulate or inferior temporal cortex as well as atrophy,
Both nonpharmacologic and pharmacologic approaches are hypometabolism, and hypoperfusion in these regions.
used to manage agitation in patients with AD [12]. Abnormalities in cholinergic, GABAergic, and dopaminergic
Nonpharmacologic approaches are based on psychosocial par- function have also been associated with apathy, as well as
adigms for NPSs and address them based on the specific model high levels of tau and phospho-tau in the cerebrospinal fluid
used. However, several general principles apply, including a (CSF) [76]. Dopaminergic circuits have been targeted in
detailed assessment of potential unmet needs and environ- treatment trials using methylphenidate, with a significant
mental or interpersonal stressors. The intervention is tailored reduction in apathy symptoms, improvement in global
to the NPS, the person with dementia’s preferences and cogni- cognition, and minimal adverse events, for more than the
444 K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449

6-week study [77]. Interim results from another study also [88,89] and higher risk for developing dementia [90].
suggest that the combination of an acetylcholinesterase Cognitively normal people with biomarkers of AD have
inhibitor (donepezil) with a cholinergic precursor slightly decreased sleep efficiency [91]. A similar relation-
(choline alphoscerate) decreased apathy and caregiver ship is seen between the presence of sleep apnea and de-
distress [78]. Open label studies of cholinesterase inhibitors mentia: those with sleep apnea are more likely to
(donepezil, galantamine, and rivastigmine) showed improve- develop MCI and dementia [92,93] and those with more
ment in apathy for all three medications [58]. severe dementia have more severe sleep apnea [94]. In
addition, sleep apnea has been associated with a younger
4.4. Depression in AD age of decline to MCI or AD [95]. Circadian rhythm
disruption has also been linked to a greater risk of MCI
Depression is found in 16% in population-based AD and dementia [96], as well as neuroinflammation and syn-
studies, and 44.3% in hospital-based studies [79]. Depres- aptic loss in mice [97].
sion is also common in MCI. A meta-analysis of 57 studies The link between disrupted sleep and dementia may be
found an omnibus prevalence of 32%, but depressive symp- the accumulation of amyloid b (Ab) in the brain, according
toms more prevalent in clinical (40%) versus community- to studies in mice [98] and humans [99]. Ab dynamics in the
based (25%) samples [68] reflecting their clinical relevance brain and CSF are strongly influenced by the sleep-wake
in the cognitively impaired. Depression is also a predictor of cycle, and sleep deprivation accelerates Ab plaque
progression from normal cognition to MCI and from MCI to deposition in mice [68,69]. Orexin, a hypothalamic
dementia. However, whether depressive symptoms represent neurotransmitter implicated in sleep homeostasis, may
a risk factor or early manifestation of the AD brain disease or play a role in Ab regulation as genetic or pharmacologic
both is unclear. Supporting the latter notion, the presence of inhibition of orexin function suppresses Ab pathology in
MCI in depression has been shown to predict later develop- mice [100,101]. The glymphatic system—an astrocyte-
ment of AD [80]. driven system of perivascular fluid flow through which the
More severe neuropathology (tau, amyloid, and vascular brain removes toxic waste products—may also play an
disease) is seen in patients with AD with depression important role because slow-wave sleep (SWS) dramati-
compared to those without. These patients also show severe cally increases glymphatic influx and efflux [102]. SWS is
loss of serotonin receptors and serotonin transporter binding, also the stage of sleep where there is a greater reduction
which may have implications for treatment [81]. in corticocortical connectivity [103–107] and cerebral
Several studies have examined antidepressant efficacy in blood flow [108,109], indicating that it may also play a
the context of AD. The Depression in Alzheimer’s Disease role in decreasing Ab production (which is closely
Study (DIADS) demonstrated improvement with sertraline associated with neuronal activity [110]) as shown by two
compared with placebo in patients with AD with major recent human studies in which slow-wave activity was asso-
depression for more than 12 weeks [82]. A follow-up study, ciated with decreased CSF Ab40 and Ab42 levels in healthy
DIADS-2, included patients meeting criteria for “depression control subjects [111].
of AD,” but found no differences in depression outcomes for These data suggest that behavioral and pharmacologic
more than 12 weeks of treatment with sertraline or placebo interventions targeting SWS enhancement, sleep disorders,
[83]. There were no differences in outcome in a 13-week and circadian disturbances may improve cognitive func-
trial comparing sertraline, mirtazapine, or placebo [84]. tion or slow deterioration. These include both nonpharma-
A meta-analysis of SSRI and serotonin-norepinephrine reup- cologic approaches such as bright-light therapy [112,113]
take inhibitors (SRNI) treatment for depression in AD found and pharmacologic approaches using cholinesterase
“small to null”effect sizes [85], with small responses in those inhibitors, trazodone, melatonin or melatonin agonists,
with subsyndromal levels of depression. Standardized psy- and orexin antagonists. However, a recent meta-analysis
chosocial interventions were given to caregivers concomi- of drug treatment versus placebo for sleep disorders in pa-
tantly in both DIADS and DIADS-2. There is some tients with AD found insufficient evidence to help guide
evidence that psychosocial interventions such as these, as drug treatment of sleep problems in AD [114], and there
well as exercise, increasing social contact, reminiscent ther- is some uncertainty about the balance of benefits and risks
apy, and others can be efficacious (reviewed in [64,86]). For associated with these common treatments. Continuous
example, cognitive rehabilitation may decrease depression positive airway pressure for sleep apnea may also delay
among women with AD and mild dementia [87]. the progression of cognitive impairment in patients with
sleep apnea [95,115,116]. Given the growing evidence of
4.5. Sleep disturbances and AD
the link between sleep and dementia, and the high
A bidirectional relationship between sleep disturbances prevalence of sleep disorders in patients with AD,
and dementia has been demonstrated in both humans and objective sleep measures should be included in all AD
animals. For example, sleep problems and sleep disruption studies and NPS measures should be included in all
are associated with increased risk of cognitive decline sleep research in AD.
