139
Views
20
CrossRef citations to date
0
Altmetric
Review

Optimal treatment of Alzheimer’s disease psychosis: challenges and solutions

&
Pages 2253-2262 | Published online: 24 Nov 2014

Abstract

Psychotic symptoms emerging in the context of neurodegeneration as a consequence of Alzheimer’s disease was recognized and documented by Alois Alzheimer himself in his description of the first reported case of the disease. Over a quarter of a century ago, in the context of attempting to develop prognostic markers of disease progression, psychosis was identified as an independent predictor of a more-rapid cognitive decline. This finding has been subsequently well replicated, rendering psychotic symptoms an important area of exploration in clinical history taking – above and beyond treatment necessity – as their presence has prognostic significance. Further, there is now a rapidly accreting body of research that suggests that psychosis in Alzheimer’s disease (AD+P) is a heritable disease subtype that enjoys neuropathological specificity and localization. There is now hope that the elucidation of the neurobiology of the syndrome will pave the way to translational research eventuating in new treatments. To date, however, the primary treatments employed in alleviating the suffering caused by AD+P are the atypical antipsychotics. These agents are approved by the US Food and Drug Administration for the treatment of schizophrenia, but they have only marginal efficacy in treating AD+P and are associated with troubling levels of morbidity and mortality. For clinical approaches to AD+P to be optimized, this syndrome must be disentangled from other primary psychotic disorders, and recent scientific advances must be translated into disease-specific therapeutic interventions. Here we provide a review of atypical antipsychotic efficacy in AD+P, followed by an overview of critical neurobiological observations that point towards a frontal, tau-mediated model of disease, and we suggest a new preclinical animal model for future translational research.

Introduction

There is urgency to the problem of the need for efficacious drug treatment in the management of Alzheimer’s disease with psychosis (AD+P). AD+P has grave consequence, particularly for caregivers, as it is associated with physical aggression, and caregivers are most often the victims.Citation1Citation3 Violent behavior that is directed towards caregivers is disruptive of care at the very least, but can also be dangerous for those charged with providing a safe environment for those suffering with disease. Not only is aggressive behavior inherently troublesome for families, but perhaps in part because of the difficulty of managing these behaviors, the rates of institutionalization are much higher in AD patients who have exhibited violent behavior.Citation4,Citation5 Notably, in one study of participants with elevated levels of physical aggression, 80% had delusions.Citation6 This suggests that psychosis at least aggravates the clinical course of AD, and creates profound caregiver stress that compounds the difficulty of taking care of a loved one with the cognitive impairment that is a defining feature of the illness.

Unfortunately, the current treatment approaches to AD+P, relying mostly on agents approved for the treatment of schizophrenia, have been largely disappointing. The efficacy of the atypical antipsychotics in the treatment of behavioral and psychological symptoms of dementia – derived from randomized placebo-controlled trials – has been parsed many ways, owing in part to the broad range of rating scales employed as outcome measures and the spectrum of neurobehavioral syndromes associated with AD. Indeed, trials that have enrolled over 5,000 patients to date have included a number of different psychometric scales of behavioral impairment, including the Neuropsychiatric Inventory (NPI); the Brief Psychiatric Rating Scale (BPRS); the Cohen-Mansfield Agitation Inventory; the Clinical Global Impression of Change; the Clinical Global Impression of Severity; and the Behavioral Pathology in Alzheimer’s Disease (BEHAVE-AD). Evaluating the efficacy of currently available treatments in AD+P is challenging, as the only available data comprises subscales distilled from comprehensive agitation inventories.

The most recent and most comprehensive meta-analysis to date of randomized placebo-controlled trials in the treatment of the behavioral and psychological symptoms of dementiaCitation7 suggests a consistent superiority of atypical agents (aripiprazole, quetiapine, olanzapine, risperidone) over placebo as measured with the aforementioned behavioral rating scales. In fact, when atypical agents are amalgamated and their efficacy data is compared with placebo, the most recent meta-analysis reveals that antipsychotics demonstrate a convincing superiority over placebo in the treatment of behavioral disturbances. Calculated mean differences and 95% confidence intervals on behavioral rating scales support this contention: BPRS, −1.58 (−2.52 to −0.65); Cohen-Mansfield Agitation Inventory, −1.84 (−3.01 to −0.61); NPI, −2.81 (−4.35 to −1.28); Clinical Global Impression of Change, −0.32 (−0.44 to −0.20); Clinical Global Impression of Severity, −0.19 (−0.3 to −0.09). These data, especially in light of the BPRS differences, could be taken as evidence that atypical antipsychotics are efficacious in the treatment of AD+P.

