Hostname: page-component-848d4c4894-ttngx Total loading time: 0 Render date: 2024-06-12T11:00:34.345Z Has data issue: false hasContentIssue false

Dementia-related psychosis and the potential role for pimavanserin

Published online by Cambridge University Press:  19 August 2020

Jeffery L. Cummings*
Affiliation:
Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada at Las Vegas (UNLV) and Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, Nevada, USA
D. P. Devanand
Affiliation:
Department of Psychiatry, Columbia University Medical Center, New York, New York, USA
Stephen M. Stahl
Affiliation:
Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
*
*Author for correspondence: Jeffery L. Cummings, Email: jcummings@cnsinnovations.com
Rights & Permissions [Opens in a new window]

Abstract

Dementia-related psychosis (DRP) is prevalent across dementias and typically manifests as delusions and/or hallucinations. The mechanisms underlying psychosis in dementia are unknown; however, neurobiological and pharmacological evidence has implicated multiple signaling pathways and brain regions. Despite differences in dementia pathology, the neurobiology underlying psychosis appears to involve dysregulation of a cortical and limbic pathway involving serotonergic, gamma-aminobutyric acid ergic, glutamatergic, and dopaminergic signaling. Thus, an imbalance in cortical and mesolimbic excitatory tone may drive symptoms of psychosis. Delusions and hallucinations may result from (1) hyperactivation of pyramidal neurons within the visual cortex, causing visual hallucinations and (2) hyperactivation of the mesolimbic pathway, causing both delusions and hallucinations. Modulation of the 5-HT2A receptor may mitigate hyperactivity at both psychosis-associated pathways. Pimavanserin, an atypical antipsychotic, is a selective serotonin inverse agonist/antagonist at 5-HT2A receptors. Pimavanserin may prove beneficial in treating the hallucinations and delusions of DRP without worsening cognitive or motor function.

Type
Perspective
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution, and reproduction in any medium, provided the original work is properly cited.
Copyright
© The Author(s), 2020. Published by Cambridge University Press

Introduction

No pharmacological agents are approved by the U.S. Food and Drug Administration (FDA) to treat dementia-related psychosis (DRP). Pimavanserin, an atypical antipsychotic that acts as a selective serotonin inverse agonist/antagonist at 5-HT2A receptors (and to a lesser extent, at 5-HT2c receptors), is the only FDA-approved treatment for hallucinations and delusions associated with Parkinson’s disease (PD) psychosis.1, Reference Cummings, Isaacson and Mills2 A phase 2 study of pimavanserin in Alzheimer’s disease (AD) psychosis met its primary end point at week 6 (mean change in the Neuropsychiatric Inventory-Nursing Home version psychosis score of −3.76 points [SE 0.65] for pimavanserin and −1.93 points [SE 0.63] for placebo; P =0 .045) with an acceptable tolerability profile and no worsening of cognition or motor function.Reference Ballard, Banister and Khan3 Pimavanserin is being investigated (NCT03325556 and 2017-002227-13) for treating hallucinations and delusions associated with DRP across five common neurodegenerative dementias: AD dementia, PD dementia, dementia with Lewy bodies (DLB), frontotemporal dementia (FTD), and vascular dementia (VaD).4-6 This article discusses the emerging understanding of the neurobiology of psychosis, the current state of knowledge about the neurobiology of psychotic symptoms in dementia syndromes, and the hypothesized role of pimavanserin in treating DRP.

Approximately 7.9 million people in the United States have dementia, with this number projected to rise as the elderly population grows.7-9 Although AD dementia is most common in the United States (60%-80% of cases), other forms of dementia include PD dementia, DLB, FTD, and VaD. While distinctive features characterize each form of dementia, broad symptom overlap is observed.10

Dementia is a syndrome characterized by a decline in one or more cognitive domains (eg, memory, language, executive function, problem-solving, attention, and social cognition) sufficiently severe to compromise daily function.11 Besides the well-recognized cognitive deficits characteristic of dementia, noncognitive symptoms (often referred to as behavioral and psychological symptoms of dementia or neuropsychiatric symptoms [NPS]) are estimated to occur in up to 90% of individuals during the course of dementia. NPS include behavioral symptoms (such as agitation, aggression, disinhibition, elation, and irritability), aberrant motor behavior, anxiety, apathy, appetite changes, depression, sleep disturbances, and symptoms of psychosis.Reference Cerejeira, Lagarto and Mukaetova-Ladinska12

Although some dementia patients display no or only a few noncognitive symptoms, multiple NPS commonly occur simultaneously.Reference Cerejeira, Lagarto and Mukaetova-Ladinska12 The Agitation Definition Work Group of the International Psychogeriatric Association defines agitation as occurring in patients with a cognitive impairment or dementia syndrome; exhibiting behavior consistent with emotional distress; manifesting excessive motor activity, verbal aggression, or physical aggression; and evidencing behaviors that cause excess disability and are not solely attributable to another disorder.Reference Cummings, Mintzer and Brodaty13 In contrast, DRP is defined by delusions or hallucinations occurring after the onset of cognitive decline, persisting for at least 1 month, and not better explained by delirium or some other mental illness.11 Both hallucinations and delusions are associated with behavioral symptoms such as physical and verbal aggression in patients with dementia.14Reference Lopez, Becker and Sweet16

