Apathy in The Setting of Alzheimer`s Disease and Related Disorders

advertisement

APPENDIX B

Apathy in the setting of Alzheimer’s disease and related disorders: overview and research recommendations

Lanctôt, K.L.

1,2

, Rosenberg, P.B.

3

, Sultzer, D.L.

4

, Francis, P.T.

5

, Brodaty, H.

6

, Padala, P.R.

7,8

,

Onyike, C.U.

3

, Agüera Ortiz, L.

9,10

, Geda, Y.E.

11 for the Neuropsychiatric Syndromes Professional Interest Area of ISTAART

1 Senior Scientist, Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON; 2 Departments of Psychiatry and

Pharmacology/Toxicology, University of Toronto

3 Division of Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University School of Medicine, Baltimore, MD

4 Department of Psychiatry and Biobehavioral Sciences, UCLA School of Medicine, Los Angeles, CA

5 Wolfson Centre for Age Related Diseases, King's College London, England

6 Dementia Collaborative Research Centre, University of New South Wales, Sydney, Australia

7 Geriatric Research Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR; 8

Department of Psychiatry, University of Arkansas for Medical Sciences, Little Rock, AR

9 Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM; 10 University Hospital Doce de Octubre, Madrid,

Spain

11 Departments of Psychiatry, Neurology and Health Sciences Research Mayo Clinic, Scottsdale, AZ

1

Abstract

An international panel of experts in apathy in Alzheimer’s disease (AD) was brought together to form a workgroup to outline a future research agenda for this neuropsychiatric symptom (NPS). The high prevalence and negative impact of apathy was acknowledged. While apathy is reasonably well-defined, the need for refinement in its definition and measurement, with particular attention to subdomains was recognized. Current knowledge includes neuroimaging research and studies focusing on neurobiological and neurochemical bases of apathy. A focus on brain-behavior relationships, interrelationships between

NPS, and evaluating the neurobiologic basis of the distinct components using multimodal imaging techniques was recommended. Animal models and genetic studies are lacking. Clinical trials that target apathy, taking advantage of advances in neuroimaging and biomarkers, and individually tailored nonpharmacological interventions are key. The ultimate goal is to alleviate apathy by identifying treatment targets and neurobiological factors that mediate successful treatment response and by well designed clinical trials.

2

Introduction

Apathy is a syndrome primarily characterized by a marked loss of motivation [1, 2]. It is common throughout the spectrum of cognitive disorders from mild cognitive impairment to severe

Alzheimer’s disease (AD), as well as in a variety of other neuropsychiatric disorders. In the last

20 years, considerable progress has been made describing the phenomenology and epidemiology, developing measurements, formulating diagnostic criteria, exploring neurophysiological substrates, and proposing and testing interventions for apathy. There is still much to learn about the boundaries of apathy and its etiology, so that effective pharmacologic and behavioral interventions can be developed.

In this paper the Neuropsychiatric Syndromes

Professional Interest Area (NPS-PIA) apathy workgroup (comprised of specialists and investigators from the fields of geriatric medicine, neuropsychiatry, behavioural neurology and neuropsychology) reviews progress to date and makes recommendations for future research.

Prevalence

The high prevalence of apathy has been well documented in both clinical and community samples, and throughout the spectrum of severity of neurodegenerative conditions. The frequency of apathy is generally higher in tertiary settings than in population-based samples due to referral bias [3];[4]. In AD, a one month prevalence of 72% among 50 outpatients with mild to severe AD has been reported based on the apathy subscale of the Neuropsychiatric inventory ( NPI ) [5]. Extending these findings to

Mild Cognitive Impairment ( MCI ), the same group reported the prevalence of apathy to be

51% in mild AD (n = 124), 39% in MCI (n = 28) and 2% in cognitively normal persons (n = 50)

[6]. Similarly, Copeland and colleagues [7] observed a 41% prevalence of ‘passivity’ in a clinic sample of elders suffering a mild cognitive disorder. In contrast, the Cardiovascular Heart

Study (CHS), found a 15% prevalence of NPI defined apathy among MCI subjects, and a prevalence of 35.8% in subjects with dementia

3

[8]. These figures contrasted to a prevalence of

3.2% among cognitively normal persons in the

Cache County Study [9]. The Mayo Clinic Study of Aging (MCSA) [10] measured NPIQ apathy in a countywide probability sample and reported prevalence rates of 18.5% in MCI and 4.8% among cognitively normal persons. The population-based Kungsholmen project and others have also reported that apathy is common in AD and may be a predictor of disease progression [11]. The association of apathy with cognitive progression among normal elders was also suggested by in findings in the Cache

County Study[12]. The prevalence of apathy in

AD and related conditions is customarily reported in crude frequency rates. However, these rates could be driven by demographic variables such as age [13]. Therefore, future studies should report by age, sex and other traditional confounder variables.

Impact

In addition to a high prevalence, research has documented the negative impact of apathy on both patients and caregivers. AD patients with apathy are 2.8 times more likely than nonapathetic patients to be impaired in one of the activities of daily living ( ADL s), and 3.2 times more likely to be impaired in all six activities

[14]. In another study of AD patients, apathy was the only behavioral problem that was significantly associated with both Instrumental

Activities of Daily Living (IADL) and ADL impairment. Apathy independently accounted for 27% of the variance in IADL [15].

Consistent with its association with decreased function, apathy has a negative impact on caregiver burden. Caregivers of patients with

AD complain more frequently of apathy than any other patient behavior and their distress correlates significantly with apathy [16]; [17]. In addition, caregivers of apathetic AD patients report significant levels of caregiver burden and fewer positive experiences with caregiving.

Apathy is also associated with lower cognitive and functional performance in normal elders and those with MCI [12].