A careful look at these studies, however, suggests the ironic truth that amongst therapies for behavioral disturbances in AD, the atypical antipsychotics are inconsistent at best in the treatment of psychosis. Positive results from the above referenced meta-analytic data on BPRS total score outcomes do not necessarily reflect improvement in psychosis. The BPRS, (Overall and Gorham 1962, Psychological Report)Citation8 an inventory of psychopathology that has been extensively utilized in the evaluation of outcomes in schizophrenia and other psychotic illnesses, is a remarkably broad 18-item scale.Citation9 Only a few items could be interpreted with any relevance in AD+P: hostility, suspiciousness, hallucinatory behavior, and perhaps unusual thought content and uncooperativeness. Although not included in the most recent meta-analysis, data on psychosis can be extracted from the majority of placebo-controlled clinical trials of antipsychotics in dementia populations by looking at psychosis subscores derived from the NPI, the BEHAVE-AD, and the BPRS (). Olanzapine has been evaluated in comparison with placebo over 6–36 weeks in nursing home patients and outpatients.Citation10Citation12 In the first study, olanzapine at a dose of 5 mg and 10 mg, but not 15 mg, was superior to placebo on the psychosis subscale total score derived from the nursing home version of the NPI, the NPI-NH ().Citation10 In a second study of 1.0 mg, 2.5 mg, 5.0 mg, and 7.5 mg doses of olanzapine, only the 7.5 mg dose was superior to placebo as measured by the NPI-NH psychosis subscale.Citation11 In the Clinical Antipsychotics Trials of Intervention Effectiveness-Alzheimer’s Disease (CATIE-AD) study, an outpatient study comparing flexible doses of atypical antipsychotics (olanzapine, quetiapine, risperidone) to placebo, olanzapine at a mean dose of 5.5 mg was not superior to placebo on the psychosis factor of the BPRS.Citation12 Two 10-week randomized placebo-controlled trials have investigated aripiprazole in the treatment of AD+P.Citation13,Citation14 In the first study, an outpatient study employing a flexible dose, aripiprazole at a mean dose of 10 mg demonstrated superiority to placebo on the BPRS psychosis subscale but not on the NPI psychosis subscale.Citation13 In a nursing home study, comparing aripiprazole at doses of 2 mg, 5 mg, and 10 mg, only 10 mg was superior to placebo on the NPI-NH psychosis subscale.Citation14 Two studies, of 6 weeks and 36 weeks duration in nursing facilities and outpatient populations, respectively, have evaluated the efficacy of quetiapine in comparison with placebo for the treatment of AD+P.Citation12,Citation15 No evidence of efficacy in treating psychosis was generated for quetiapine from either study on the basis of the BPRS psychosis subscales. Prior to CATIE-AD, there were four randomized placebo-controlled trials of risperidone in AD+P, ranging from 8–36 weeks in both the nursing home residents and outpatients.Citation16Citation19 A meta-analysis of these four studies revealed that risperidone was superior to placebo on the BEHAVE-AD psychosis subscale, with an effect size of 0.154 (95% confidence interval 0.038–0.269), suggesting only a modest effect of drug treatment.Citation20 Data from CATIE-AD also evidenced a benefit of risperidone over placebo on the BPRS psychosis factor, the only agent to beat placebo on this measure.Citation12 The data on atypical antipsychotics, while inconsistent, do suggest some utility for most agents, with some superiority for risperidone; however, the magnitudes of treatment effects are not robust, and highly efficacious agents are desperately needed. One solution to this problem is to develop novel treatments based on the emerging neurobiology of AD+P, rather than relying on existing treatments for other psychotic conditions.

Table 1 Randomized placebo-controlled trials of atypical antipsychotics in the treatment of Alzheimer’s disease with psychosis

Phenomenology in AD+P: theoretical considerations

Psychotic symptoms in AD comprise delusions, with a reported prevalence of between 30%–40%,Citation21Citation24 and, to a smaller extent, hallucinations, with a prevalence of 5%–20%.Citation21,Citation23,Citation25Citation27 The heritability of AD+P has been firmly establishedCitation28 and is estimated to be between 30%–60%.Citation29 The persistence of untreated psychosis over time in Alzheimer’s disease in individual patients has not been firmly established, but available evidence suggests that approximately 40%–50% of psychotic individuals will continue to manifest psychotic symptoms after 1 year.Citation30 Rather than the bizarre delusions and complex network of unsubstantiated belief systems commonly seen in schizophrenia typified by logical impossibility and experiences alien to “ordinary life situations,”Citation31 the delusions in AD+P generally involve rather narrow elaborations and distortions of actual experience. The delusions of AD have been largely reconciled into two categories,Citation32,Citation33 delusions of persecution (eg, theft, harm, jealousy, abandonment)Citation2,Citation34Citation36 and delusions of misidentification (eg, phantom boarder, television images are animate, Capgras syndrome, house is not one’s home),Citation2,Citation35Citation38 the latter having once been conceptualized as representing perceptual abnormalities, but actually representing ideational fixations.Citation32,Citation39,Citation40 A category of “autobiographical delusions,”Citation32 for instance that a dead relative is still alive, has also been suggested,Citation41 but these may be regarded as an extension of the misidentification domain.

A few theoretical models have been developed to explain the common emergence of psychosis in AD – particularly focused on delusions – in those otherwise not afflicted with mental illness. Carefully reviewed by Reeves et alCitation32 the earliest of these ratiocinations suggested that psychosis results from a “logical attempt to understand the environment” in the context of degraded cognitive integrity.Citation42 For instance, the common delusions in AD – theft, jealousy, misidentification, abandonment, phantom boarder, television images are animateCitation21,Citation43,Citation44 – are possible narrative confabulations that fill in the gaps or normalize the experiences of those confronted with a history they have become unmoored from, and who rely on a gradually eroding apparatus for discriminating object from representation. The most intuitive support for this model comes from the most prevalent delusion in AD, that people are hiding or stealing things,Citation45Citation47 a potential confabulation that is entirely consistent with the primary cognitive deficit in AD, accelerated forgetting. Alternatively, some have suggested that brain disease results in “release symptomatology”Citation48 or that cognitive deficits germinate delusional ideas as a consequence of a “lack of corrective judgment” from other cortical structures.Citation49 These conceptualizations, while interesting, are broad and speculative and do not really generate any testable scientific predictions. More recent work has been focused on concrete neurobiological correlates of psychotic AD, and from that a glimpse of disease pathophysiology has begun to emerge.

Cognitive course and mortality in AD+P: evidence for a distinct syndrome?