K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449 445

5. Design for NPS trials symptom-specific severity ratings. They may therefore differ
in detecting response to treatment, so researchers should
Only two drugs are indicated for NPSs in dementia: pima- select the scale that maximizes sensitivity in their population
vanserin for hallucinations and delusions associated with PD and indication.
psychosis (USA), and risperidone for short-term treatment Psychosocial interventions and concomitant medications,
of persistent aggression in moderate-to-severe AD (only in if introduced during a trial, increase variability and affect the
Canada and Europe). There are 16 ongoing trials, mostly placebo response. Ideally, both should be in place (and their
for agitation and aggression. This low productivity raises benefit assessed) before trials begin. Standardized psychoso-
several questions: are the drugs ineffective or the trials inap- cial interventions such as those used in DIADS [82],
propriately designed? Are syndromes phenomenologically DIADS-2 [83], and the Citalopram for agitation in
similar but neurobiologically diverse? Are diagnostic Alzheimer’s Disease (CitAD) study [119]) enhance recruit-
criteria and assessment tools for behavioral syndromes ment and retention, and minimize variability by standard-
capturing study populations that will enable detection of a izing procedures across sites.
positive signal? Do nonpharmacologic paradigms and treat-
ments show more promise than pharmacologic ones? Has an
6. Regulatory considerations
effective dose range been established and are sufficient dose
arms used? If titration is used, has enough time been allowed US Food and Drug Administration and Health Canada
to reach the optimal treatment effect? Which outcome mea- representatives at the Roundtable were asked whether com-
sures should be used? What is a clinically significant effect posite end points for NPSs in AD might be accepted. They
size? noted that composite end points are approved when validated
NPS treatment studies are particularly affected by large and when accepted as clinically meaningful by academics,
placebo effects. Sequential parallel comparison designs physicians, patients, and caregivers. Also, trials may be
[117] use a two-stage approach to mitigate these effects: required to use a primary end point and a coprimary global
placebo nonresponders from the first randomization are end point. To obtain approval for therapies to prevent the
rerandomized to receive active treatment or placebo. Studies emergence of symptoms, the key for regulators is that a
using historical data, run-in designs, or multiple clinical and meaningful outcome is measurable and can be interpreted.
biomarker variables might enroll more homogeneous popu- Pimavanserin was recently approved to treat hallucina-
lations and more accurately predict effect sizes. tions and delusions associated with psychosis in Parkinson’s
Several tools are available for assessing NPSs. Each has disease. The sponsor was granted breakthrough therapy
its pros and cons. The appropriate tool depends on the clin- designation by the Food and Drug Administration, allowing
ical condition and trial design. Informant-based measures approval based on a single small study rather than the usual
such as the NPI-Q, Behavioral Pathology in Alzheimer’s requirement for two pivotal studies. Would NPS therapies in
Disease Rating Scale (BEHAVE-AD), and Cohen- AD qualify for breakthrough designation? Regulators
Mansfield Agitation Inventory are low cost and clinically emphasized that breakthrough status and priority review
relevant but reflect the biases of the informant. Their validity are for potentially transformative therapies that address un-
depends on the extent of contact the informant has with the met medical needs.
person with dementia and caregiver depression and cogni-
tive status [118]. Of the informant-based tools, only the
7. Conclusions
NPI prescribes the minimum contact the caregiver must
have with the patient: 4 hours per day at least 4 days per There is renewed enthusiasm among industry, academic,
week, and knowledge of daytime and nighttime behaviors. regulatory, and patient and family advocacy groups to accel-
Sponsors set lower standards: all seven current trials have erate the development of new treatments for NPSs in AD. Ma-
less stringent NPI criteria. jor challenges for clinical trials include the need to (1) allocate
Clinician ratings such as with NPI-C are relatively free sufficient funds to develop nonpharmacologic interventions
from this “caregiver filter.” They have higher inter-rater reli- and implement them in routine clinical practice, these funds
ability but cost more. Video recordings are more objective are crucial as nonpharmacologic intervention studies gener-
but are not appropriate for low-frequency behaviors. They ally do not have a sponsor; (2) better define entry criteria
are intrusive, costly, and can have questionable clinical rele- (e.g., diagnosis and severity of patients and the presence of co-
vance. Wearable sensors are good for low-frequency behav- morbidities); (3) develop, validate, and standardize outcome
iors but may introduce noise and have questionable clinical measures that are clinically meaningful to patients and care-
relevance [33]. givers; (4) continue to seek biomarkers of NPSs; (5) stan-
The commonly used rating scales (e.g., BEHAVE-AD, dardize issues related to caregiver, informant involvement
Neurobehavioral Rating Scale, and NPI) characterize symp- and care setting (outpatient vs. nursing home) in clinical tri-
toms similarly, but they assess severity differently. For als; (6) incorporate designs that account for increased placebo
example, BEHAVE-AD uses an impact scale, NPI-Q as- responses; and (7) reach consensus on the use of psychosocial
sesses frequency and severity, and NPI-C produces interventions as background therapy in trials.