Beyond the associations with decreased cognition and function, apathy is a marker for

poor prognosis. In MCI, apathy has consistently been linked to higher rates of conversion to dementia. For example, among 131 patients, those with both amnestic-MCI and apathy had an almost sevenfold greater risk of progressing to AD than amnestic-MCI patients without apathy (HR=6.9; 2.3-20.6). Furthermore, in another MCI group, the risk of developing AD increased 30% per point on the NPI apathy item

(HR=1.3; 1.1-1.4) [18]. In 1821 MCI patients followed for a median of 1.6 years, the risk of developing AD increased 16% per point on the

NPI apathy item (HR=1.16; 1.01-1.30) [19]. In those with AD, the presence of apathy has been associated with treatment response and the speed of progression. The presence of apathy was a predictor of poor behavioral response to donepezil in patients with AD [20]. In addition, apathy at baseline was associated with faster cognitive (p = 0.0007) and functional decline (p

= 0.006) in a cohort of 354 AD patients [21].

What remains to be seen is whether treatment of apathy can modify its association with poor prognosis.

Measurement of Apathy

The last two decades have witnessed growing interest in the investigation of apathy. This is in part fuelled by three measurement advances.

First a practical step was taken to operationalize the definition of apathy, and subsequently, measure it using a validated scale [1, 2]. Second, investigators developed additional scales to measure apathy and related neuropsychiatric symptoms in the context of dementia [22, 23].

Third, diagnostic criteria for apathy associated with primary dementia have been proposed.

Operationalization of the Definition . Apathy is associated with varied concepts including indifference, lack of motivation, and procrastination. The syndrome has cognitive, affective, and behavioral dimensions leading to difficulties in operationalization and lack of consistency of use of the term in the literature.

Further complicating assessment is the overlap of symptoms between apathy and other syndromes such as depression, anxiety and motor retardation. Marin’s model [24] defined apathy using three dimensions as a composite of reductions in emotional response, goal-directed cognitive activity, and purposeful behavior.

These dimensions are consistent with previous neurobiological models [25, 26] where three subtypes of disrupted processing are defined based on specific underlying lesions: emotionalaffective, cognitive, and auto-activation.

Measurement of Severity. Scales for apathy assessment include: the Apathy Evaluation Scale

(AES) [27] the Structured Clinical Interview for

Apathy (SCIA) [28], the Apathy Scale (AS)

[29], the Apathy Inventory (AI) [30], the

Dementia Apathy Interview and Rating (DAIR)

[31], and the Lille Apathy Rating Scale (LARS)

[32]. These assessments are largely based on

Marin’s model, which is focused on lack of motivation. The recently published APADEM scale [33] has shown good measurement properties in those with moderate to severe dementia, where ceiling effects are often problematic. Investigators from the American

Psychiatric Association and their collaborators examined the psychometric properties of 15 scales that measure apathy across various disorders including schizophrenia, depression, traumatic brain injury, stroke, Parkinson’s disease etc. They concluded that the apathy subscale of Neuropsychiatric inventory (NPI), and AES [27] are the two most psychometrically robust measures for assessing apathy across various disease conditions [34].

Diagnostic Criteria. In 2009, an international workgroup [35] proposed Consensus Diagnostic

Criteria for apathy using a contemporary concept, definition and construct (shown in

Table 1). These criteria 1) define apathy in terms of its core feature of diminished motivation, 2) introduce additional cognitive, affective and behavioral requirements, 3) assert a time criterion (symptoms must be present for at least 4 weeks) and 4) require demonstrable functional impairment attributable to apathy.

They also stipulate exclusion criteria for discounting entities that mimic apathy [36]. The criteria operationalize present-day concepts of apathy, denoting persistent declines in motivation, self-directed action and emotional vitality in a format that facilitates bedside ascertainment. By incorporating affective

4

indicators (i.e., diminished feelings and reactiveness) and noting persistency, the criteria extend earlier work [37]; [38] that highlighted cognitive and behavioral indicators. High reliability (Kappa, κ, = 0.93) was demonstrated in a study of 306 patients with dementia of the

Alzheimer type (DAT), mixed dementia, MCI,

Parkinson’s disease, schizophrenia and major depression. Reliability was solid for each criterion (κ = 0.9−1 for A, C and D, and 0.6-0.8 for B1−3)[39].

While progress has been made defining and measuring apathy, more refinement is needed.

In particular subdomains of apathy should be defined and modified as understanding improves, e.g. scales that sort symptoms into cognitive, behavioural, and emotional domains, with adequate validity and reliability for these domains. Adoption of diagnostic criteria facilitates communication and collaboration, as well as compilation and comparison of findings.

Thus promotion of the new diagnostic criteria is a high priority requiring replication of their validation in larger samples, and in diverse clinical populations and settings. It is also of vital importance to establish the sensitivity and specificity of these criteria, using for comparison diagnoses derived from specialist examination and diagnostic consensus, so as to determine if refinements are needed. In addition, the field might develop physiologic markers of apathy using neuroimaging, neurobiological, genetic and other (e.g., actigraphy [40]) methods as outlined below. These markers can be used to validate clinical diagnosis and serve as quantitative surrogate measures Ultimately, the usefulness of diagnostic criteria will be demonstrated by their ability to define an endophenotype that is linked to specific neurobiologic substrates and treatment response.

Neuroimaging

Structural and functional neuroimaging can reveal relationships between regional brain structure or function and the presence or extent of clinical apathy. Neuroimaging can illuminate such relationships in vivo, address state or trait expressions, and explore relevant relationships with specific brain physiologies in distinct brain regions.

Structural . Studies using MRI to measure regional cortical volume reported that apathy in

AD was associated with smaller volumes of the anterior cingulate gyrus, orbitofrontal cortex, or other regions in the frontal cortex or basal ganglia. In addition, apathy was associated with either presence of white matter lesions overall

[41] or volume of frontal white matter hyperintensities [42]. Diffusion tensor imaging showed that white matter microstructural integrity was disrupted in the left anterior cingulate among AD patients with apathy [43].

Functional . Studies using PET imaging to measure cortical metabolic activity or perfusion found that apathy in AD was associated with low metabolism or perfusion in anterior cingulate or orbitofrontal cortex (e.g. Marshall et al. [44]).

Hypoperfusion of other regions in frontal cortex or basal ganglia was inconsistently reported.

One study found particular aspects of the apathy syndrome were associated with hypoperfusion of individual cortical regions [45]. Ligand neuroimaging measures of neuroreceptor sites in

AD have demonstrated lower dopamine transporter binding in the bilateral putamen associated with poor initiative [46], and lower cholinergic receptor binding in the left frontal cortex associated with blunted affect, emotional withdrawal, and motor slowing [47].