The earliest specific reference to the problem of psychosis in the modern literature on Alzheimer’s disease emerges in a Russian neuropsychiatric journal in 1964.Citation50 It was another 20 years before critical observations were made that contributed to a reconceptualization of psychotic phenomena as expressions of a unique pathophysiological syndrome rather than a mere behavioral nuisance. In the context of searching for clinical signs that would predict the velocity of decline in AD, psychosis – in addition to myoclonus and extrapyramidal signs – was found to be an early marker heralding a hastened deterioration.Citation51,Citation52 This observation was quickly replicated in several studies reinforcing the prognostic value of psychosis in relationship to decline.Citation53,Citation54 Studies focused on the exploration of risk factors for psychotic symptoms in AD have also consistently identified a greater burden of cognitive impairment as an independent risk factor for psychosis, with acceleration of decline emerging prior to the onset of psychosis.Citation24,Citation46,Citation55,Citation56

The features of accelerated cognitive impairment in AD+P suggest some degree of frontal localization. In one study of AD+P and Alzheimer’s disease without psychosis (AD-P) that compared subjects of similar duration of illness, age, and education, AD+P subjects evidenced more impairment on tasks of frontal-lobe function, including the Wisconsin Card Sorting Test and the similarities subtest of the Wechsler Adult Intelligence Scale.Citation57 Recent studies suggest that working memory performance, a test of frontal function depending largely on the dorsolateral prefrontal cortex, enjoys a unique association with psychosis in AD. Early evidence for this emerged from an epidemiological study of AD+P risk factors, wherein performance on the digit span, a task of working memory, was observed to be a robust predictor of future psychosis in AD.Citation24 Impressively, a one-standard-deviation improvement in this task reduced the risk of developing hallucinations or delusions by two thirds. In our own study of cognition in AD+P, we evaluated AD and performed stepwise regression of cognitive domains in order to determine whether any cognitive domains (memory, executive function, language) predicted psychosis. We found that of the wide spectrum of domains of cognition interrogated in the analysis, only working memory interacted with psychosis.Citation58

In order to replicate the finding and to determine whether working memory, beyond its mere association with the state of psychosis, might track uniquely with the accelerated decline in AD+P, we performed an analysis of cognitive data from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) study database. The ADNI database includes longitudinal cognitive data on subjects some of whom destined to become psychotic, as evidenced by psychometric ratings, over the course of the study. We hypothesized that digit-span performance, amongst available cognitive metrics, would decline more rapidly over time as a marker of the accelerated decline of AD+P. Results of the analysis demonstrated that digit-span performance is one of only two cognitive measures that decline more rapidly in AD+P when compared with AD-P.Citation59 It is not clear how impairment in working memory is etiologically related to AD+P. One possibility is that it is merely a byproduct of frontal degeneration, and not directly etiologically related; another is that accelerated frontal degeneration engenders working memory deficits, and that failures in working memory impair successful environmental surveillance in AD, culminating in misinterpretations and psychosis.

Evidence of a more aggressive cognitive phenotype as a marker of virulence would predict an acceleration of overall decline and decreased survival rates in those with AD+P. It is puzzling, given the established relationship between rate of cognitive decline and mortality in AD,Citation60,Citation61 that most,Citation53,Citation60Citation65 but not all,Citation66 early studies did not find an increased mortality rate in AD+P. More recent studies, with larger sample sizes and an enhanced follow-up with a more powerful statistical approach (Cox proportional hazard regression analysis) have consistently observed the predictive value of psychosis in AD survival.Citation67Citation70

A parsimonious hypothesis that explains the accelerated cognitive deterioration with some specificity and the early demise of patients with AD+P is that psychosis is a behavioral expression of a more-rapid neurodegenerative process that may be regional. If so, what regions are implicated? What are the neurochemical and neuropathological hallmarks of this distinct AD syndrome? Can answers to these questions point the way forward in developing new anti-AD+P treatments?

Imaging correlates in AD+P: a predominance of frontal localization

In the search for cerebral localization of deficit patterns in AD+P, investigators have, with varying levels of success related to sample sizes and heterogeneous subject composition, availed themselves of structural and functional neuroimaging. The literature on imaging in AD+P has been recently reviewed and thoughtfully organized by Murray et alCitation71 who separated the studies by methodological approach, including computed tomography, magnetic resonance imaging (MRI), single-photon emission computed tomography, and [18F]-fluorodeoxyglucose position emission tomography (PET) studies. As the authors highlight, with some variability and inconsistency, the preponderance of imaging evidence across methodologies implicate the frontal lobes.Citation71 A computed tomography study investigating regional brain atrophy in AD+P found that subjects with delusions demonstrated frontal and temporal patterns of asymmetric atrophy, compared with AD-P subjects.Citation72 MRI studies of AD+P have generally implicated the frontal cortex, with both orbitofrontal and medial frontal involvement.Citation73,Citation74 PET studies – with only one exceptionCitation75 – have consistently shown frontal hypometabolism in AD+P relative to AD-P.Citation76Citation80 In an analysis of a large, well-controlled sample drawn from the ADNI database, we observed a specific relationship between orbitofrontal hypometabolism and AD+P.Citation59

Neurotransmission in AD+P

An early theory promulgated in an effort to explain the prevalence of psychosis in AD suggested that a preservation of dopaminergic tone in the context of early cholinergic deficits may result in a relative “hyperdopaminergic” state, with resultant psychosis.Citation81 However evidence for a relative excess of dopaminergic tone has been limited. An early postmortem study of neurotransmission in psychotic AD did not identify any abnormalities in markers of dopamine signaling.Citation82 One small postmortem study suggested a selective increase in D3 receptor availability.Citation83 Further, using [11C]-raclopride PET, a modest increase in tracer binding in the striatum was observed in a cohort of delusional patients, suggesting some increase in D3 availability.Citation84 Yet, a recent comprehensive postmortem study of monoaminergic correlates of behavioral symptoms in AD did not observe any relationship between alterations in dopamine or dopamine metabolite concentrations in nine behaviorally and neurochemically relevant brain regions and psychotic symptoms in AD.Citation85

Perturbations in serotonergic systems have been reported in postmortem studies of psychotic AD. A reduction of serotonin (5-HT) has been reported in several studies across several brain regions,Citation82,Citation86 as have altered 5-HT6 receptor functionCitation87 and a reduction in cell counts in the dorsal raphe nucleus.Citation88 In a recent study, a significant reduction in serotonergic tone comprising lower 5-hydroxyindoleacetic acid concentrations in the hippocampus and lower 5-HT concentrations in the superior temporal gyrus and cerebral cortex, correlated with dementia severity.Citation85 It may be that the association observed in previous studies between psychosis in AD and reduced 5-HT may be related to regional disease severity in psychotic AD, and not an independent risk factor for the emergence of the syndrome.