446 K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449

[10] Gonzalez-Salvador MT, Arango C, Lyketsos CG, Barba AC. The


RESEARCH IN CONTEXT stress and psychological morbidity of the Alzheimer patient care-
giver. Int J Geriatr Psychiatry 1999;14:701–10.
[11] Murman DL, Chen Q, Powell MC, Kuo SB, Bradley CJ, Colenda CC.
The incremental direct costs associated with behavioral symptoms in
1. Systematic Review: This review summarizes the pre- AD. Neurology 2002;59:1721–9.
sentations made at the May 2016 Research Roundta- [12] Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of
ble Meeting. Each presenter reviewed the literature behavioral and psychological symptoms of dementia. BMJ 2015;
of recent work of their specific topic area within 350:h369.
[13] Cuthbert BN. Research Domain Criteria: toward future psychiatric
the overall area of neuropsychiatric symptoms in nosologies. Dialogues Clin Neurosci 2015;17:89–97.
Alzheimer’s disease. [14] Cummings JL, Mega M, Gray K, Rosenberg-Thompson S,
Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehen-
2. Interpretation: The present article posits that new sive assessment of psychopathology in dementia. Neurology 1994;
treatments for neuropsychiatric symptoms in Alz- 44:2308–14.
heimer’s disease are needed and with sufficient [15] Zhao QF, Tan L, Wang HF, Jiang T, Tan MS, Tan L, et al. The prev-
research support can be developed to improve the alence of neuropsychiatric symptoms in Alzheimer’s disease: sys-
quality of life for people with dementia and their tematic review and meta-analysis. J Affect Disord 2016;190:264–71.
[16] Lai CK. The merits and problems of Neuropsychiatric Inventory as an
care partners. assessment tool in people with dementia and other neurological dis-
3. Future directions: Develop, validate, and standardize orders. Clin Interv Aging 2014;9:1051–61.
[17] de Medeiros K, Robert P, Gauthier S, Stella F, Politis A, Leoutsakos J,
outcome measures that are clinically meaningful to et al. The Neuropsychiatric Inventory-Clinician rating scale (NPI-C):
patients and caregivers and incorporate the use of reliability and validity of a revised assessment of neuropsychiatric
biomarkers to help enable the development of better symptoms in dementia. Int Psychogeriatr 2010;22:984–94.
treatments. [18] Donovan NJ, Amariglio RE, Zoller AS, Rudel RK, Gomez-Isla T,
Blacker D, et al. Subjective cognitive concerns and neuropsychiatric
predictors of progression to the early clinical stages of Alzheimer dis-
ease. Am J Geriatr Psychiatry 2014;22:1642–51.
[19] Duara R, Loewenstein DA, Greig MT, Potter E, Barker W, Raj A,
et al. Pre-MCI and MCI: neuropsychological, clinical, and imaging
features and progression rates. Am J Geriatr Psychiatry 2011;
References 19:951–60.
[20] Banks SJ, Raman R, He F, Salmon DP, Ferris S, Aisen P, et al. The
[1] Lyketsos CG, Carrillo MC, Ryan JM, Khachaturian AS, Trzepacz P, Alzheimer’s disease cooperative study prevention instrument project:
Amatniek J, et al. Neuropsychiatric symptoms in Alzheimer’s dis- longitudinal outcome of behavioral measures as predictors of cogni-
ease. Alzheimers Dement 2011;7:532–9. tive decline. Dement Geriatr Cogn Dis Extra 2014;4:509–16.
[2] Geda YE, Schneider LS, Gitlin LN, Miller DS, Smith GS, Bell J, [21] Geda YE, Roberts RO, Mielke MM, Knopman DS, Christianson TJ,
et al. Neuropsychiatric symptoms in Alzheimer’s disease: past Pankratz VS, et al. Baseline neuropsychiatric symptoms and the risk
progress and anticipation of the future. Alzheimers Dement of incident mild cognitive impairment: a population-based study. Am
2013;9:602–8. J Psychiatry 2014;171:572–81.
[3] Steinberg M, Shao H, Zandi P, Lyketsos CG, Welsh-Bohmer KA, [22] Feldman H, Scheltens P, Scarpini E, Hermann N, Mesenbrink P,
Norton MC, et al. Point and 5-year period prevalence of neuropsychi- Mancione L, et al. Behavioral symptoms in mild cognitive impair-
atric symptoms in dementia: the Cache County Study. Int J Geriatr ment. Neurology 2004;62:1199–201.