While still speculative, these studies collectively suggest that apathy in AD is associated with atrophy and dysfunction of medial and inferior frontal regions that mediate behavioral initiation, motivation, and reward mechanisms. Reduced dopaminergic input to these regions and low frontal cholinergic binding may be relevant.

Dysfunction of additional regions in frontal heteromodal association cortex or interconnected basal ganglia structures may also contribute to the expression of apathy.

Several research approaches using neuroimaging can improve understanding of apathy in AD and contribute to treatment development. Steps forward include the assessment of brainbehavior relationships in larger samples with well-characterized apathy symptoms and other behavioral syndromes. Interrelationships among

5

behavioral syndromes and relevant neurobiological factors can thus be examined.

Neuroimaging studies should evaluate the brain basis of distinct components of the apathy syndrome (cognitive, emotional, and behavioral). Different biologic contributions to each component may be revealed, with heuristic value and treatment implications. Longitudinal studies can demonstrate relationships over the course of the neurodegenerative process, and can address causal factors more directly.

Multimodality imaging studies (e.g., structure, metabolism, neuroreceptor, and/or amyloid) can reveal relationships among multiple pathophysiological processes. Finally, neuroimaging studies in preclinical or clinical treatment studies can identify treatment targets and neurobiological factors that mediate successful treatment response.

Neurobiology and Biomarkers

Imaging data suggest that regional substrates of apathy include structural and functional alterations in the anterior cingulate, orbitofrontal cortex, or other components of frontal circuits such as the dorsolateral frontal cortex, frontal white matter, or basal ganglia. From a neuropathological perspective, apathy may result from atrophy and white matter tract changes

(described above), likely indicating loss of neurons and synapses innervating and connecting these particular regions [48, 49]. A biomarker is a biological test or marker of disease state or disease mechanism. Biomarkers are intertwined with an understanding of the biological mechanisms that may underlie apathy in AD.

Neurochemical . Neurochemical correlates of apathy include reduced indices of dopamine in the putamen [50], reduced acetylcholine in lateral frontal cortex [51] and lower plasma

GABA [52]. Pharmacologic challenge has shown that AD patients with apathy have a blunted subjective reward after administration of the dopaminergic agent dextroamphetamine[53].

These changes provide a rational basis for the success in some trials of both cholinergic [54] and dopaminergic agents [55] in reducing apathy in people with dementia. Whilst it does not always follow that simply correcting a perceived reduction in a particular neurotransmitter will produce benefit, these data appear to confirm the hypothesis that dopaminergic circuits linking the basal ganglia with the anterior cingulate and frontal cortices, normally involved in motivation and reward, are dysfunctional in people with AD and apathy [55]. Further neurochemical indices have not been explored as biomarkers. For example, for dopaminergic neurotransmission, potential biomarkers include the precursor l-3,4dihydroxyphenylalanine (L-DOPA), dopamine

(DA) and metabolite 3,4-dihydroxyphenylacetic acid (DOPAC). There are reports of increased

CSF DOPAC in AD vs. controls [56] but no difference in plasma DA [57]. Urine levels of L-

DOPA, DA, and DOPA are 2-3X higher in controls than AD [58]. CSF levels of dopamine transporter are quantified by Western blot [59].

Amyloid and tau . Higher NPI apathy scores were associated with higher CSF tau and phosphorylated tau, suggesting increased tangle formation, in 32 AD patients [60]. No such associations were observed between CSF amyloid, and NPI depression or psychosis scores.

Inflammatory markers. Both mood disorders and AD may be associated with inflammation

[61, 62] with specific increases in blood levels of Interleukin-6 (IL-6) and Tumor Necrosis

Factor alpha (TNF-α) [63, 64]. Given the overlap of symptoms between depression and apathy, it is possible that apathy is associated with an altered inflammatory state in AD. Other potential biomarkers of inflammation include

CSF antimicroglial antibodies [65], serum neopterin [66] and macrophage-colony stimulating factor [67].

Genetics. There are few data on genetic associations of apathy in AD. There are reports of null associations with polymorphisms of

ApoE, COMT, and the 5HT promoter [68-70] but no reports on associations with AD susceptibility genes recently identified in genome-wide association studies [71] [72]

Key steps forward to aid our understanding of the neurobiology of apathy in dementia should include a hypothesis-generating autopsy investigation of biochemical correlates of apathy in people with dementia, controlling for other

6

neuropsychiatric disturbances. Such studies can provide opportunities for focused imaging studies in well-chosen patient populations and will drive experimental medicine studies of licensed drugs in the first instance, and stimulate industry to undertake drug development based on these new targets.

There is much need for investigation of apathy biomarkers in AD, such as tau/phosphorylated tau, DA, and pro-inflammatory cytokines.

While these are unlikely to be solely associated with apathy, regional distribution of biomarkers for AD may be important. More specific biomarkers for apathy will emerge with greater understanding of the neurobiology and may include those related to specific neurotransmitters. In addition, such data are subject to marked variance, and CSF levels of amyloid and cytokines vary with time of day, sleep, and lumbar puncture itself [73-75] and clearly there is significant need for validation.

Animal models

While there have been few investigations of animal models of apathy, there are promising clues in models of mood disorders and addiction.

For example, chronic mild stress and psychosocial stress rodent models of

“depression” exhibit behaviours that are similar to apathy in humans, namely decreased motivation evidenced by decreased desire to consume sucrose solution, or decreased socialization and sexual behavior [76]. Another mechanism for inducing apathy in rodents is dopamine receptor antagonism by exogenous antipsychotics [77]. The progressive-ratio schedule of reinforcement is widely used in animal models of reinforcement including cocaine [78] and food administration [79], and may be a viable tool for assessment of motivation. Both these approaches might be applied to the AD transgenic mouse models or to the development of a novel model of apathy in

AD which would represent an important advance.