Although currently available agents have not achieved satisfying rates of success in treating AD+P, continued exploration of perturbations in neurotransmitter systems in AD+P, together with the advent of more-sophisticated supportive therapeutics, may prove helpful in treating this condition.

Neuropathology in AD+P: a focus on tau

The classical protein pathologies in Alzheimer’s disease comprise extracellular amyloid-beta (Aβ) plaques and intracellular neurofibrillary tangles, composed primarily of hyperphosphorylated tau proteins. As AD+P is a more-rapidly progressing disease variant, with evidence of some localization of disease to the frontal cortex, neuropathological correlation studies are critical in the search for disease etiology. In general, evidence collected from neuropathological studies supports an association of AD+P with tau, rather than Aβ pathology.Citation44 This is not entirely surprising, since in AD distribution of tangle pathology, rather than Aβ pathology, correlates most closely with disease severity.Citation89 Postmortem studies consistently point to a heavier burden of tangle pathology in AD+P, with some localization in the frontal cortex.Citation82,Citation88,Citation90,Citation91

In order to explore tau pathology in AD+P, our group accessed the ADNI database to look for cerebrospinal fluid (CSF) evidence of increased tau in AD+P. Previous reports have linked elevations of CSF total tau with the postmortem burden of neurofibrillary tangles,Citation92,Citation93 so we approached the data with the hypothesis that we would find elevations in total tau in AD+P relative to AD-P as a reflection of tangle pathology. Indeed, we found that of the three core CSF AD biomarkers (Aβ, total tau, phospho-tau), only elevation of total tau in CSF was statistically significantly associated with psychosis in AD, driven by a robust association in females with AD+P.Citation94 In a contemporaneous postmortem immunohistochemical study of the frontal cortex conducted by colleagues in the laboratory of Dr Robert Sweet, hyperphosphorylated tau (the main component of neurofibrillary tangles)Citation95 was overabundant in subjects with AD+P compared with AD-P, strengthening the association of AD+P with tau.Citation96

Recently, in collaboration with Dr. Sweet, we conducted a biochemical analysis of phospho-tau epitopes in AD+P using a high-sensitivity enzyme-linked immunosorbent assay (ELISA) developed in the laboratory of Dr Peter Davies.Citation97 The ELISAs utilize highly specific monoclonal antibodies to phospho-tau epitopes that are representative of a range of tau pathology, from neuritic to pretangle to mature tangles.Citation98 Our experiments were conducted on postmortem tissue samples from the superior frontal gyri of 71 subjects. As α-synuclein pathology is known to contribute to a psychotic phenotype in dementia with Lewy bodies (DLB), we controlled for α-synuclein pathology.Citation99 We observed in women with AD+P, but not in men with AD+P, a robust association with tau pathology.Citation100 In females, while levels of nonpathogenic total tau as measured with DA31 antibody ELISA did not differ between AD+P and AD-P groups, pathogenic phospho-tau species did differ significantly. AD+P females had significantly higher levels of phosphorylated tau than did AD-P females at each epitope interrogated: Ser396/404 (PHF-1), Ser202 (CP13), Thr231 (RZ3), and Ser199/202/Thr205 (AT8). Males with AD+P, all of whom were not suffering with clinical DLB prior to death, nonetheless had an increase in α-synuclein pathology. In males this suggests that α-synuclein may make a unique contribution to AD+P, while in females tau pathology may be the source of vulnerability.

Previously conducted neuropathology studies have not been parsed by sex. There is evidence, however, that female sex is an independent risk factor for psychosis in AD.Citation30,Citation46,Citation101,Citation102 There is some support for biological underpinnings of this distinction from imaging studies. An MRI study in AD+P evidenced reduced frontotemporal cortical thickness in women, rather than in men, with AD+P.Citation74 Additionally, cerebral perfusion patterns in AD+P have been shown to be sex-specific.Citation103 The significance of the sex specificity of the association of AD+P with tau pathology is not yet clear, and more research is necessary to confirm this relationship.

Future directions: a pathology-guided approach

The published research in AD+P points toward an accelerated cognitive decline, with a particular burden of impairment affecting performance on frontal tasks. There is strong evidence that degeneration is accelerated in the frontal cortex, likely mediated by tau pathology, especially in females. This suggests a model of AD+P in which normative temporo–parietal deficits of AD are complicated by an accelerated extension to the frontal cortex. There may be some sex specificity to this model, and the degree to which the frontal tauopathy of AD+P is a female problem is not yet clear. However, the model, in identifying pathogenic proteins and localization, does enable translational research that may aid in illuminating in just what regions, and to what degree, tau pathology may contribute to the psychotic phenotype.

Animal models of psychiatric disease pose a particular challenge, in that the core of psychiatric illness is experience, something inaccessible in observations of animals that are language deficient. However, there is a long history of observing animals under conditions designed to serve as analogues of precipitants of human behavioral disorders and of studying physiological changes in these animals; a more recent history of observing animals with physiological changes known to be associated with human disease and of studying behavioral changes. In AD, translational research relies to a large degree on the advent of transgenic mice carrying autosomal dominant mutations in human genes that are known to cause the disease, or to cause pathology associated with the disease. The mice, depending on the identity of the mutation, produce an abundance of Aβ-driven plaque or tau-driven neurofibrillary tangle pathology and display dense cognitive deficits.Citation104 Curiously, to date, these models have only infrequently been employed to study the behavioral syndromes so common in AD. This raises interesting questions. Are there outcome measures in mice relevant to psychosis that could be utilized in such an exploration? If so, could extant mouse models be employed to elucidate the neuropathological substrate responsible for AD+P? If AD+P in humans emerges, in part, secondary to frontal neurodegeneration driven by tau, could a mouse model aid in a more-precise localization of the frontal deficits responsible?