Prevalence estimates for psychosis range from 10% for FTD to 75% for DLB (Table 1).16Reference Mourik, Rosso and Niermeijer30 In the United States, an estimated 2.34 million people suffer from DRP.16Reference Mourik, Rosso and Niermeijer30 Visual hallucinations occur in all forms of dementia and are commonly observed in PD dementia and DLB.Reference Fenelon, Soulas, Zenasni and Cleret de Langavant31, Reference McKeith, Boeve and Dickson32 Delusions are also observed in all forms of dementia, most commonly paranoid delusions (eg, theft or spousal infidelity) and misidentifications, though the latter is sometimes considered a type of memory deficit rather than psychosis.Reference Fenelon, Soulas, Zenasni and Cleret de Langavant31, Reference Devanand, Jacobs and Tang33 DRP varies within and across patients in the psychotic symptoms manifested and in the severity of symptoms during the course of illness.Reference Devanand, Jacobs and Tang33 DRP is more frequently observed in patients with more advanced dementia. DRP has consistently been associated with greater caregiver burden and more rapid progression to severe dementia, institutionalization, and death.34Reference Scarmeas, Brandt and Albert38

Table 1. Prevalence ranges for psychosis, delusions, and hallucinations in AD dementia, VaD, DLB, PD Dementia, and FTD

Many forms of dementia, aside from VaD, result from neurodegenerative disease and are associated with various proteinopathies characterized by protein misfolding and aggregation. Aberrant aggregated proteins in AD produce β-amyloid plaques and neurofibrillary tangles (hyperphosphorylated tau aggregates).Reference Rovelet-Lecrux, Hannequin and Raux39, Reference Hyman, Phelps and Beach40 In DLB and PD dementia, Lewy bodies and Lewy neurites, composed primarily of phosphorylated α-synuclein aggregates, accumulate preferentially in limbic brain regions.41Reference Hamilton43 FTD may be associated with either aggregated tau protein or aggregated trans-activator regulatory DNA-binding 43 (TDP-43) protein.Reference Nelson, Dickson and Trojanowski44

Post-mortem analyses have revealed most dementia patients exhibit mixed pathology comprising abnormal protein aggregates plus vascular changes.Reference Brenowitz, Keene and Hawes45, Reference Schneider, Arvanitakis, Bang and Bennett46 In one study of community-dwelling adults, 56% of dementia patients were diagnosed with multiple underlying pathologies (AD in combination with either PD/DLB, infarctions representing vascular brain injury, or both).Reference Schneider, Arvanitakis, Bang and Bennett46 After adjusting for age, individuals with multiple diagnoses were deemed to be nearly three times more likely to develop dementia as those with a single underlying pathology.Reference Schneider, Arvanitakis, Bang and Bennett46 In a separate study, database analyses revealed that 59% to 68% of patients with AD neuropathology also displayed Lewy body pathology or vascular brain injury.Reference Brenowitz, Keene and Hawes45

Neurobiology of Psychosis

The underlying mechanisms behind psychosis in dementia are unknown. The neurobiology of psychosis has primarily been examined in studies of patients with schizophrenia and animals investigated in pharmacological probe paradigms.Reference Gründer and Cumming47 Taken together, the evidence suggests alterations in multiple signaling pathways—including dopaminergic, gamma-aminobutyric acid (GABA)ergic, glutamatergic, and serotonergic neurotransmission—may contribute to psychosis.Reference Hirvonen and Hietala48 Excess dopamine signaling in the mesolimbic pathway, which projects from the ventral tegmental area (VTA) to the nucleus accumbens, has been demonstrated to promote positive symptoms, primarily delusions and hallucinations.Reference Watanabe, Morimoto, Nakamura and Suwaki49 Under basal conditions, GABAergic interneurons provide inhibitory regulation of the activity of cortical pyramidal neurons. Inhibition of N-methyl-d-aspartate (NMDA) receptors reduces the inhibitory activity of GABAergic interneurons, resulting in glutamatergic hyperfunction. In rodent models, NMDA receptor antagonism or cortical and hippocampal NMDA receptor deletion results in psychotic-like behaviors.Reference Zhou, Zhang and Li50, Reference Nakazawa, Zsiros and Jiang51 Similarly, ketamine-induced NMDA antagonism significantly increases positive symptoms in haloperidol-treated patients diagnosed with schizophrenia.Reference Lahti, Koffel, LaPorte and Tamminga52 Glutamatergic projections from the prefrontal cortex provide tonic control of dopaminergic neurons in the VTA. Microdialysis and electrical stimulation rodent models reveal that glutamatergic hyperfunction increases burst firing of dopaminergic neurons in the VTA, stimulating the mesolimbic pathway.Reference Almodovar-Fabregas, Segarra and Colon53, Reference Karreman and Moghaddam54