Pharmacotherapy

There are few data to guide clinicians in treating apathy in AD. Results from a randomized withdrawal study [80] as well a randomized,

7 placebo-controlled trial in moderate-to-severe

[81, 82] AD patients supported donepezil treatment for apathy. A randomized, placebocontrolled trial in mild-to-moderate AD suggests efficacy in delaying the emergence of apathy

[83]. Improvements in apathy were reported with galantamine treatment for mild-to-moderate

AD in a randomized, placebo-controlled trial

[84]. Similar results were reported in an open label galantamine study [85] and with rivastigmine in moderate-to-severe AD patients

[86]. Double-blind, placebo-controlled trials with metrifonate, [87, 88] and an open-label study with tacrine [89] found a significant reduction in NPI apathy scores in AD patients.

However, it should be noted that data supporting cholinesterase inhibitors for treating apathy in

AD have evaluated apathy as a secondary outcome only. Furthermore, patients were not recruited because of apathy.

For off-label use, a case report of modafinil

(without a formal diagnosis of AD) [90] and a randomized controlled trial for nefiracetam

(without AD) [91] provide suggestions of effectiveness warranting additional study. Two randomized, controlled studies, subject to the same limitations as with the cholinesterase trials, found significant improvements in apathy for patients with AD or vascular dementia (VaD) treated with memantine [92]; [93]. A series of small studies with mixed results suggest possible effectiveness of dopaminergic agents including amantadine [94]; [95], bupropion [96] and ropinirole [97] for apathy in patients with a range of cognitive impairments.

Methylphenidate for apathy has been explored in many patient populations including severely demented nursing home patients, [98] VaD patients, [99] AD patients [100] and in a small number of patients with mixed diagnoses in a series of individual cross-over, double-blind, randomized “N of 1” trials [101]. A small, crossover randomized controlled trial in outpatients with mild to moderate AD demonstrated that methylphenidate was efficacious in the treatment of apathy [102]. These results were supported by a randomized placebo controlled parallel group trial [103]. Currently, a multicentre, doubleblind, placebo-controlled trial is investigating methylphenidate treatment for apathetic patients

with mild-to-moderate AD with results expected in 2012 [104] Lastly, double-blind, placebocontrolled trials have found improvements in apathy with ginkgo biloba extract EGb 761 in

AD and VaD [105] as well as probable AD patients [106] as reported in secondary analyses.

It should be apparent that there is a need for placebo-controlled trials with apathy as the primary target and outcome. These studies should use current diagnostic criteria and take advantage of advances in neuroimaging and biomarkers, combined with pharmacologic challenge, to better understand the relationship between apathy and treatment response. Trials should include patients with carefully defined apathy symptoms, paying particular attention to delineating subtypes of apathy. Ultimately, the validity of diagnostic criteria will be related to whether or not they can identify patients with a higher probability of responding to specific treatments.

Non-pharmacological management

A systematic review of the literature on nonpharmacological interventions for apathy in dementia focused on randomized or pseudorandomized trials of interventions grouped for convenience into six categories. The review noted many other studies that were small, open label or qualitative, which reported benefits, but that could not be captured quantitatively. Only one category, a heterogeneous group of interventions, labelled Therapeutic Activities, contained sufficient, high research quality studies [107]. Therapeutic Activities included stimulation, creative activities, cooking,

Montessori methods or incentive based behavioral approaches, often individually tailored to patient needs. This category of interventions was found to be the most effective.

Positive results, although less impressive, were reported in other categories of intervention, namely music, exercise, multi-sensory stimulation, pet therapy and special care units.

However the evidence for efficacy of these interventions was limited as the standard of research was generally low.

It may be that individual attention in activities personally rewarding to recipients is the key element and that application of a single type of

8 intervention across a broad range of people with dementia and apathy will not be enough to demonstrate clinically and statistically significant outcomes. Even where such outcomes can be shown, effects usually wane once interventions cease unless activities are continued.

Future research may be best designed to match activities to individual interests, personal history and retained skills to engage persons with dementia and maintain involvement. Useful outcome measures are scales to evaluate prespecified goals such as goal attainment scales

[108] or quality of life indicators, as well as apathy itself. Given the increased prevalence or worsening of apathy with dementia progression, delay of emergence, or stabilization of severity over long periods might be important outcomes.

Alternative strategies may have to be considered over the broad span of dementia severities.

Confounds that should be accounted for are depression, medication effects (therapeutic and adverse), physical problems that cause pain, lethargy or depletion, premorbid behavioral dispositions, and environmental factors and interventions (such as structured activity programs). Combination of non-pharmacological and drug treatments such as cholinesterase inhibitors should be evaluated. Finally, context is important in demonstrating efficacy and sustainability. Nursing homes that incorporate interventions into usual care or family caregivers that can continue use of individual activities are more likely to effect lasting improvements. By its nature, recruitment is difficult in undertaking apathy trials; proxy consent from family caregivers and provision of clear information to institutional ethics review boards are important.

Immediate Next Steps

Much progress has been made in defining apathy as a distinct syndrome. Key questions that should be studied in the next 5 years include:

1) Apathy is recognized as a marker of poor prognosis. Do the new diagnostic criteria also predict poor prognosis in

MCI and dementia? Are there valid subdomains of apathy, and can they be

reliably measured? Do subtypes of apathy confer a greater risk?

2) Neuroimaging studies collectively suggest that apathy in AD is associated with regions that mediate behavioral initiation, motivation, and reward mechanisms. Do the distinct components of the apathy syndrome

(cognitive, emotional, and behavioral) differ in this regard? Can they be used to predict treatment response?

3) There is little understanding of the role of genetics in presentation and response to treatment for those with apathy, and animal models have not been developed.

These areas have the potential to improve understanding of apathy and as a result, suggest new treatment targets.

4) Apathy is increasingly being recognized as a therapeutic target. The first RCTs have been completed. Do the diagnostic criteria define a population who responds to specific interventions? Will treatment of apathy change its relationship with poor prognosis?

5) There is a need for both pharmacologic and nonpharmacologic trials with apathy as a primary outcome measure. For nonpharmacologic studies, interventions may have to be individualized.

Collectively, these studies should use current diagnostic criteria and take advantage of advances in neuroimaging and biomarkers, combined with pharmacologic challenge to better understand the relationship between apathy and treatment response.