Currently employed paradigms for modeling psychosis in mice include induction of locomotor hyperactivity and disruption of sensorimotor gating, quantified with prepulse inhibition (PPI) of acoustic startle.Citation105 Locomotor hyperactivity, as a model of psychosis in rodents resulting from exogenous agents promoting dopamine release, is the conceptual offspring of the hyperdopaminergic hypothesis of schizophrenia;Citation106 deficits of PPI represent an endophenotype of psychosis itself that has been associated with schizophrenia and may have broad relevance in all psychotic states.Citation107 PPI is defined as the reduction of the intensity of motor startle in response to a startle stimulus (for example, acoustic) when the stimulus is preceded by a diminished, nonstartle stimulus. In a normal state, the startle induced by the startle stimulus should be significantly gated by the attention paid to the preceding stimulus. In psychosis, this ability to inhibit startle is disrupted by a failure of normal gaiting, perhaps representing a diminished capacity to monitor environmental stimuli appropriately.Citation108 PPI of acoustic startle as a phenotypic marker of psychosis may be best described as “the interface of psychosis and cognition,”Citation105,Citation109 representing a cognitive failure that potentially influences environmental surveillance and may promote psychotic misinterpretation. The confluence of cognitive impairment and environmental misapprehension implicit in PPI deficiency makes PPI an attractive outcome measure in investigating potential animal models of AD+P.

In order to develop a preclinical mouse model of AD+P, we recently selected an AD model manifesting early frontal tauopathy, the transgenic rTg(tauP301L)4510 mouse,Citation110 and characterized PPI deficits over time in relationship to tau pathology.Citation111 We demonstrated that PPI deficits accrued over time in female rTg(tauP301L)4510 mice, and that these deficits were driven by the same pathogenic hyperphosphorylated tau molecules that we had previously observed in the frontal cortex of human female AD+P subjects.Citation100 This suggests that the rTg(tauP301L)4510 mouse may be a candidate for preclinical study of AD+P biology and the development of novel therapeutics. There is more work to be done prior to settling on any animal model of AD+P. Future studies involving this model will include expanding the phenotypic characterization of this mouse beyond PPI to include other cognitive and behavioral markers of psychosis, and exploring the sex specificity of the observed behavioral characteristics.

The way forward in the treatment of AD+P may transcend traditional psychiatric treatments of psychosis and may involve the reduction of pathogenic proteins rather than supporting aberrant neurotransmitter systems that are the downstream effects of disease pathology. Future treatments may focus on tau pathology, α-synuclein pathology, or both. Several tau reduction agents are currently in the drug development stage for the treatment of AD, and even if they do not provide a cure for the primary illness, they warrant exploration in the service of combating a particularly morbid and common manifestation of the disease.

Disclosure

The authors report no conflicts of interest in this work.