These findings are supported by basic pharmacological intervention studies showing that multiple signaling pathways contribute to psychosis. These observations include dopamine D2 receptor activation via psychostimulants, glutamate NMDA receptor inhibition via dissociative anesthetics, and serotonin 5-HT2A receptor activation via psychedelics, all of which have been reported to precipitate psychotic symptoms.Reference Rolland, Jardri and Amad55 Methamphetamine, which is a substrate for the dopamine transporter (DAT), increases the firing rate of cultured rat dopamine neurons, triggering an excitatory response at dopamine concentrations lower than those required for D2 autoreceptor activation.Reference Ingram, Prasad and Amara56 Both methamphetamine and the DAT inhibitor cocaine, which increases extracellular dopamine levels via uptake inhibition, induce paranoid delusions as well as auditory and tactile hallucinations in stimulant-dependent individuals.Reference Mahoney, Kalechstein, De La Garza and Newton57, Reference McKetin, Baker, Dawe, Voce and Lubman58 Ketamine, a noncompetitive NMDA receptor antagonist, significantly increases hallucinations and delusions in schizophrenic patients following administration of subanesthetic doses and increases regional cerebral blood flow to the anterior cingulate cortex, while decreasing blood flow to the hippocampus and primary visual cortex.Reference Lahti, Holcomb, Medoff and Tamminga59, Reference Krystal, Karper and Seibyl60 In addition, subanesthetic doses of ketamine induces acute auditory verbal hallucinations and paranoid delusions in control subjects without psychosis in a reduced-stimulation environment.Reference Powers, Gancsos, Finn, Morgan and Corlett61 Administration of the serotonin 5-HT2A receptor agonist lysergic acid diethylamide (LSD) to healthy volunteers results in increased cerebral blood flow and enhanced resting-state functional connectivity in the visual cortex, which strongly correlates with visual hallucinations.Reference Carhart-Harris, Muthukumaraswamy and Roseman62 Administration of the 5-HT2A receptor agonist psilocybin to hallucinogen-naïve adults precipitates mystical delusions that persist at 14-month follow-up in 60% of subjects.Reference Griffiths, Richards, Johnson, McCann and Jesse63 In a separate study in healthy human volunteers, psychotic symptoms that closely mimicked those observed in first-episode schizophrenic patients (including sensory misperceptions and thought-process disruption) occur within 20 to 30 minutes of psilocybin administration. Pretreatment with the 5-HT2A receptor antagonist ketanserin inhibits psilocybin-induced psychosis in a dose-dependent manner, while pretreatment with the D2 receptor antagonist haloperidol does not affect psilocybin-induced hallucinations, further supporting the idea that certain symptoms of hallucinogen-induced psychosis (such as visual hallucinations) result from 5-HT2A agonism, while others (such as auditory hallucinations) are more strongly linked to the D2 receptor.Reference Vollenweider, Vollenweider-Scherpenhuyzen, Babler, Vogel and Hell64 Additional support for multifactorial signaling stems from the observation that conventional antipsychotics exert their effects primarily via D2 receptor inhibition, while atypical antipsychotics act as serotonin–dopamine antagonists, inhibiting both D2 and 5-HT2A receptors.Reference Stahl65

Taken together, neurobiological and pharmacological evidence points to a common, interconnected cortical–limbic psychosis pathway (Figure 1). Both the occurrence of hallucinogen 5-HT2A receptor effects in the prefrontal and visual cortices, and the observation that loss of serotonin nerve terminals in the prefrontal and visual cortices of patients with PD psychosis leads to upregulation of 5-HT2A receptors in the cortex support this hypothesis. A possible convergence of many of these pathways into 5-HT2A-modulated systems is suggested by the observation that the 5-HT2A receptor is affected by essentially all atypical antipsychotics, which led to the development of pimavanserin, a 5-HT2A-selective inverse agonist/antagonist.Reference Hacksell, Burstein, McFarland, Mills and Williams66 In preclinical rodent studies, pimavanserin reduced both amphetamine- and NMDA receptor antagonist-induced hyperactivity when combined with haloperidol or haloperidol and risperidone, respectively.Reference Gardell, Vanover and Pounds67 Additionally, pimavanserin was shown to reverse psychosis-like behaviors in rodent models of PD psychosis without impairing motor performance or interfering with the efficacy of PD medications.Reference McFarland, Price and Bonhaus68 Finally, pimavanserin increased dopamine release in the medial prefrontal cortex but not in the nucleus accumbens, suggesting pimavanserin may be beneficial for cognitive, negative, and psychotic symptoms.Reference Li, Ichikawa and Huang69

Figure 1. Hypothesized cortical–limbic psychosis pathway and proposed mechanism of disease for DRP. Neurobiological and pharmacological evidence suggests that hallucinations and delusions are precipitated by overactivation of the mesolimbic pathway, while visual hallucinations are mediated via overactivation of the visual cortex. Dissociative anesthetic-induced (ie, PCP, ketamine) glutamate NMDA receptor antagonism, psychedelic-induced (ie, LSD, psilocybin) serotonin 5-HT2A receptor agonism, and psychostimulant-induced (ie, amphetamine, cocaine) dopamine D2 receptor agonism/DAT antagonism have all been reported to precipitate hallucinations and delusions. In contrast, antipsychotic-mediated D2 and 5-HT2A antagonism treat both hallucinations and delusions. GABAergic interneuron or NMDA receptor dysfunction, and excess serotonin or 5-HT2A receptor upregulation in the cerebral cortex can result in sustained activation of pyramidal neurons and may lead to hyperactive glutamatergic signaling to the VTA, resulting in excess dopamine or D2 receptor upregulation in the ventral striatum, triggering hallucinations and delusions in DRP.Reference Rolland, Jardri and Amad55Reference Ingram, Prasad and Amara56, Reference Kometer, Schmidt, Jancke and Vollenweider70Reference Braak and Braak87

Changes in Dementia that may Result in Psychosis Pathway Dysfunction

The cortical–limbic psychosis pathway provides numerous points where dysfunction could trigger delusions and/or hallucinations. The pathology and dysfunction across the dementias leading to delusions and/or hallucinations may be different in specific dementias but each is positioned to affect the function of the cortical–limbic system thought to mediate psychosis. The evidence for dysfunction which could disrupt the cortical–limbic psychosis pathway in each dementia is described below.