Psychiatry 2008;23:170–7. [23] Rosenberg PB, Mielke MM, Appleby BS, Oh ES, Geda YE,
[4] Tschanz JT, Corcoran CD, Schwartz S, Treiber K, Green RC, Lyketsos CG. The association of neuropsychiatric symptoms in
Norton MC, et al. Progression of cognitive, functional, and neuropsy- MCI with incident dementia and Alzheimer disease. Am J Geriatr
chiatric symptom domains in a population cohort with Alzheimer de- Psychiatry 2013;21:685–95.
mentia: the Cache County Dementia Progression Study. Am J Geriatr [24] Masters MC, Morris JC, Roe CM. “Noncognitive” symptoms of early
Psychiatry 2011;19:532–42. Alzheimer disease: a longitudinal analysis. Neurology 2015;84:1–6.
[5] Steinberg M, Tschanz JT, Corcoran C, Steffens DC, Norton MC, [25] Ismail Z, Smith EE, Geda Y, Sultzer D, Brodaty H, Smith G, et al.
Lyketsos CG, et al. The persistence of neuropsychiatric symptoms Neuropsychiatric symptoms as early manifestations of emergent de-
in dementia: the Cache County Study. Int J Geriatr Psychiatry mentia: provisional diagnostic criteria for mild behavioral impair-
2004;19:19–26. ment. Alzheimers Dement 2016;12:195–202.
[6] Lyketsos CG, Steele C, Baker L, Galik E, Kopunek S, Steinberg M, [26] Ismail Z, Aguera-Ortiz L, Brodaty H, Cieslak A, Cummings J,
et al. Major and minor depression in Alzheimer’s disease: prevalence Fischer CE, et al. The Mild Behavioral Impairment Checklist
and impact. J Neuropsychiatry Clin Neurosci 1997;9:556–61. (MBI-C): a rating scale for neuropsychiatric symptoms in pre-
[7] Gonzalez-Salvador T, Lyketsos CG, Baker A, Hovanec L, Roques C, dementia populations. J Alzheimers Dis 2017;56:929–38.
Brandt J, et al. Quality of life in dementia patients in long-term care. [27] Mortby ME, Black SE, Gauthier S, Miller DS, Porsteinsson A,
Int J Geriatr Psychiatry 2000;15:181–9. Smith EE, et al. Mild behavioral impairment: implications for clinical
[8] Steele C, Rovner B, Chase GA, Folstein M. Psychiatric symptoms trials. Int Psychogeriatr 2017. submitted.
and nursing home placement of patients with Alzheimer’s disease. [28] Rabins PV, Schwartz S, Black BS, Corcoran C, Fauth E, Mielke M,
Am J Psychiatry 1990;147:1049–51. et al. Predictors of progression to severe Alzheimer’s disease in an
[9] Peters ME, Schwartz S, Han D, Rabins PV, Steinberg M, Tschanz JT, incidence sample. Alzheimers Dement 2013;9:204–7.
et al. Neuropsychiatric symptoms as predictors of progression to se- [29] Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological in-
vere Alzheimer’s dementia and death: the Cache County Dementia terventions for neuropsychiatric symptoms of dementia. Am J Psy-
Progression Study. Am J Psychiatry 2015;172:460–5. chiatry 2012;169:946–53.
K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449 447

[30] Cohen-Mansfield J. Assessment of disruptive behavior/agitation in [51] Ismail Z, Nguyen MQ, Fischer CE, Schweizer TA, Mulsant BH. Neu-
the elderly: function, methods, and difficulties. J Geriatr Psychiatry roimaging of delusions in Alzheimer’s disease. Psychiatry Res 2012;
Neurol 1995;8:52–60. 202:89–95.
[31] Cohen-Mansfield J. Measurement of inappropriate behavior associ- [52] Forstl H, Burns A, Levy R, Cairns N. Neuropathological correlates of
ated with dementia. J Gerontol Nurs 1999;25:42–51. psychotic phenomena in confirmed Alzheimer’s disease. Br J Psychi-
[32] Cohen-Mansfield J, Golander H. The measurement of psychosis in atry 1994;165:53–9.
dementia: a comparison of assessment tools. Alzheimer Dis Assoc [53] Ismail Z, Nguyen MQ, Fischer CE, Schweizer TA, Mulsant BH,
Disord 2011;25:101–8. Mamo D. Neurobiology of delusions in Alzheimer’s disease. Curr
[33] Cohen-Mansfield J, Martin LS. Assessment of agitation in older Psychiatry Rep 2011;13:211–8.
adults. In: Lichtenberg PA, ed. Handbook of assessment in clinical [54] Lyketsos CG, Baker L, Warren A, Steele C, Brandt J, Steinberg M,
gerontology. London: Elsevier; 2010. p. 381–404. et al. Depression, delusions, and hallucinations in Alzheimer’s dis-
[34] Arnold SE, Hyman BT, Flory J, Damasio AR, Van Hoesen GW. The ease: no relationship to apolipoprotein E genotype. J Neuropsychi-
topographical and neuroanatomical distribution of neurofibrillary atry Clin Neurosci 1997;9:64–7.