References

  • AarslandDCummingsJLYennerGMillerBRelationship of aggressive behavior to other neuropsychiatric symptoms in patients with Alzheimer’s diseaseAm J Psychiatry199615322432478561206
  • DeutschLHBylsmaFWRovnerBWSteeleCFolsteinMFPsychosis and physical aggression in probable Alzheimer’s diseaseAm J Psychiatry19911489115911631882992
  • KunikMESnow-TurekALIqbalNContribution of psychosis and depression to behavioral disturbances in geropsychiatric inpatients with dementiaJ Gerontol A Biol Sci Med Sci1999543M157M16110191845
  • KnopmanDSKittoJDeinardSHeiringJLongitudinal study of death and institutionalization in patients with primary degenerative dementiaJ Am Geriatr Soc19883621081123339214
  • O’DonnellBFDrachmanDABarnesHJPetersonKESwearerJMLewRAIncontinence and troublesome behaviors predict institutionalization in dementiaJ Geriatr Psychiatry Neurol19925145521571074
  • GilleyDWWilsonRSBeckettLAEvansDAPsychotic symptoms and physically aggressive behavior in Alzheimer’s diseaseJ Am Geriatr Soc1997459107410799288014
  • MaHHuangYCongZThe efficacy and safety of atypical antipsychotics for the treatment of dementia: a meta-analysis of randomized placebo-controlled trialsJ Alzheimers Dis201442391593725024323
  • OverallJEGorhamDRThe Brief Psychiatric Rating ScalePsychological Report196210799812
  • RhoadesHMOverallJEThe semistructured BPRS interview and rating guidePsychopharmacol Bull19882411011043290934
  • StreetJSClarkWSGannonKSOlanzapine treatment of psychotic and behavioral symptoms in patients with Alzheimer disease in nursing care facilities: a double-blind, randomized, placebo-controlled trial. The HGEU Study GroupArch Gen Psychiatry2000571096897611015815
  • De DeynPPCarrascoMMDeberdtWOlanzapine versus placebo in the treatment of psychosis with or without associated behavioral disturbances in patients with Alzheimer’s diseaseInt J Geriatr Psychiatry200419211512614758577
  • SultzerDLDavisSMTariotPNCATIE-AD Study GroupClinical symptom responses to atypical antipsychotic medications in Alzheimer’s disease: phase 1 outcomes from the CATIE-AD effectiveness trialAm J Psychiatry2008165784485418519523
  • De DeynPJesteDVSwaninkRAripiprazole for the treatment of psychosis in patients with Alzheimer’s disease: a randomized, placebo-controlled studyJ Clin Psychopharmacol200525546346716160622
  • MintzerJETuneLEBrederCDAripiprazole for the treatment of psychoses in institutionalized patients with Alzheimer dementia: a multicenter, randomized, double-blind, placebo-controlled assessment of three fixed dosesAm J Geriatr Psychiatry2007151191893117974864
  • TariotPNSchneiderLKatzIRQuetiapine treatment of psychosis associated with dementia: a double-blind, randomized, placebo-controlled clinical trialAm J Geriatr Psychiatry200614976777616905684
  • KatzIRJesteDVMintzerJEClydeCNapolitanoJBrecherMComparison of risperidone and placebo for psychosis and behavioral disturbances associated with dementia: a randomized, double-blind trial. Risperidone Study GroupJ Clin Psychiatry199960210711510084637
  • De DeynPPRabheruKRasmussenAA randomized trial of risperidone, placebo, and haloperidol for behavioral symptoms of dementiaNeurology199953594695510496251
  • BrodatyHAmesDSnowdonJA randomized placebo-controlled trial of risperidone for the treatment of aggression, agitation, and psychosis of dementiaJ Clin Psychiatry200364213414312633121
  • MintzerJGreenspanACaersIRisperidone in the treatment of psychosis of Alzheimer disease: results from a prospective clinical trialAm J Geriatr Psychiatry200614328029116505133
  • KatzIde DeynPPMintzerJGreenspanAZhuYBrodatyHThe efficacy and safety of risperidone in the treatment of psychosis of Alzheimer’s disease and mixed dementia: a meta-analysis of 4 placebo-controlled clinical trialsInt J Geriatr Psychiatry200722547548417471598
  • SultzerDLPsychosis and antipsychotic medications in Alzheimer’s disease: clinical management and research perspectivesDement Geriatr Cogn Disord2004171–2789014566100
  • FlintAJDelusions in dementia: a reviewJ Neuropsychiatry Clin Neurosci1991321211301687962
  • SultzerDLLevinHSMahlerMEHighWMCummingsJLAssessment of cognitive, psychiatric, and behavioral disturbances in patients with dementia: the Neurobehavioral Rating ScaleJ Am Geriatr Soc19924065495551587970
  • PaulsenJSSalmonDPThalLJIncidence of and risk factors for hallucinations and delusions in patients with probable ADNeurology200054101965197110822438
  • DevanandDPJacobsDMTangMXThe course of psychopathologic features in mild to moderate Alzheimer diseaseArch Gen Psychiatry19975432572639075466
  • MegaMSCummingsJLFiorelloTGornbeinJThe spectrum of behavioral changes in Alzheimer’s diseaseNeurology19964611301358559361
  • LyketsosCGSteinbergMTschanzJTNortonMCSteffensDCBreitnerJCMental and behavioral disturbances in dementia: findings from the Cache County Study on Memory in AgingAm J Psychiatry2000157570871410784462
  • SweetRANimgaonkarVLDevlinBLopezOLDeKoskySTIncreased familial risk of the psychotic phenotype of Alzheimer diseaseNeurology200258690791111914406
  • BacanuSADevlinBChowdariKVDeKoskySTNimgaonkarVLSweetRAHeritability of psychosis in Alzheimer diseaseAm J Geriatr Psychiatry200513762462716009739
  • SchneiderLSDagermanKSPsychosis of Alzheimer’s disease: clinical characteristics and historyJ Psychiatr Res200438110511114690773
  • CermolacceMSassLParnasJWhat is bizarre in bizarre delusions? A critical reviewSchizophr Bull201036466767920142381
  • ReevesSJGouldRLPowellJFHowardRJOrigins of delusions in Alzheimer’s diseaseNeurosci Biobehav Rev201236102274228722910677
  • CookSEMiyaharaSBacanuSAPsychotic symptoms in Alzheimer disease: evidence for subtypesAm J Geriatr Psychiatry200311440641312837669
  • BurnsAJacobyRLevyRPsychiatric phenomena in Alzheimer’s disease. I: disorders of thought contentBr J Psychiatry1990157727692942397365
  • ReisbergBBorensteinJSalobSPFerrisSHFranssenEGeorgotasABehavioral symptoms in Alzheimer’s disease: phenomenology and treatmentJ Clin Psychiatry198748Suppl9153553166