9

Acknowledgements

KLL receives research support funding from the National Institutes of Health (5R01AG033032),

Canadian Institute for Health Research (MOP-114913), Alzheimer Society of Canada (#12-74),

Heart and Stroke Foundation (#NA 7220) and the Ontario Mental Health Foundation. PBR receives support from NIA grants 1R01AG039384, 5K08AG029157, 5R01AG033032 and the

American Foundation for Aging Research. DLS receives support from the Department of

Veterans Affairs (Merit Review Award) and the NIMH (MH56031). PTF receives research support from the UK charities Alzheimer’s Society and Alzheimer’s Research UK (Brains for

Dementia Research), Alzheimer’s Society and The Edmund J Safra Foundation. PRP receives research support from the Department of Veterans Affairs and the Alzheimer’s Association.

YEG receives research support funding from the National Institutes of Health (AG06786,

RR024150 (Mayo Clinic CTSA [Career Transition Award]), RWJ Foundation and the European

Union Regional Development Fund (Project FNUSA-ICRC: CZ.1.05/1.1.00/02.0123).

Conflict of Interest

The authors have no conflict of interest to report. The views expressed in this paper are those of the authors, and do not necessarily represent the official views of their sponsoring agencies.

10

Table 1. Diagnostic criteria for apathy in neurodegenerative disease

Diagnosis requires fulfillment of criteria A, B, C and D

A.

Loss of or diminished motivation in comparison to the patient’s previous level of functioning and which is not consistent with his age or culture. These changes in motivation may be reported by the patient himself or by the observations of others

B.

Presence of at least one symptom in at least two of the three following domains for a period of at least four weeks and present most of the time

Domain B1 – Behavior Domain B2 – Cognition

Loss of, or diminished, goaldirected behavior as evidenced by at least one of the following:

Loss of, or diminished, goaldirected cognitive activity as evidenced by at least one of the following:

Loss of self-initiated behavior

(e.g., in
starting conversation, doing basic tasks of day-to-day living, seeking social activities, communicating choices)

Loss of environmentstimulated behavior (e.g., in responding to conversation, participating in social activities)

Loss of spontaneous ideas and curiosity for routine and new events (i.e., challenging tasks, recent news, social opportunities, personal/family and social affairs).

Loss of environmentstimulated ideas and curiosity for routine and new events

(i.e., in the person’s residence, neighborhood or community).

Domain B3 – Emotion

Loss of, or diminished, emotion as evidenced by at least one of the following:

Loss of spontaneous emotion, observed or self-reported (e.g., subjective feeling of weak or absent emotions, or observation by others of a blunted affect)

Loss of emotional responsiveness to positive or negative stimuli or events

(e.g., observer-reports of unchanging affect, or of little emotional reaction to exciting events, personal loss, serious illness, emotional-laden news)

C.

These symptoms (A & B) cause clinically significant impairment in personal, social, occupational, or other important areas of functioning

D.

The symptoms (A & B) are not exclusively explained or due to physical disabilities (e.g. blindness and loss of hearing), to motor disabilities, to diminished level of consciousness or to the direct physiological effects of a substance (e.g. drug of abuse, a medication)

11

References

1.

2.

3.

4.

5.

6.

7.

Marin, R.S., Differential diagnosis and classification of apathy. The American journal of psychiatry, 1990. 147 (1): p. 22-30.

Marin, R.S., Apathy: a neuropsychiatric syndrome. The Journal of neuropsychiatry and clinical neurosciences, 1991. 3 (3): p. 243-54.

Kokmen, E., et al., Impact of referral bias on clinical and epidemiological studies of

Alzheimer's disease. Journal of clinical epidemiology, 1996. 49 (1): p. 79-83.

Tsuang, D., et al., Impact of sample selection on APOE epsilon 4 allele frequency: a comparison of two Alzheimer's disease samples. J Am Geriatr Soc, 1996. 44 (6): p. 704-7.

Mega, M.S., et al., The spectrum of behavioral changes in Alzheimer's disease.

Neurology, 1996. 46 (1): p. 130-5.

Hwang, T.J., et al., Mild cognitive impairment is associated with characteristic neuropsychiatric symptoms. Alzheimer disease and associated disorders, 2004. 18 (1): p.

17-21.

Copeland, M.P., et al., Psychiatric symptomatology and prodromal Alzheimer's disease.

Alzheimer disease and associated disorders, 2003. 17 (1): p. 1-8.

8. Lyketsos, C.G., et al., Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the cardiovascular health study. JAMA : the journal of the American Medical Association, 2002. 288 (12): p. 1475-83.

9. Lyketsos, C.G., et al., Mental and behavioral disturbances in dementia: findings from the

Cache County Study on Memory in Aging. The American journal of psychiatry, 2000.

157 (5): p. 708-14.

10. Geda, Y.E., et al., Prevalence of neuropsychiatric symptoms in mild cognitive impairment and normal cognitive aging: population-based study. Archives of general psychiatry, 2008. 65 (10): p. 1193-8.

11. Palmer, K., et al., Predictors of progression from mild cognitive impairment to Alzheimer disease. Neurology, 2007. 68 (19): p. 1596-602.

12. Onyike, C.U., et al., Epidemiology of apathy in older adults: the Cache County Study.

Am J Geriatr Psychiatry, 2007. 15 (5): p. 365-75.

13. Brodaty, H., et al., Do people become more apathetic as they grow older? A longitudinal study in healthy individuals. Int Psychogeriatr, 2010. 22 (3): p. 426-36.

14. Freels, S., et al., Functional status and clinical findings in patients with Alzheimer's disease. J Gerontol, 1992. 47 (6): p. M177-82.

15. Boyle, P.A., et al., Executive dysfunction and apathy predict functional impairment in

Alzheimer disease. Am J Geriatr Psychiatry, 2003. 11 (2): p. 214-21.

12

16. Landes, A.M., et al., Apathy in Alzheimer's disease. J Am Geriatr Soc, 2001. 49 (12): p.

1700-7.

17. Thomas, P., et al., Family, Alzheimer's disease and negative symptoms. Int J Geriatr

Psychiatry, 2001. 16 (2): p. 192-202.