tangles and neuritic plaques in the cerebral cortex of patients with [55] Schneider LS, Dagerman K, Insel PS. Efficacy and adverse effects
Alzheimer’s disease. Cereb Cortex 1991;1:103–16. of atypical antipsychotics for dementia: meta-analysis of random-
[35] Kromer Vogt LJ, Hyman BT, Van Hoesen GW, Damasio AR. Patho- ized, placebo-controlled trials. Am J Geriatr Psychiatry 2006;
logical alterations in the amygdala in Alzheimer’s disease. Neurosci- 14:191–210.
ence 1990;37:377–85. [56] Maust DT, Kim HM, Seyfried LS, Chiang C, Kavanagh J,
[36] Sassin I, Schultz C, Thal DR, Rub U, Arai K, Braak E, et al. Evolution Schneider LS, et al. Antipsychotics, other psychotropics, and the
of Alzheimer’s disease-related cytoskeletal changes in the basal nu- risk of death in patients with dementia: number needed to harm.
cleus of Meynert. Acta Neuropathol 2000;100:259–69. JAMA Psychiatry 2015;72:438–45.
[37] Grudzien A, Shaw P, Weintraub S, Bigio E, Mash DC, Mesulam MM. [57] Cohen-Mansfield J, Golander H, Ben-Israel J, Garfinkel D. The
Locus coeruleus neurofibrillary degeneration in aging, mild cognitive meanings of delusions in dementia: a preliminary study. Psychiatry
impairment and early Alzheimer’s disease. Neurobiol Aging 2007; Res 2011;189:97–104.
28:327–35. [58] Cohen-Mansfield J, Golander H, Cohen R. Rethinking psychosis in
[38] Schneider JA, Li JL, Li Y, Wilson RS, Kordower JH, Bennett DA. dementia: an analysis of antecedents and explanations. Am J Alz-
Substantia nigra tangles are related to gait impairment in older per- heimers Dis Other Demen 2017;32:265–71.
sons. Ann Neurol 2006;59:166–73. [59] Ballard C, Orrell M, YongZhong S, Moniz-Cook E, Stafford J,
[39] Simic G, Babic Leko M, Wray S, Harrington CR, Delalle I, Jovanov- Whittaker R, et al. Impact of antipsychotic review and nonpharmaco-
Milosevic N, et al. Monoaminergic neuropathology in Alzheimer’s logical intervention on antipsychotic use, neuropsychiatric symp-
disease. Prog Neurobiol 2016;151:101–38. toms, and mortality in people with dementia living in nursing
[40] Byne W, Mattiace L, Kress Y, Davies P. Alz-50 immunoreactivity in homes: a Factorial Cluster-Randomized Controlled Trial by the
the hypothalamus of the normal and Alzheimer human and the rat. J Well-Being and Health for People With Dementia (WHELD) Pro-
Comp Neurol 1991;306:602–12. gram. Am J Psychiatry 2016;173:252–62.
[41] Arendt T, Bruckner MK, Morawski M, Jager C, Gertz HJ. Early neu- [60] Cummings J, Mintzer J, Brodaty H, Sano M, Banerjee S,
rone loss in Alzheimer’s disease: cortical or subcortical? Acta Neuro- Devanand DP, et al. Agitation in cognitive disorders: International
pathol Commun 2015;3:10. Psychogeriatric Association provisional consensus clinical and
[42] Jeste DV, Finkel SI. Psychosis of Alzheimer’s disease and related de- research definition. Int Psychogeriatr 2015;27:7–17.
mentias. Diagnostic criteria for a distinct syndrome. Am J Geriatr [61] Rosenberg PB, Nowrangi MA, Lyketsos CG. Neuropsychiatric
Psychiatry 2000;8:29–34. symptoms in Alzheimer’s disease: what might be associated brain cir-
[43] Ballard C, Bannister C, Graham C, Oyebode F, Wilcock G. Associa- cuits? Mol Aspects Med 2015;43–44:25–37.
tions of psychotic symptoms in dementia sufferers. Br J Psychiatry [62] Tsai CF, Hung CW, Lirng JF, Wang SJ, Fuh JL. Differences in brain
1995;167:537–40. metabolism associated with agitation and depression in Alzheimer’s
[44] Ballard CG, O’Brien JT, Coope B, Wilcock G. Psychotic symptoms disease. East Asian Arch Psychiatry 2013;23:86–90.
in dementia and the rate of cognitive decline. J Am Geriatr Soc 1997; [63] Cohen-Mansfield J. Theoretical frameworks for behavioral problems
45:1031–2. in dementia. Alzheimers Care Q 2000;1:18–21.
[45] Leroi I, Voulgari A, Breitner JC, Lyketsos CG. The epidemiology of [64] Cohen-Mansfield J. Behavioral and psychological symptoms of de-
psychosis in dementia. Am J Geriatr Psychiatry 2003;11:83–91. mentia. In: Washington, DC, US: American Psychological Associa-
[46] Farber NB, Rubin EH, Newcomer JW, Kinscherf DA, Miller JP, tion; 2015. p. 271–317.