  • RubinEHDrevetsWCBurkeWJThe nature of psychotic symptoms in senile dementia of the Alzheimer typeJ Geriatr Psychiatry Neurol19881116203266997
  • BurnsAJacobyRLevyRPsychiatric phenomena in Alzheimer’s disease. II: disorders of perceptionBr J Psychiatry1990157768192942397366
  • MerriamAEAronsonMKGastonPWeySLKatzIThe psychiatric symptoms of Alzheimer’s diseaseJ Am Geriatr Soc19883617123335733
  • GormleyNRizwanMRPrevalence and clinical correlates of psychotic symptoms in Alzheimer’s diseaseInt J Geriatr Psychiatry19981364104149658277
  • HwangJPYangCHTsaiSJPhantom boarder symptom in dementiaInt J Geriatr Psychiatry200318541742012766918
  • VenneriAShanksMFStaffRTDella SalaSNurturing syndrome: a form of pathological bereavement with delusions in Alzheimer’s diseaseNeuropsychologia200038221322410660231
  • RabinsPVMaceNLLucasMJThe impact of dementia on the familyJAMA198224833333357087127
  • JesteDVFinkelSIPsychosis of Alzheimer’s disease and related dementias. Diagnostic criteria for a distinct syndromeAm J Geriatr Psychiatry200081293410648292
  • MurrayPSKumarSDemichele-SweetMASweetRAPsychosis in Alzheimer’s diseaseBiol Psychiatry201475754255224103379
  • CummingsJLMillerBHillMANeshkesRNeuropsychiatric aspects of multi-infarct dementia and dementia of the Alzheimer typeArch Neurol19874443893933827694
  • HironoNMoriEYasudaMFactors associated with psychotic symptoms in Alzheimer’s diseaseJ Neurol Neurosurg Psychiatry19986456486529598682
  • RopackiSAJesteDVEpidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003Am J Psychiatry2005162112022203016263838
  • BerriosGENon-cognitive symptoms and the diagnosis of dementia. Historical and clinical aspectsBr J Psychiatry Suppl1989411162690875
  • MalloyPFRichardsonEDThe frontal lobes and content-specific delusionsJ Neuropsychiatry Clin Neurosci1994644554667841816
  • ShakhmatovNFOn psychotic pictures observed during the course of Alzheimer’s disease and Pick’s diseaseZh Nevropatol Psikhiatr Im S S Korsakova196464265269 Russian14140453
  • MayeuxRSternYSpantonSHeterogeneity in dementia of the Alzheimer type: evidence of subgroupsNeurology19853544534613982631
  • SternYMayeuxRSanoMHauserWABushTPredictors of disease course in patients with probable Alzheimer’s diseaseNeurology19873710164916533658173
  • DrevetsWCRubinEHPsychotic symptoms and the longitudinal course of senile dementia of the Alzheimer typeBiol Psychiatry198925139482912509
  • RosenJZubenkoGSEmergence of psychosis and depression in the longitudinal evaluation of Alzheimer’s diseaseBiol Psychiatry19912932242322015329
  • WilsonRSGilleyDWBennettDABeckettLAEvansDAHallucinations, delusions, and cognitive decline in Alzheimer’s diseaseJ Neurol Neurosurg Psychiatry200069217217710896689
  • EmanuelJELopezOLHouckPRTrajectory of cognitive decline as a predictor of psychosis in early Alzheimer disease in the cardiovascular health studyAm J Geriatr Psychiatry201119216016820808116
  • JesteDVWraggRESalmonDPHarrisMJThalLJCognitive deficits of patients with Alzheimer’s disease with and without delusionsAm J Psychiatry199214921841891734737
  • KoppelJGoldbergTEGordonMLRelationships between behavioral syndromes and cognitive domains in Alzheimer disease: the impact of mood and psychosisAm J Geriatr Psychiatry20122011994100022048323
  • KoppelJSundaySGoldbergTEDaviesPChristenEGreenwaldBSAlzheimer’s Disease Neuroimaging InitiativePsychosis in Alzheimer’s disease is associated with frontal metabolic impairment and accelerated decline in working memory: findings from the Alzheimer’s Disease Neuroimaging InitiativeAm J Geriatr Psychiatry201422769870723672944
  • HuiJSWilsonRSBennettDABieniasJLGilleyDWEvansDARate of cognitive decline and mortality in Alzheimer’s diseaseNeurology200361101356136114638955
  • HeymanAWilkinsonWEHurwitzBJEarly-onset Alzheimer’s disease: clinical predictors of institutionalization and deathNeurology19873769809843587649
  • BurnsALewisGJacobyRLevyRFactors affecting survival in Alzheimer’s diseasePsychol Med19912123633701876641
  • LopezOLWisniewskiSRBeckerJTBollerFDeKoskySTPsychiatric medication and abnormal behavior as predictors of progression in probable Alzheimer diseaseArch Neurol199956101266127210520944
  • SamsonWNvan DuijnCMHopWCHofmanAClinical features and mortality in patients with early-onset Alzheimer’s diseaseEur Neurol19963621031068654478
  • SternYTangMXAlbertMSPredicting time to nursing home care and death in individuals with Alzheimer diseaseJAMA1997277108068129052710
  • MoritzDJFoxPJLuscombeFAKraemerHCNeurological and psychiatric predictors of mortality in patients with Alzheimer disease in CaliforniaArch Neurol19975478788859236577
  • ScarmeasNBrandtJAlbertMDelusions and hallucinations are associated with worse outcome in Alzheimer diseaseArch Neurol200562101601160816216946
  • WilsonRSKruegerKRKamenetskyJMHallucinations and mortality in Alzheimer diseaseAm J Geriatr Psychiatry2005131198499016286442
  • Vilalta-FranchJLópez-PousaSCalvó-PerxasLGarre-OlmoJPsychosis of Alzheimer disease: prevalence, incidence, persistence, risk factors, and mortalityAm J Geriatr Psychiatry201321111135114323567368
  • LopezOLBeckerJTChangYFThe long-term effects of conventional and atypical antipsychotics in patients with probable Alzheimer’s diseaseAm J Psychiatry201317091051105823896958
  • MurrayPSKumarSDemichele-SweetMASweetRAPsychosis in Alzheimer’s diseaseBiol Psychiatry201475754255224103379
  • GeroldiCBrescianiLZanettiOFrisoniGBRegional brain atrophy in patients with mild Alzheimer’s disease and delusionsInt Psychogeriatr200214436537812670058
  • BruenPDMcGeownWJShanksMFVenneriANeuroanatomical correlates of neuropsychiatric symptoms in Alzheimer’s diseaseBrain2008131Pt 92455246318669506
  • WhiteheadDTunnardCHurtCAddNeuroMed ConsortiumFrontotemporal atrophy associated with paranoid delusions in women with Alzheimer’s diseaseInt Psychogeriatr20122419910721740613