18. Palmer, K., et al., Neuropsychiatric predictors of progression from amnestic-mild cognitive impairment to Alzheimer's disease: the role of depression and apathy. J

Alzheimers Dis, 2010. 20 (1): p. 175-83.

19. Rosenberg, P., et al., The associations of neuropsychiatric symptoms in MCI with incident dementia and Alzheimer's Disease. Am J Geriatr Psychiatry, 2012. (In press) .

20. Mega, M.S., et al., The spectrum of behavioral responses to cholinesterase inhibitor therapy in Alzheimer disease. Arch Neurol, 1999. 56 (11): p. 1388-93.

21. Starkstein, S.E., et al., A prospective longitudinal study of apathy in Alzheimer's disease.

J Neurol Neurosurg Psychiatry, 2006. 77 (1): p. 8-11.

22. Cummings, J.L., et al., The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology, 1994. 44 (12): p. 2308-14.

23. Kaufer, D.I., et al., Validation of the NPI-Q, a brief clinical form of the Neuropsychiatric

Inventory. The Journal of neuropsychiatry and clinical neurosciences, 2000. 12 (2): p.

233-9.

24. Marin, R.S., Differential diagnosis and classification of apathy. Am J Psychiatry, 1990.

147 (1): p. 22-30.

25. Cummings, J.L., Frontal-subcortical circuits and human behavior. Arch Neurol, 1993.

50 (8): p. 873-80.

26. Levy, R. and B. Dubois, Apathy and the functional anatomy of the prefrontal cortex-basal ganglia circuits. Cereb Cortex, 2006. 16 (7): p. 916-28.

27. Marin, R.S., R.C. Biedrzycki, and S. Firinciogullari, Reliability and validity of the

Apathy Evaluation Scale. Psychiatry Res, 1991. 38 (2): p. 143-62.

28. Starkstein, S.E., et al., On the overlap between apathy and depression in dementia. J

Neurol Neurosurg Psychiatry, 2005. 76 (8): p. 1070-4.

29. Starkstein, S.E., et al., Reliability, validity, and clinical correlates of apathy in

Parkinson's disease. J Neuropsychiatry Clin Neurosci, 1992. 4 (2): p. 134-9.

30. Robert, P.H., et al., The apathy inventory: assessment of apathy and awareness in

Alzheimer's disease, Parkinson's disease and mild cognitive impairment. Int J Geriatr

Psychiatry, 2002. 17 (12): p. 1099-105.

31. Strauss, M.E. and S.D. Sperry, An informant-based assessment of apathy in Alzheimer disease. Neuropsychiatry Neuropsychol Behav Neurol, 2002. 15 (3): p. 176-83.

32. Sockeel, P., et al., The Lille apathy rating scale (LARS), a new instrument for detecting and quantifying apathy: validation in Parkinson's disease. J Neurol Neurosurg Psychiatry,

2006. 77 (5): p. 579-84.

13

33.

Agüera Ortiz, L., et al., Proceso de creación de la escala APADEM-NH para la medición de la apatía en pacientes con demencia institucionalizados. Psicogeriatría, 2011.

3 (1): p.

29-36.

34. Clarke, D.E., et al., Are the available apathy measures reliable and valid? A review of the psychometric evidence. Journal of psychosomatic research, 2011. 70 (1): p. 73-97.

35. Robert, P., et al., Proposed diagnostic criteria for apathy in Alzheimer's disease and other neuropsychiatric disorders. Eur Psychiatry, 2009. 24 (2): p. 98-104.

36. Robert, P., et al., Proposed diagnostic criteria for apathy in Alzheimer's disease and other neuropsychiatric disorders. European psychiatry : the journal of the Association of

European Psychiatrists, 2009. 24 (2): p. 98-104.

37. Marin, R.S., Apathy: a neuropsychiatric syndrome. J Neuropsychiatry Clin Neurosci,

1991. 3 (3): p. 243-54.

38. Starkstein, S.E., et al., Syndromic validity of apathy in Alzheimer's disease. Am J

Psychiatry, 2001. 158 (6): p. 872-7.

39. Mulin, E., et al., Diagnostic criteria for apathy in clinical practice. Int J Geriatr

Psychiatry, 2011. 26 (2): p. 158-65.

40. Zeitzer, J.M., et al., Phenotyping Apathy in Individuals With Alzheimer Disease Using

Functional Principal Component Analysis. Am J Geriatr Psychiatry, 2012.

41. Jonsson, M., et al., Apathy is a prominent neuropsychiatric feature of radiological whitematter changes in patients with dementia. Int J Geriatr Psychiatry, 2010. 25 (6): p. 588-95.

42. Starkstein, S.E., et al., Neuroimaging correlates of apathy and depression in Alzheimer's disease. J Neuropsychiatry Clin Neurosci, 2009. 21 (3): p. 259-65.

43. Kim, J.W., et al., Microstructural alteration of the anterior cingulum is associated with apathy in Alzheimer disease. Am J Geriatr Psychiatry, 2011. 19 (7): p. 644-53.

44. Marshall, G.A., et al., Positron emission tomography metabolic correlates of apathy in

Alzheimer's disease. Archives of Neurology, 2007. 64 (7): p. 1015-1020.

45. Benoit, M., et al., Brain perfusion correlates of the apathy inventory dimensions of

Alzheimer's disease. Int J Geriatr Psychiatry, 2004. 19 (9): p. 864-9.

46. David, R., et al., Striatal dopamine transporter levels correlate with apathy in neurodegenerative diseases A SPECT study with partial volume effect correction. Clin

Neurol Neurosurg, 2008. 110 (1): p. 19-24.

47. Sultzer, D., et al., Cholinergic receptor imaging in Alzheimer's disease: method and early results, in Annual Meeting of the American Association for Geriatric Psychiatry. March

2010, Am J Geriatr Psychiatry. p. S71-72.

48. Tekin, S., et al., Orbitofrontal and anterior cingulate cortex neurofibrillary tangle burden is associated with agitation in Alzheimer disease. Annals of Neurology, 2001. 49 (3): p.

355-361.

49. Marshall, G.A., et al., Neuropathologic correlates of apathy in Alzheimer's disease.

Dementia and geriatric cognitive disorders, 2006. 21 (3): p. 144-7.