Morris JC, et al. Increased neocortical neurofibrillary tangle density [65] Cohen-Mansfield J, Thein K, Marx MS, Dakheel-Ali M, Freedman L.
in subjects with Alzheimer disease and psychosis. Arch Gen Psychi- Efficacy of nonpharmacologic interventions for agitation in advanced
atry 2000;57:1165–73. dementia: a randomized, placebo-controlled trial. J Clin Psychiatry
[47] Murray PS, Kirkwood CM, Gray MC, Fish KN, Ikonomovic MD, 2012;73:1255–61.
Hamilton RL, et al. Hyperphosphorylated tau is elevated in Alz- [66] Cohen-Mansfield J. Nonpharmacologic treatment of behavioral dis-
heimer’s disease with psychosis. J Alzheimers Dis 2014;39:759–73. orders in dementia. Curr Treat Options Neurol 2013;15:765–85.
[48] Zubenko GS, Moossy J, Martinez AJ, Rao G, Claassen D, Rosen J, [67] Sampson EL, White N, Lord K, Leurent B, Vickerstaff V, Scott S,
et al. Neuropathologic and neurochemical correlates of psychosis et al. Pain, agitation, and behavioural problems in people with de-
in primary dementia. Arch Neurol 1991;48:619–24. mentia admitted to general hospital wards: a longitudinal cohort
[49] Shinno H, Inagaki T, Miyaoka T, Okazaki S, Kawamukai T, Utani E, study. Pain 2015;156:675–83.
et al. A decrease in N-acetylaspartate and an increase in myoinositol [68] Husebo BS, Ballard C, Sandvik R, Nilsen OB, Aarsland D. Efficacy
in the anterior cingulate gyrus are associated with behavioral and psy- of treating pain to reduce behavioural disturbances in residents of
chological symptoms in Alzheimer’s disease. J Neurol Sci 2007; nursing homes with dementia: cluster randomised clinical trial.
260:132–8. BMJ 2011;343:d4065.
[50] Ting WK, Fischer CE, Millikin CP, Ismail Z, Chow TW, [69] Porsteinsson AP, Drye LT, Pollock BG, Devanand DP, Frangakis C,
Schweizer TA. Grey matter atrophy in mild cognitive impairment/ Ismail Z, et al. Effect of citalopram on agitation in Alzheimer
early Alzheimer disease associated with delusions: a voxel-based disease: the CitAD randomized clinical trial. JAMA 2014;
morphometry study. Curr Alzheimer Res 2015;12:165–72. 311:682–91.
448 K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449

[70] Garay RP, Grossberg GT. AVP-786 for the treatment of agitation in [89] Miyata S, Noda A, Iwamoto K, Kawano N, Okuda M, Ozaki N. Poor
dementia of the Alzheimer’s type. Expert Opin Investig Drugs sleep quality impairs cognitive performance in older adults. J Sleep
2017;26:121–32. Res 2013;22:535–41.
[71] Thomas P, Clement JP, Hazif-Thomas C, Leger JM. Family, Alz- [90] Benedict C, Byberg L, Cedernaes J, Hogenkamp PS, Giedratis V,
heimer’s disease and negative symptoms. Int J Geriatr Psychiatry Kilander L, et al. Self-reported sleep disturbance is associated with Alz-
2001;16:192–202. heimer’s disease risk in men. Alzheimers Dement 2015;11:1090–7.
[72] Guercio BJ, Donovan NJ, Munro CE, Aghjayan SL, Wigman SE, [91] Ju YE, McLeland JS, Toedebusch CD, Xiong C, Fagan AM,
Locascio JJ, et al. The Apathy Evaluation Scale: a comparison of sub- Duntley SP, et al. Sleep quality and preclinical Alzheimer disease.
ject, informant, and clinician report in cognitively normal elderly and JAMA Neurol 2013;70:587–93.
mild cognitive impairment. J Alzheimers Dis 2015;47:421–32. [92] Spira AP, Blackwell T, Stone KL, Redline S, Cauley JA, Ancoli-
[73] Robert P, Onyike CU, Leentjens AF, Dujardin K, Aalten P, Israel S, et al. Sleep-disordered breathing and cognition in older
Starkstein S, et al. Proposed diagnostic criteria for apathy in Alz- women. J Am Geriatr Soc 2008;56:45–50.
heimer’s disease and other neuropsychiatric disorders. Eur Psychiatry [93] Yaffe K, Laffan AM, Harrison SL, Redline S, Spira AP, Ensrud KE,
2009;24:98–104. et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive
[74] Marin RS, Biedrzycki RC, Firinciogullari S. Reliability and validity impairment and dementia in older women. JAMA 2011;306:613–9.
of the Apathy Evaluation Scale. Psychiatry Res 1991;38:143–62. [94] Ancoli-Israel S, Klauber MR, Butters N, Parker L, Kripke DF. De-
[75] Benoit M, Berrut G, Doussaint J, Bakchine S, Bonin-Guillaume S, mentia in institutionalized elderly: relation to sleep apnea. J Am Ger-
Fremont P, et al. Apathy and depression in mild Alzheimer’s disease: iatr Soc 1991;39:258–63.
a cross-sectional study using diagnostic criteria. J Alzheimers Dis [95] Osorio RS, Gumb T, Pirraglia E, Varga AW, Lu SE, Lim J, et al.