PD dementia

While dopamine depletion in the dorsal striatum due to loss of nigrostriatal neurons results in the characteristic motor symptoms of PD, serotonin dysfunction is thought to be the underlying cause of PD psychosis. Post-mortem analyses indicate Lewy body and Lewy neurite deposition in the raphe nucleus magnus, obscurus, and pallidus, and the gigantocellular nucleus of the medullary lateral reticular formation nuclei (which comprise the serotonergic caudal brainstem complex) precedes deposition in dopaminergic midbrain neurons.Reference Braak, Del Tredici and Rub88 In addition, significant decreases in serotonin, the serotonin transporter (SERT), and the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) are observed in the caudate in post-mortem analyses of PD patients.Reference Kish, Tong and Hornykiewicz72 During the early stages of PD, SERT expression is diminished in the forebrain, but preserved in the caudate, and increased serotonergic uptake is observed in the thalamus and raphe nuclei, indicating a shift toward mesolimbic and mesocortical dysfunction.Reference Albin, Koeppe and Bohnen73, Reference Joutsa, Johansson, Seppanen, Noponen and Kaasinen74 As PD progresses, SERT expression in the caudate decreases and is correlated with disease stage.Reference Kerenyi, Ricaurte and Schretlen75 In PD patients with visual hallucinations, upregulation of 5-HT2A receptors has been observed using positron emission tomography in the inferolateral temporal cortex—in complex visual processing—as well as other portions of the ventral visual pathway, including the bilateral inferior occipital gyrus and right fusiform gyrus.Reference Ballanger, Strafella and van Eimeren76, Reference Huot, Johnston and Darr77 5-HT2A receptor upregulation may represent a compensatory mechanism by which the brain attempts to counteract reductions in serotonin levels.Reference Huot, Johnston and Darr77 In a double-blind, randomized, placebo-controlled phase 3 study of the efficacy of pimavanserin in the treatment of hallucinations and delusions associated with PD psychosis, a post-hoc subgroup analysis revealed patients with cognitive impairment (Mini-Mental State Examination scores 21-24) demonstrate a significantly greater improvement in Scale for the Assessment of Positive Symptoms-PD scores from baseline to week 6 with pimavanserin as compared to placebo-treated patients (P =0.002), suggesting pimavanserin may exhibit a more robust effect in cognitively impaired patients.Reference Cummings, Ballard and Tariot89

Imaging studies demonstrate that PD dementia is associated with more widespread cortical cholinergic depletion than that observed in patients with PD who do not exhibit dementia.Reference Bohnen, Kaufer and Ivanco90, Reference Kuhl, Minoshima and Fessler91 Modulation of cholinergic activity has been reported to decrease hallucinations and delusions in PD dementia patients. Treatment with the acetylcholinesterase inhibitor donepezil significantly reduces hallucinations and paranoid ideation, as well as overall rating scale scores for PD psychosis.Reference Fabbrini, Barbanti and Aurilia92 At present, the underlying mechanism for these effects is unknown. Serotonin has been reported to inhibit acetylcholine release from cortical cholinergic nerve terminalsReference Lanctôt, Herrmann and Mazzotta93 and 5-HT receptor inverse agonism/antagonism may represent another therapeutic strategy to modulate cortical cholinergic activity.

Dementia with Lewy bodies

In DLB, the density of Lewy bodies in limbic areas (highest density in the amygdala) is significantly higher than in neocortical areas.Reference Rezaie, Cairns, Chadwick and Lantos94 As in PD, serotonin dysfunction is thought to be the underlying cause of psychosis in DLB. Cortical 5-HT2 binding differs between patients with DLB with and without hallucinations. Significant deficits in 5-HT2 binding are observed in cortical layers III and V (deep cortical layers that contain pyramidal neurons) in patients who did not experience hallucinations, whereas a 5-HT2 binding deficit was observed only in one upper cortical layer in patients who did experience hallucinations, suggesting 5-HT2 receptor preservation in the temporal cortex may contribute to hallucinations in this population.Reference Cheng, Ferrier and Morris78 This hypothesis is further supported by the observation that serotonergic receptor binding and 5-HIAA levels were significantly decreased in nonhallucinating vs hallucinating patients with DLB in a neurochemical analysis of the temporal cortex. In the same study, downregulation of choline acetyltransferase activity was observed in the temporal and parietal cortices, particularly in those experiencing hallucinations.Reference Perry, Marshall and Kerwin79 Treatment with the cholinesterase inhibitor rivastigmine decreased delusions and hallucinations in patients with DLB compared to placebo.Reference McKeith, Del Ser and Spano95 The mechanism underlying the efficacy of cholinesterase inhibition in the treatment of DLB remains unresolved. However, hallucinations in patients with DLB have been proposed to result from imbalances between the serotonergic and cholinergic inputs to the cortex, suggesting restoration of the balance between the two inputs may account for this effect.Reference Perry, Marshall and Thompson96

AD dementia

Although serotonergic signaling is altered in AD, it may not be the primary dysfunction contributing to psychosis. When compared to healthy controls, significant reductions in 5-HT2 receptor binding and expression have been observed in the frontal, temporal, and cingulate cortices, as well as the amygdala and hippocampus, in patients with AD.97Reference Jansen, Faull, Dragunow and Synek101 In a study of psychopathology in late-onset AD patients, the 5-HT2A receptor polymorphism 102-T/C was significantly associated with visual and auditory hallucinations, and the 5-HT2C receptor polymorphism Cys23Ser was significantly associated with visual hallucinations, implicating neurodegeneration in the biology of the psychotic symptoms of individuals expressing these genetic variations. However, the 5-HT2A 102-T/C polymorphism reduces 5-HT2A receptor expression while the 5-HT2C Cys23Ser polymorphism leads to functional downregulation of the 5-HT2C receptor.102Reference Okada, Northup and Ozaki104