14

50. David, R., et al., Striatal dopamine transporter levels correlate with apathy in neurodegenerative diseases A SPECT study with partial volume effect correction.

Clinical Neurology and Neurosurgery, 2008. 110 (1): p. 19-24.

51. Sultzer D., M.R., Campa OM., Achamallah N., Harwood DG., Brody AL., Walston A.,

Maldelkern MA., Cholinergic receptor imaging in Alzheimer's disease: method and early results. Am J Geriatr Psychiatry, 2010. 18 (Suppl 1): p. S71-72.

52. Lanctot, K.L., et al., Behavioral correlates of GABAergic disruption in Alzheimer's disease. International psychogeriatrics / IPA, 2007. 19 (1): p. 151-8.

53. Lanctot, K.L., et al., Apathy associated with Alzheimer disease: use of dextroamphetamine challenge. Am J Geriatr Psychiatry, 2008. 16 (7): p. 551-7.

54. Feldman, H., et al., A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer's disease. Neurology, 2001. 57 (4): p. 613-620.

55. Mitchell, R.A., N. Herrmann, and K.L. Lanctot, The role of dopamine in symptoms and treatment of apathy in Alzheimer's disease. CNS neuroscience & therapeutics, 2011.

17 (5): p. 411-27.

56. Zubenko, G.S., et al., Cerebrospinal fluid levels of angiotensin-converting enzyme, acetylcholinesterase, and dopamine metabolites in dementia associated with Alzheimer's disease and Parkinson's disease: a correlative study. Biol Psychiatry, 1986. 21 (14): p.

1365-81.

57. Umegaki, H., et al., Low plasma epinephrine in elderly female subjects of dementia of

Alzheimer type. Brain Res, 2000. 858 (1): p. 67-70.

58. Liu, L., et al., Simultaneous determination of catecholamines and their metabolites related to Alzheimer's disease in human urine. J Sep Sci, 2011. 34 (10): p. 1198-204.

59. Duarte, S.T., et al., Analysis of synaptic proteins in the cerebrospinal fluid as a new tool in the study of inborn errors of neurotransmission. J Inherit Metab Dis. 34 (2): p. 523-8.

60. Skogseth, R., et al., Neuropsychiatric correlates of cerebrospinal fluid biomarkers in

Alzheimer's disease. Dement Geriatr Cogn Disord, 2008. 25 (6): p. 559-63.

61. Rosenberg, P.B., Clinical aspects of inflammation in Alzheimer's disease. Int Rev

Psychiatry, 2005. 17 (6): p. 503-14.

62. Dantzer, R., et al., Identification and treatment of symptoms associated with inflammation in medically ill patients. Psychoneuroendocrinology, 2008. 33 (1): p. 18-29.

63. Dowlati, Y., et al., A meta-analysis of cytokines in major depression. Biol Psychiatry,

2009. 67 (5): p. 446-57.

64. Swardfager, W., et al., A meta-analysis of cytokines in Alzheimer's disease. Biol

Psychiatry, 2010. 68 (10): p. 930-41.

65. McRae, A., et al., Cerebrospinal fluid antimicroglial antibodies in Alzheimer disease: a putative marker of an ongoing inflammatory process. Exp Gerontol, 2007. 42 (4): p. 355-

63.

15

66. Maes, M., Depression is an inflammatory disease, but cell-mediated immune activation is the key component of depression. Prog Neuropsychopharmacol Biol Psychiatry, 2011.

35 (3): p. 664-75.

67. Laske, C., et al., Macrophage colony-stimulating factor (M-CSF) in plasma and CSF of patients with mild cognitive impairment and Alzheimer's disease. Curr Alzheimer Res,

2010. 7 (5): p. 409-14.

68. Albani, D., et al., The serotonin transporter promoter polymorphic region is not a risk factor for Alzheimer's disease related behavioral disturbances. J Alzheimers Dis, 2009.

18 (1): p. 125-30.

69. Borroni, B., et al., Genetic correlates of behavioral endophenotypes in Alzheimer disease: role of COMT, 5-HTTLPR and APOE polymorphisms. Neurobiol Aging, 2006. 27 (11): p. 1595-603.

70. Chen, C.S., et al., Apolipoprotein E Polymorphism and Behavioral and Psychological

Symptoms of Dementia in Patients With Alzheimer Disease. Alzheimer Dis Assoc

Disord, 2011.

71. Hollingworth, P., et al., Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1,

CD33 and CD2AP are associated with Alzheimer's disease. Nat Genet, 2011. 43 (5): p.

429-35.

72. Naj, A.C., et al., Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nat Genet, 2011. 43 (5): p. 436-41.

73. Bateman, R.J., et al., Fluctuations of CSF amyloid-beta levels: implications for a diagnostic and therapeutic biomarker. Neurology, 2007. 68 (9): p. 666-9.

74. van Munster, B.C., et al., Time-course of cytokines during delirium in elderly patients with hip fractures. J Am Geriatr Soc, 2008. 56 (9): p. 1704-9.

75. Kang, J.E., et al., Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science, 2009. 326 (5955): p. 1005-7.

76. Krishnan, V. and E.J. Nestler, Animal models of depression: molecular perspectives.

Curr Top Behav Neurosci, 2011. 7 : p. 121-47.

77. Wise, R.A., Dopamine, learning and motivation. Nat Rev Neurosci, 2004. 5 (6): p. 483-

94.

78. McGregor, A., G. Baker, and D.C. Roberts, Effect of 6-hydroxydopamine lesions of the amygdala on intravenous cocaine self-administration under a progressive ratio schedule of reinforcement. Brain Res, 1994. 646 (2): p. 273-8.

79. Weinberg, Z.Y., M.L. Nicholson, and P.J. Currie, 6-Hydroxydopamine lesions of the ventral tegmental area suppress ghrelin's ability to elicit food-reinforced behavior.

Neurosci Lett, 2011. 499 (2): p. 70-3.

80. Holmes, C., et al., The efficacy of donepezil in the treatment of neuropsychiatric symptoms in Alzheimer disease. Neurology, 2004. 63 (2): p. 214-9.