  • HironoNMoriEIshiiKAlteration of regional cerebral glucose utilization with delusions in Alzheimer’s diseaseJ Neuropsychiatry Clin Neurosci19981044334399813789
  • MentisMJWeinsteinEAHorwitzBAbnormal brain glucose metabolism in the delusional misidentification syndromes: a positron emission tomography study in Alzheimer diseaseBiol Psychiatry19953874384498672604
  • SultzerDLMahlerMEMandelkernMAThe relationship between psychiatric symptoms and regional cortical metabolism in Alzheimer’s diseaseJ Neuropsychiatry Clin Neurosci1995744764848555751
  • LopezOLSmithGBeckerJTMeltzerCCDeKoskySTThe psychotic phenomenon in probable Alzheimer’s disease: a positron emission tomography studyJ Neuropsychiatry Clin Neurosci2001131505511207329
  • GradyCLHaxbyJVSchapiroMBSubgroups in dementia of the Alzheimer type identified using positron emission tomographyJ Neuropsychiatry Clin Neurosci1990243733842136389
  • SultzerDLBrownCVMandelkernMADelusional thoughts and regional frontal/temporal cortex metabolism in Alzheimer’s diseaseAm J Psychiatry2003160234134912562582
  • CummingsJLBackCThe cholinergic hypothesis of neuropsychiatric symptoms in Alzheimer’s diseaseAm J Geriatr Psychiatry199862 Suppl 1S64S789581223
  • ZubenkoGSMoossyJMartinezAJNeuropathologic and neurochemical correlates of psychosis in primary dementiaArch Neurol19914866196241710105
  • SweetRAHamiltonRLHealyMTAlterations of striatal dopamine receptor binding in Alzheimer disease are associated with Lewy body pathology and antemortem psychosisArch Neurol200158346647211255451
  • ReevesSBrownRHowardRGrasbyPIncreased striatal dopamine (D2/D3) receptor availability and delusions in Alzheimer diseaseNeurology200972652853419204262
  • VermeirenYVan DamDAertsTEngelborghsSDe DeynPPBrain region-specific monoaminergic correlates of neuropsychiatric symptoms in Alzheimer’s diseaseJ Alzheimers Dis201441381983324685637
  • Garcia-AllozaMGil-BeaFJDiez-ArizaMCholinergic-serotonergic imbalance contributes to cognitive and behavioral symptoms in Alzheimer’s diseaseNeuropsychologia200543344244915707619
  • MarcosBGarcía-AllozaMGil-BeaFJInvolvement of an altered 5-HT-{6} receptor function in behavioral symptoms of Alzheimer’s diseaseJ Alzheimers Dis2008141435018525126
  • FörstlHBurnsALevyRCairnsNNeuropathological correlates of psychotic phenomena in confirmed Alzheimer’s diseaseBr J Psychiatry1994165153597953058
  • ArriagadaPVGrowdonJHHedley-WhyteETHymanBTNeurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer’s diseaseNeurology1992423 Pt 16316391549228
  • ZubenkoGSMolecular neurobiology of Alzheimer’s disease (syndrome?)Harv Rev Psychiatry1997541772139427013
  • FarberNBRubinEHNewcomerJWIncreased neocortical neurofibrillary tangle density in subjects with Alzheimer disease and psychosisArch Gen Psychiatry200057121165117311115331
  • ClarkCMXieSChittamsJCerebrospinal fluid tau and beta-amyloid: how well do these biomarkers reflect autopsy-confirmed dementia diagnoses?Arch Neurol200360121696170214676043
  • TapiolaTOvermyerMLehtovirtaMThe level of cerebrospinal fluid tau correlates with neurofibrillary tangles in Alzheimer’s diseaseNeuroreport1997818396139639462474
  • KoppelJSundaySButhornJGoldbergTDaviesPGreenwaldBAlzheimer’s Disease Neuroimaging InitiativeElevated CSF Tau is associated with psychosis in Alzheimer’s diseaseAm J Psychiatry2013170101212121324084821
  • CrowtherRAOlesenOFJakesRGoedertMThe microtubule binding repeats of tau protein assemble into filaments like those found in Alzheimer’s diseaseFEBS Lett199230921992021505683
  • MurrayPSKirkwoodCMGrayMCHyperphosphorylated tau is elevated in Alzheimer’s disease with psychosisJ Alzheimers Dis201439475977324270207
  • AckerCMForestSKZinkowskiRDaviesPd’AbramoCSensitive quantitative assays for tau and phospho-tau in transgenic mouse modelsNeurobiol Aging201334133835022727277
  • AugustinackJCSchneiderAMandelkowEMHymanBTSpecific tau phosphorylation sites correlate with severity of neuronal cytopathology in Alzheimer’s diseaseActa Neuropathol20021031263511837744
  • McKeithIGDicksonDWLoweJet al; Consortium onDLBDiagnosis and management of dementia with Lewy bodies: third report of the DLB ConsortiumNeurology200565121863187216237129
  • KoppelJAckerCDaviesPPsychotic Alzheimer’s disease is associated with gender-specific tau phosphorylation abnormalitiesNeurobiol Aging20143592021202824731519
  • RockwellEKrullAJDimsdaleJJesteDVLate-onset psychosis with somatic delusionsPsychosomatics199435166728134531
  • LeroiIVoulgariABreitnerJCLyketsosCGThe epidemiology of psychosis in dementiaAm J Geriatr Psychiatry2003111839112527543
  • MoranEKBeckerJASatlinALyooIKFischmanAJJohnsonKAPsychosis of Alzheimer’s disease: Gender differences in regional perfusionNeurobiol Aging20082981218122517408808
  • LaFerlaFMGreenKNAnimal models of Alzheimer diseaseCold Spring Harb Perspect Med2012211
  • van den BuuseMModeling the positive symptoms of schizophrenia in genetically modified mice: pharmacology and methodology aspectsSchizophr Bull201036224627019900963
  • LaruelleMAbi-DarghamAGilRKegelesLInnisRIncreased dopamine transmission in schizophrenia: relationship to illness phasesBiol Psychiatry1999461567210394474
  • BraffDStoneCCallawayEGeyerMGlickIBaliLPrestimulus effects on human startle reflex in normals and schizophrenicsPsychophysiology1978154339343693742
  • OuagazzalAMJenckFMoreauJLDrug-induced potentiation of prepulse inhibition of acoustic startle reflex in mice: a model for detecting antipsychotic activity?Psychopharmacology (Berl)20011562–327328311549229
  • DesbonnetLWaddingtonJLO’TuathaighCMMutant models for genes associated with schizophreniaBiochem Soc Trans200937Pt 130831219143653
  • SantacruzKLewisJSpiresTTau suppression in a neurodegenerative mouse model improves memory functionScience2005309573347648116020737
  • KoppelJJimenezHAzoseMPathogenic tau species drive a psychosis-like phenotype in a mouse model of Alzheimer’s diseaseBehav Brain Res2014275C273325151619