Concomitant serotonergic and cholinergic deficits have been observed in the frontal and temporal cortices in AD patients as compared to healthy controls, and the ratio of serotonin to acetylcholinesterase in the temporal cortex is correlated with psychosis in female patients.Reference Garcia-Alloza, Gil-Bea and Diez-Ariza105 Cholinesterase inhibitors used in the treatment of AD may improve hallucinations and delusions in some patients.Reference Cummings, McRae and Zhang106

There is evidence of GABAergic and glutamatergic dysfunction in AD which might disrupt the cortical–limbic psychosis pathway. Significant reductions in GABA concentrations in numerous cortical areas, including the temporal, frontal (orbitofrontal and premotor cortex), parietal, and occipital cortices, are observed in biopsy and autopsy specimens obtained from AD patients.107-111 Alterations in NMDA-mediated glutamatergic signaling appears to vary throughout the course of AD. Under normal circumstances, glutamate regulates the inhibitory tone of these GABA neurons; amyloid appears to increase the sensitivity of these receptors to glutamate, leading to glutamatergic hyperactivation and GABAergic neuronal degeneration. During the later stages of disease progression, excessive GABAergic neuronal degeneration results in NMDA receptor hypofunction.Reference Olney, Wozniak and Farber80 Evidence of GABAergic or glutamatergic dysfunction has yet to be directly associated with psychosis in AD. Treatment of AD patients with the NMDA receptor antagonist memantine, which was shown to inhibit the excitotoxic effects of NMDA glutamate receptors, has occasionally been reported to worsen or induce new visual hallucinations in AD patients.Reference Monastero, Camarda, Pipia and Camarda81

Frontotemporal dementia

Deficiencies in the serotonergic system have been reported in imaging studies, post-mortem tissue analyses, and cerebrospinal fluid studies of patients with FTD. Decreased 5-HT2A receptor expression has been observed in the orbitofrontal, frontal medial, and cingulate cortices of such patients.Reference Huey, Putnam and Grafman82 Data regarding how GABAergic and glutamatergic signaling are affected in FTD are incomplete; however, loss of glutamatergic pyramidal cells and GABAergic neurons in the upper layers of the frontal and temporal cortices has been reported.Reference Ferrer83 MRI studies have revealed widespread limbic atrophy in FTD,Reference Meyer, Mueller and Stuke112, Reference Boccardi, Sabattoli and Laakso113 suggesting disruption of the cortical–limbic psychosis pathway may occur. However, data regarding dysfunction specific to psychosis in this population are not yet reported, possibly because psychosis is less common in FTD.

Vascular dementia

Serotonergic dysfunction is present in VaD, with increased 5-HT1A and 5-HT2A receptor binding observed in the temporal cortex of post-mortem tissue samples from multi-infarct VaD patients.Reference Elliott, Ballard and Kalaria84 The relationship between this increased receptor binding and psychosis is unknown. Data regarding the roles of GABAergic and glutamatergic signaling in VaD is lacking.

Neurotransmission alterations across dementias

While the strength of the evidence varies across these dementias, all have been associated with alterations in neurotransmission which have the potential to impact the cortical–limbic psychosis pathway. Each of these conditions is highly heterogeneous and the likelihood an individual patient will develop psychosis varies greatly. Further research is required to confirm how etiological factors, such as the anatomical locations of neurodegeneration, compensatory mechanisms, and genetic influences contribute to the development of psychosis.

Proposed Mechanism for DRP

The proposed mechanism for DRP is believed to involve a common cortical–limbic psychosis pathway (Figure 1). Cortical GABAergic interneuron or NMDA receptor dysfunction is hypothesized to result in loss of inhibitory tone, leading to hyperactivity of glutamatergic neurons that signal to the VTA. Alternatively, excessive signaling via 5-HT2A receptors on pyramidal glutamate neurons can lead to hyperactive glutamatergic neurons that signal to the VTA. Sustained hyperactive glutamatergic signaling then leads to mesolimbic dopamine pathway hyperactivation, resulting in hallucinations and delusions. Excess signaling via 5-HT2A receptors in the visual cortex may be responsible specifically for visual hallucinations.64Reference Hacksell, Burstein, McFarland, Mills and Williams66, Reference Kometer, Schmidt, Jancke and Vollenweider70, 72Reference Braak and Braak87

The cortical–limbic psychosis pathway can be triggered in a variety of different ways across dementias based on which neurons are lost or damaged and which signaling pathways become disordered but appears to be responsive to serotonin modulation. Cortical GABA interneuron dysfunction, excess serotonin, cortical 5-HT2A receptor upregulation, excess striatal dopamine, striatal D2 receptor upregulation, and excess glutamate signaling all have the potential to contribute to pathway dysfunction.

Proposed Mechanism of Action of Pimavanserin

Across the underlying causes of cortical–limbic psychosis pathway hyperactivation, 5-HT2A receptor antagonism represents a common point of regulation and for treatment intervention with antipsychotics.Reference Hacksell, Burstein, McFarland, Mills and Williams66 Pimavanserin is a selective serotonin inverse agonist/antagonist at 5-HT2A receptors, with 40-fold less activity at 5-HT2C receptors and no affinity for dopaminergic, histaminergic, muscarinic, or adrenergic receptors, and is proposed to act as a targeted serotonergic modulator of circuits (Figure 2).Reference Hacksell, Burstein, McFarland, Mills and Williams66 Pimavanserin is thought to reduce the activity of these receptors to below basal levels and regulate the effects of both cortical GABAergic deficits and excess cortical serotonergic signaling. This is posited to decrease visual hallucinations and attenuate glutamate signaling to the VTA and mesolimbic pathway, further decreasing delusions and hallucinations.