81. Gauthier, S., et al., Efficacy of donepezil on behavioral symptoms in patients with moderate to severe Alzheimer's disease. Int Psychogeriatr, 2002. 14 (4): p. 389-404.

16

82. Feldman, H., et al., A 24-week, randomized, double-blind study of donepezil in moderate to severe Alzheimer's disease. Neurology, 2001. 57 (4): p. 613-20.

83. Waldemar, G., et al., Effect of donepezil on emergence of apathy in mild to moderate

Alzheimer's disease. Int J Geriatr Psychiatry, 2011. 26 (2): p. 150-7.

84. Herrmann, N., et al., Galantamine treatment of problematic behavior in Alzheimer disease: post-hoc analysis of pooled data from three large trials. Am J Geriatr Psychiatry,

2005. 13 (6): p. 527-34.

85. Brodaty, H., et al., A naturalistic study of galantamine for Alzheimer's disease. CNS

Drugs, 2006. 20 (11): p. 935-43.

86. Cummings, J.L., et al., Effects of rivastigmine treatment on the neuropsychiatric and behavioral disturbances of nursing home residents with moderate to severe probable

Alzheimer's disease: a 26-week, multicenter, open-label study. Am J Geriatr

Pharmacother, 2005. 3 (3): p. 137-48.

87. Dubois, B., et al., A multicentre, randomized, double-blind, placebo-controlled study to evaluate the efficacy, tolerability and safety of two doses of metrifonate in patients with mild-to-moderate Alzheimer's disease: the MALT study. Int J Geriatr Psychiatry, 1999.

14 (11): p. 973-82.

88. Kaufer, D., Beyond the cholinergic hypothesis: the effect of metrifonate and other cholinesterase inhibitors on neuropsychiatric symptoms in Alzheimer's disease. Dement

Geriatr Cogn Disord, 1998. 9 Suppl 2 : p. 8-14.

89. Kaufer, D., J.L. Cummings, and D. Christine, Differential neuropsychiatric symptom responses to tacrine in Alzheimer's disease: relationship to dementia severity. J

Neuropsychiatry Clin Neurosci, 1998. 10 (1): p. 55-63.

90. Padala, P.R., W.J. Burke, and S.C. Bhatia, Modafinil therapy for apathy in an elderly patient. Ann Pharmacother, 2007. 41 (2): p. 346-9.

91. Robinson, R.G., et al., Double-blind treatment of apathy in patients with poststroke depression using nefiracetam. J Neuropsychiatry Clin Neurosci, 2009. 21 (2): p. 144-51.

92. Pantev M, R.R., Gortelmeyer R, Clinical and behavioural eveluation in long-term care patients with mild to modertae dementia under memantine treatment. Zeitschrt fur

Gerontopsychologie Und-Psychiatrie, 1993. 6 : p. S103-S117

93. Winblad, B. and N. Poritis, Memantine in severe dementia: results of the 9M-Best Study

(Benefit and efficacy in severely demented patients during treatment with memantine).

Int J Geriatr Psychiatry, 1999. 14 (2): p. 135-46.

94. Van Reekum, R., et al., N of 1 study: amantadine for the amotivational syndrome in a patient with traumatic brain injury. Brain Inj, 1995. 9 (1): p. 49-53.

95. Drayton, S.J., et al., Amantadine for executive dysfunction syndrome in patients with dementia. Psychosomatics, 2004. 45 (3): p. 205-9.

96. Corcoran, C., M.L. Wong, and V. O'Keane, Bupropion in the management of apathy. J

Psychopharmacol, 2004. 18 (1): p. 133-5.

17

97. Kohno, N., et al., Successful treatment of post-stroke apathy by the dopamine receptor agonist ropinirole. J Clin Neurosci, 2010. 17 (6): p. 804-6.

98. Maletta, G.J.W., T., Reversal of anorexia by methylphenidate in apathetic, severely demented nursing home patients. American Journal of Geriatric Psychiatry, 1993. 1 (3): p.

234-243.

99. Galynker, I., et al., Methylphenidate treatment of negative symptoms in patients with dementia. J Neuropsychiatry Clin Neurosci, 1997. 9 (2): p. 231-9.

100. Padala, P.R., et al., Methylphenidate for apathy and functional status in dementia of the

Alzheimer type. Am J Geriatr Psychiatry, 2010. 18 (4): p. 371-4.

101. Jansen, I.H., et al., Toward individualized evidence-based medicine: five "N of 1" trials of methylphenidate in geriatric patients. J Am Geriatr Soc, 2001. 49 (4): p. 474-6.

102. Herrmann, N., et al., Methylphenidate for the treatment of apathy in Alzheimer disease: prediction of response using dextroamphetamine challenge. J Clin Psychopharmacol,

2008. 28 (3): p. 296-301.

103. Padala, P.R., Burke, W.J.. , Treatment of apathy: Results of a randomized double blind placebo controlled clinical trial. . Presented at the 2011 International Conference on

Alzheimer’s Disease in Paris, France., 2011.

104. Drye, L.T., et al., Designing a Trial to Evaluate Potential Treatments for Apathy in

Dementia: The Apathy in Dementia Methylphenidate Trial (ADMET): The Apathy in

Dementia Methylphenidate Trial (ADMET). Am J Geriatr Psychiatry, 2012.

105. Scripnikov, A., A. Khomenko, and O. Napryeyenko, Effects of Ginkgo biloba extract

EGb 761 on neuropsychiatric symptoms of dementia: findings from a randomised controlled trial. Wien Med Wochenschr, 2007. 157 (13-14): p. 295-300.

106. Bachinskaya, N., R. Hoerr, and R. Ihl, Alleviating neuropsychiatric symptoms in dementia: the effects of Ginkgo biloba extract EGb 761. Findings from a randomized controlled trial. Neuropsychiatr Dis Treat, 2011. 7 : p. 209-15.

107. Brodaty, H. and K. Burns, Nonpharmacological Management of Apathy in Dementia: A

Systematic Review. Am J Geriatr Psychiatry, 2011.

108. Kiresuk, R.S.T., Goal attainment scaling: A general method for evaluating comprehensive community mental health programs Community Mental Health Journal,

1968. 4 (6): p. 443-453.

18

Download