2012;31:325–34. Sleep-disordered breathing advances cognitive decline in the elderly.
[76] Lanctot KL, Aguera-Ortiz L, Brodaty H, Francis PT, Geda YE, Neurology 2015;84:1964–71.
Ismail Z, et al. Apathy associated with neurocognitive disorders: [96] Tranah GJ, Blackwell T, Stone KL, Ancoli-Israel S, Paudel ML,
recent progress and future directions. Alzheimers Dement 2017; Ensrud KE, et al. Circadian activity rhythms and risk of incident de-
13:84–100. mentia and mild cognitive impairment in older women. Ann Neurol
[77] Rosenberg PB, Lanctot KL, Drye LT, Herrmann N, Scherer RW, 2011;70:722–32.
Bachman DL, et al. Safety and efficacy of methylphenidate for [97] Musiek ES, Lim MM, Yang G, Bauer AQ, Qi L, Lee Y, et al. Circa-
apathy in Alzheimer’s disease: a randomized, placebo-controlled dian clock proteins regulate neuronal redox homeostasis and neuro-
trial. J Clin Psychiatry 2013;74:810–6. degeneration. J Clin Invest 2013;123:5389–400.
[78] Rea R, Carotenuto A, Traini E, Fasanaro AM, Manzo V, Amenta F. [98] Kang JE, Lim MM, Bateman RJ, Lee JJ, Smyth LP, Cirrito JR, et al.
Apathy treatment in Alzheimer’s disease: interim results of the AS- Amyloid-beta dynamics are regulated by orexin and the sleep-wake
COMALVA Trial. J Alzheimers Dis 2015;48:377–83. cycle. Science 2009;326:1005–7.
[79] Panza F, Frisardi V, Capurso C, D’Introno A, Colacicco AM, [99] Huang Y, Potter R, Sigurdson W, Kasten T, Connors R, Morris JC,
Imbimbo BP, et al. Late-life depression, mild cognitive impairment, et al. beta-Amyloid dynamics in human plasma. Arch Neurol 2012;
and dementia: possible continuum? Am J Geriatr Psychiatry 2010; 69:1591–7.
18:98–116. [100] Roh JH, Jiang H, Finn MB, Stewart FR, Mahan TE, Cirrito JR, et al.
[80] Steffens DC, McQuoid DR, Potter GG. Amnestic mild cognitive Potential role of orexin and sleep modulation in the pathogenesis of
impairment and incident dementia and Alzheimer’s disease in geri- Alzheimer’s disease. J Exp Med 2014;211:2487–96.
atric depression. Int Psychogeriatr 2014;26:2029–36. [101] Liguori C, Romigi A, Nuccetelli M, Zannino S, Sancesario G,
[81] Holmes C, Arranz M, Collier D, Powell J, Lovestone S. Depression in Martorana A, et al. Orexinergic system dysregulation, sleep impair-
Alzheimer’s disease: the effect of serotonin receptor gene variation. ment, and cognitive decline in Alzheimer disease. JAMA Neurol
Am J Med Genet B Neuropsychiatr Genet 2003;119B:40–3. 2014;71:1498–505.
[82] Lyketsos CG, Del Campo L, Steinberg M, Miles Q, Steele CD, [102] Xie L, Kang H, Xu Q, Chen MJ, Liao Y, Thiyagarajan M, et al. Sleep
Munro C, et al. Treating depression in Alzheimer disease: efficacy drives metabolite clearance from the adult brain. Science 2013;
and safety of sertraline therapy, and the benefits of depression reduc- 342:373–7.
tion: the DIADS. Arch Gen Psychiatry 2003;60:737–46. [103] Horovitz SG, Braun AR, Carr WS, Picchioni D, Balkin TJ,
[83] Rosenberg PB, Drye LT, Martin BK, Frangakis C, Mintzer JE, Fukunaga M, et al. Decoupling of the brain’s default mode network
Weintraub D, et al. Sertraline for the treatment of depression in Alz- during deep sleep. Proc Natl Acad Sci U S A 2009;106:11376–81.
heimer disease. Am J Geriatr Psychiatry 2010;18:136–45. [104] Larson-Prior LJ, Power JD, Vincent JL, Nolan TS, Coalson RS,
[84] Banerjee S, Hellier J, Dewey M, Romeo R, Ballard C, Baldwin R, Zempel J, et al. Modulation of the brain’s functional network archi-
et al. Sertraline or mirtazapine for depression in dementia (HTA- tecture in the transition from wake to sleep. Prog Brain Res 2011;
SADD): a randomised, multicentre, double-blind, placebo-controlled 193:277–94.