Figure 2. Pimavanserin-mediated 5-HT2A receptor inhibition: hypothesized modulation of signaling through a variety of neurotransmitters. Through 5-HT2A antagonism/reverse agonism, pimavanserin is proposed to act as a targeted serotonergic modulator of circuits, mitigating the effects of GABAergic deficits and excess serotonergic signaling, while also reducing hyperactive glutamatergic signaling and mesolimbic pathway activation.

Summary

DRP is a commonly occurring phenomenon across dementias, yet no pharmacological agents are currently approved by the FDA for DRP treatment. Commonly used typical and atypical antipsychotics have an increased risk of death and other treatment-limiting side effects.Reference Schneider, Dagerman and Insel114, 115 While the etiology of DRP is unknown, neurobiological and pharmacological evidence supports a common, interconnected cortical–limbic psychosis pathway mediating DRP, which may be modified via 5-HT2A receptor inverse agonism/antagonism. The only FDA-approved agent to treat hallucinations and delusions associated with PD psychosis, pimavanserin (proposed to act as a targeted serotonergic modulator of circuits), is being investigated to treat DRP in a phase 3 trial completed in 2019.5 The selectivity of pimavanserin presents a potential novel mechanism for the management of hallucinations and delusions associated with DRP.Reference Hacksell, Burstein, McFarland, Mills and Williams66, Reference Cummings, Ballard and Tariot89

Acknowledgments

The authors received editorial assistance in the preparation of this manuscript from Arbor Scientia, Inc., which was supported by Acadia Pharmaceuticals Inc., San Diego, CA. Dr. Cummings is supported by Keep Memory Alive (KMA), National Institute of General Medical Sciences (NIGMS) grant P20GM109025, National Institute of Neurological Disorders and Stroke (NINDS) grant U01NS093334, and National Institute of Aging (NIA) grant R01AG053798. Dr. Devanand is supported by the NIA.

Disclosures

Dr. Cummings has provided consultation to Acadia, Actinogen, Alkahest, Allergan, Alzheon, Avanir, Axsome, BiOasis, Biogen, Cassava, Cerecin, Cortexyme, Diadem, EIP Pharma, Eisai, Foresight, Genentech, Green Valley, Grifols, Otsuka, Resverlogix, Roche, Samumed, Samus, Signant, Third Rock, Toyama, and United Neuroscience pharmaceutical and assessment companies. Dr. Cummings has stock options in Prana, Neurokos, Adamas, MedAvante-ProPhase, Anovis, and BiOasis.

Dr. Devanand has provided consultation to Acadia, BXcel, Corium, Eisai, Genentech, and Grifols.

Dr. Stahl has served as a consultant to Acadia, Alkermes, Allergan, Arbor Pharmaceuticals, Axovant, Axsome, Celgene, Concert, Clearview, EMD Serono, Eisai Pharmaceuticals, Ferring, Impel NeuroPharma, Intra-Cellular Therapies, Ironshore Pharmaceuticals, Janssen, Lilly, Lundbeck, Merck, Otsuka, Pfizer, Sage Therapeutics, Servier, Shire, Sunovion, Takeda, Taliaz, Teva, Tonix, Tris Pharma, and Vifor Pharma; he is a board member of Genomind; he has served on speakers bureaus for Acadia, Lundbeck, Otsuka, Perrigo, Servier, Sunovion, Takeda and Vertex; and he has received research and/or grant support from Acadia, Avanir, Braeburn Pharmaceuticals, Eli Lilly, Intra-Cellular Therapies, Ironshore Pharmaceuticals, ISSWSH, Neurocrine, Otsuka, Shire, Sunovion, and TMS NeuroHealth Centers.

References

NUPLAZID® [package insert]. San Diego, CA: Acadia Pharmaceuticals Inc.; 2019.Google Scholar
Cummings, J, Isaacson, S, Mills, R, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet. 2014;383(9916):533540.CrossRefGoogle ScholarPubMed
Ballard, C, Banister, C, Khan, Z, et al. Evaluation of the safety, tolerability, and efficacy of pimavanserin versus placebo in patients with Alzheimer’s disease psychosis: a phase 2, randomised, placebo-controlled, double-blind study. Lancet Neurol. 2018;17(3):213222.10.1016/S1474-4422(18)30039-5CrossRefGoogle ScholarPubMed
Acadia Pharmaceuticals Inc. Analyses of pimavanserin studies evaluating treatment in Alzheimer’s disease psychosis and parkinson’s disease psychosis published in the journal of prevention of Alzheimer’s disease suggest potential for treating dementia-related psychosis. Acadia Pharmaceuticals website. http://ir.acadia-pharm.com/phoenix.zhtml?c=125180&p=irol-newsArticle&ID=2366704. Updated September 2018. Accessed June 2019.Google Scholar
Relapse prevention study of pimavanserin in dementia-related psychosis. ClinicalTrials.gov identifier: NCT03325556. https://clinicaltrials.gov/ct2/show/NCT03325556. Updated April 21, 2020. Accessed July 2020.Google Scholar
EU Clinical Trials Register. Clinical trials for 2017-002227-13. https://www.clinicaltrialsregister.eu/ctr-search/search?query=2017-002227-13. Accessed July 2020.Google Scholar
Goodman, RA, Lochner, KA, Thambisetty, M, et al. Prevalence of dementia subtypes in United States Medicare fee-for-service beneficiaries, 2011-2013. Alzheimers Dement. 2017;13(1):2837.10.1016/j.jalz.2016.04.002CrossRefGoogle ScholarPubMed
Hebert, LE, Weuve, J, Scherr, PA, Evans, DA. Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology. 2013;80(19):17781783.10.1212/WNL.0b013e31828726f5CrossRefGoogle ScholarPubMed
Plassman, BL, Langa, KM, Fisher, GG, et al. Prevalence of dementia in the United States: the aging, demographics, and memory study. Neuroepidemiology. 2007;29(1–2):125132.CrossRefGoogle ScholarPubMed
Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement. 2017, 2017;13(4):325373.Google Scholar
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (DSM-5). Arlington, VA: American Psychiatric Association; 2013.Google Scholar
Cerejeira, J, Lagarto, L, Mukaetova-Ladinska, EB. Behavioral and psychological symptoms of dementia. Front Neurol. 2012;3:73CrossRefGoogle ScholarPubMed
Cummings, J, Mintzer, J, Brodaty, H, et al. Agitation in cognitive disorders: International Psychogeriatric Association provisional consensus clinical and research definition. Int Psychogeriatr. 2015;27(1):717.CrossRefGoogle ScholarPubMed
Gilley, DW, Wilson, RS, Beckett, LA, Evans, DA. Psychotic symptoms and physically aggressive behavior in Alzheimer’s disease. J Am Geriatr Soc. 1997;45(9):10741079.10.1111/j.1532-5415.1997.tb05969.xCrossRefGoogle ScholarPubMed
Leonard, R, Tinetti, ME, Allore, HG, Drickamer, MA. Potentially modifiable resident characteristics that are associated with physical or verbal aggression among nursing home residents with dementia. Arch Intern Med. 2006;166(12):12951300.CrossRefGoogle ScholarPubMed
Lopez, OL, Becker, JT, Sweet, RA, et al. Psychiatric symptoms vary with the severity of dementia in probable Alzheimer’s disease. J Neuropsychiatry Clin Neurosci. 2003;15(3):346353.10.1176/jnp.15.3.346CrossRefGoogle ScholarPubMed
Ballard, C, Neill, D, O’Brien, J, et al. Anxiety, depression and psychosis in vascular dementia: prevalence and associations. J Affect Disord. 2000;59(2):97106.CrossRefGoogle ScholarPubMed
Burns, A, Jacoby, R, Levy, R. Psychiatric phenomena in Alzheimer’s disease. I: disorders of thought content. Br J Psychiatry. 1990;157:72, 92–76, 74.Google ScholarPubMed
Burns, A, Jacoby, R, Levy, R. Psychiatric phenomena in Alzheimer’s disease. II: disorders of perception. Br J Psychiatry. 1990;157:76, 92–81, 74.10.1192/bjp.157.1.76CrossRefGoogle Scholar
Johnson, DK, Watts, AS, Chapin, BA, Anderson, R, Burns, JM. Neuropsychiatric profiles in dementia. Alzheimer Dis Assoc Disord. 2011;25(4):326–332.10.1097/WAD.0b013e31820d89b6CrossRefGoogle ScholarPubMed
Lyketsos, CG, Lopez, O, Jones, B, et al. Prevalence of neuropsychiatric symptoms in dementia and mild cognitive impairment: results from the cardiovascular health study. JAMA. 2002;288(12):14751483.10.1001/jama.288.12.1475CrossRefGoogle ScholarPubMed
Lyketsos, CG, Steinberg, M, Tschanz, JT, et al. Mental and behavioral disturbances in dementia: findings from the Cache County Study on Memory in Aging. Am J Psychiatry. 2000;157(5):708714.CrossRefGoogle Scholar
Leroi, I, Voulgari, A, Breitner, JC, Lyketsos, CG. The epidemiology of psychosis in dementia. Am J Geriatr Psychiatry. 2003;11(1):8391.CrossRefGoogle ScholarPubMed
Ballard, C, Saad, K, Patel, A, et al. The prevalence and phenomenology of psychotic symptoms in dementia sufferers. Int J Geriatr Psychiatry. 1995;10(6):477485.CrossRefGoogle Scholar
Nagahama, Y, Okina, T, Suzuki, N, et al. Classification of psychotic symptoms in dementia with Lewy bodies. Am J Geriatr Psychiatry. 2007;15(11):961967.CrossRefGoogle ScholarPubMed
Aarsland, D, Ballard, C, Larsen, JP, McKeith, I. A comparative study of psychiatric symptoms in dementia with Lewy bodies and Parkinson’s disease with and without dementia. Int J Geriatr Psychiatry. 2001;16(5):528536.10.1002/gps.389CrossRefGoogle ScholarPubMed