trial. Lancet 2011;378:403–11. [105] Samann PG, Wehrle R, Hoehn D, Spoormaker VI, Peters H, Tully C,
[85] Sepehry AA, Lee PE, Hsiung GY, Beattie BL, Jacova C. Effect of se- et al. Development of the brain’s default mode network from wake-
lective serotonin reuptake inhibitors in Alzheimer’s disease with co- fulness to slow wave sleep. Cereb Cortex 2011;21:2082–93.
morbid depression: a meta-analysis of depression and cognitive [106] Spoormaker VI, Schroter MS, Gleiser PM, Andrade KC, Dresler M,
outcomes. Drugs Aging 2012;29:793–806. Wehrle R, et al. Development of a large-scale functional brain
[86] Olazaran J, Reisberg B, Clare L, Cruz I, Pena-Casanova J, Del Ser T, network during human non-rapid eye movement sleep. J Neurosci
et al. Nonpharmacological therapies in Alzheimer’s disease: a sys- 2010;30:11379–87.
tematic review of efficacy. Dement Geriatr Cogn Disord 2010; [107] Wu CW, Liu PY, Tsai PJ, Wu YC, Hung CS, Tsai YC, et al. Variations
30:161–78. in connectivity in the sensorimotor and default-mode networks dur-
[87] Kurz A, Thone-Otto A, Cramer B, Egert S, Frolich L, Gertz HJ, et al. ing the first nocturnal sleep cycle. Brain Connect 2012;2:177–90.
CORDIAL: cognitive rehabilitation and cognitive-behavioral treat- [108] Braun AR, Balkin TJ, Wesenten NJ, Carson RE, Varga M, Baldwin P,
ment for early dementia in Alzheimer disease: a multicenter, random- et al. Regional cerebral blood flow throughout the sleep-wake cycle.
ized, controlled trial. Alzheimer Dis Assoc Disord 2012;26:246–53. An H2(15)O PET study. Brain 1997;120:1173–97.
[88] Sterniczuk R, Theou O, Rusak B, Rockwood K. Sleep disturbance is [109] Hofle N, Paus T, Reutens D, Fiset P, Gotman J, Evans AC, et al. Regional
associated with incident dementia and mortality. Curr Alzheimer Res cerebral blood flow changes as a function of delta and spindle activity
2013;10:767–75. during slow wave sleep in humans. J Neurosci 1997;17:4800–8.
K.L. Lanct^ot et al. / Alzheimer’s & Dementia: Translational Research & Clinical Interventions 3 (2017) 440-449 449

[110] Cirrito JR, Yamada KA, Finn MB, Sloviter RS, Bales KR, May PC, [115] Ancoli-Israel S, Palmer BW, Cooke JR, Corey-Bloom J,
et al. Synaptic activity regulates interstitial fluid amyloid-beta levels Fiorentino L, Natarajan L, et al. Cognitive effects of treating obstruc-
in vivo. Neuron 2005;48:913–22. tive sleep apnea in Alzheimer’s disease: a randomized controlled
[111] Ju YE, Finn MB, Sutphen CL, Herries EM, Jerome GM, study. J Am Geriatr Soc 2008;56:2076–81.
Ladenson JH, et al. Obstructive sleep apnea decreases central nervous [116] Cooke JR, Ancoli-Israel S, Liu L, Loredo JS, Natarajan L, Palmer BS,
system-derived proteins in the cerebrospinal fluid. Ann Neurol 2016; et al. Continuous positive airway pressure deepens sleep in patients
80:154–9. with Alzheimer’s disease and obstructive sleep apnea. Sleep Med
[112] McCurry SM, Pike KC, Vitiello MV, Logsdon RG, Larson EB, 2009;10:1101–6.
Teri L. Increasing walking and bright light exposure to improve sleep [117] Fava M, Evins AE, Dorer DJ, Schoenfeld DA. The problem of the
in community-dwelling persons with Alzheimer’s disease: results of placebo response in clinical trials for psychiatric disorders: culprits,
a randomized, controlled trial. J Am Geriatr Soc 2011;59:1393–402. possible remedies, and a novel study design approach. Psychother
[113] Ancoli-Israel S, Gehrman P, Martin JL, Shochat T, Marler M, Corey- Psychosom 2003;72:115–27.
Bloom J, et al. Increased light exposure consolidates sleep and [118] Schulz R, Cook TB, Beach SR, Lingler JH, Martire LM, Monin JK,
strengthens circadian rhythms in severe Alzheimer’s disease patients. et al. Magnitude and causes of bias among family caregivers rating
Behav Sleep Med 2003;1:22–36. Alzheimer disease patients. Am J Geriatr Psychiatry 2013;21:14–25.
[114] McCleery J, Cohen DA, Sharpley AL. Pharmacotherapies for sleep [119] Drye LT, Ismail Z, Porsteinsson AP, Rosenberg PB, Weintraub D,
disturbances in Alzheimer’s disease. Cochrane Database Syst Rev Marano C, et al. Citalopram for agitation in Alzheimer’s disease:
2014;CD009178. design and methods. Alzheimers Dement 2012;8:121–30.

Das könnte Ihnen auch gefallen