Ballard, C, Holmes, C, McKeith, I, et al. Psychiatric morbidity in dementia with Lewy bodies: a prospective clinical and neuropathological comparative study with Alzheimer’s disease. Am J Psychiatry. 1999;156(7):10391045.Google ScholarPubMed
Lee, W, Tsai, C, Gauthier, S, Wang, S, Fuh, J. The association between cognitive impairment and neuropsychiatric symptoms in patients with Parkinson’s disease dementia. Int Psychogeriatr. 2012;24(12):19801987.CrossRefGoogle ScholarPubMed
Mendez, MF, Shapira, JS, Woods, RJ, Licht, EA, Saul, RE. Psychotic symptoms in frontotemporal dementia: prevalence and review. Dement Geriatr Cogn Disord. 2008;25(3):206211.CrossRefGoogle ScholarPubMed
Mourik, JC, Rosso, SM, Niermeijer, MF, et al. Frontotemporal dementia: behavioral symptoms and caregiver distress. Dement Geriatr Cogn Disord. 2004;18(3–4):299306.CrossRefGoogle ScholarPubMed
Fenelon, G, Soulas, T, Zenasni, F, Cleret de Langavant, L. The changing face of Parkinson’s disease-associated psychosis: a cross-sectional study based on the new NINDS-NIMH criteria. Mov Disord. 2010;25(6):763766.10.1002/mds.22839CrossRefGoogle ScholarPubMed
McKeith, IG, Boeve, BF, Dickson, DW, et al. Diagnosis and management of dementia with Lewy bodies: fourth consensus report of the DLB Consortium. Neurology. 2017;89(1):88100.CrossRefGoogle ScholarPubMed
Devanand, DP, Jacobs, DM, Tang, MX, et al. The course of psychopathologic features in mild to moderate Alzheimer disease. Arch Gen Psychiatry. 1997;54(3):257263.10.1001/archpsyc.1997.01830150083012CrossRefGoogle ScholarPubMed
Haupt, M. Psychotherapeutic intervention in dementia. Dementia. 1996;7(4):207209.Google ScholarPubMed
Lim, L, Zhang, A, Lim, L, et al. High caregiver burden in young onset dementia: what factors need attention? J Alzheimers Dis. 2018;61(2):537543.CrossRefGoogle ScholarPubMed
Naimark, D, Jackson, E, Rockwell, E, Jeste, DV. Psychotic symptoms in Parkinson’s disease patients with dementia. J Am Geriatr Soc. 1996;44(3):296299.10.1111/j.1532-5415.1996.tb00918.xCrossRefGoogle ScholarPubMed
Peters, ME, Schwartz, S, Han, D, et al. Neuropsychiatric symptoms as predictors of progression to severe Alzheimer’s dementia and death: the Cache County Dementia Progression Study. Am J Psychiatry. 2015;172(5):460465.CrossRefGoogle ScholarPubMed
Scarmeas, N, Brandt, J, Albert, M, et al. Delusions and hallucinations are associated with worse outcome in Alzheimer disease. Arch Neurol. 2005;62(10):16011608.10.1001/archneur.62.10.1601CrossRefGoogle ScholarPubMed
Rovelet-Lecrux, A, Hannequin, D, Raux, G, et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet. 2006;38(1):2426.CrossRefGoogle ScholarPubMed
Hyman, BT, Phelps, CH, Beach, TG, et al. National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement. 2012;8(1):113.10.1016/j.jalz.2011.10.007CrossRefGoogle ScholarPubMed
Goldman, JE, Yen, SH, Chiu, FC, Peress, NS. Lewy bodies of Parkinson’s disease contain neurofilament antigens. Science. 1983;221(4615):10821084.CrossRefGoogle ScholarPubMed
Colom-Cadena, M, Pegueroles, J, Herrmann, AG, et al. Synaptic phosphorylated alpha-synuclein in dementia with Lewy bodies. Brain. 2017;140(12):32043214.CrossRefGoogle ScholarPubMed
Hamilton, RL. Lewy bodies in Alzheimer’s disease: a neuropathological review of 145 cases using alpha-synuclein immunohistochemistry. Brain Pathol. 2000;10(3):378384.CrossRefGoogle ScholarPubMed
Nelson, PT, Dickson, DW, Trojanowski, JQ, et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report. Brain. 2019;142(6):15031527.CrossRefGoogle ScholarPubMed
Brenowitz, WD, Keene, CD, Hawes, SE, et al. Alzheimer’s disease neuropathologic change, Lewy body disease, and vascular brain injury in clinic- and community-based samples. Neurobiol Aging. 2017;53:8392.CrossRefGoogle ScholarPubMed
Schneider, JA, Arvanitakis, Z, Bang, W, Bennett, DA. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology. 2007;69(24):21972204.CrossRefGoogle ScholarPubMed
Gründer, G, Cumming, P. Chapter 7—the dopamine hypothesis of schizophrenia: current status. In: Abel T, Nickl-Jockschat T, eds. The Neurobiology of Schizophrenia. San Diego, CA: Academic Press; 2016:109124.10.1016/B978-0-12-801829-3.00015-XCrossRefGoogle Scholar
Hirvonen, J, Hietala, J. Dopamine receptor imaging in schizophrenia: focus on genetic vulnerability. In: Seeman P, Madras B, eds. Imaging of the Human Brain in Health and Disease. San Diego, CA: Elsevier Inc.; 2014:341360.CrossRefGoogle Scholar
Watanabe, T, Morimoto, K, Nakamura, M, Suwaki, H. Modification of behavioral responses induced by electrical stimulation of the ventral tegmental area in rats. Behav Brain Res. 1998;93(1–2):119129.CrossRefGoogle ScholarPubMed
Zhou, Z, Zhang, G, Li, X, et al. Loss of phenotype of parvalbumin interneurons in rat prefrontal cortex is involved in antidepressant- and propsychotic-like behaviors following acute and repeated ketamine administration. Mol Neurobiol. 2015;51(2):808819.CrossRefGoogle ScholarPubMed
Nakazawa, K, Zsiros, V, Jiang, Z, et al. GABAergic interneuron origin of schizophrenia pathophysiology. Neuropharmacology. 2012;62(3):15741583.CrossRefGoogle ScholarPubMed
Lahti, AC, Koffel, B, LaPorte, D, Tamminga, CA. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology. 1995;13(1):919.CrossRefGoogle ScholarPubMed
Almodovar-Fabregas, LJ, Segarra, O, Colon, N, et al. Effects of cocaine administration on VTA cell activity in response to prefrontal cortex stimulation. Ann N Y Acad Sci. 2002;965:157171.CrossRefGoogle ScholarPubMed
Karreman, M, Moghaddam, B. The prefrontal cortex regulates the basal release of dopamine in the limbic striatum: an effect mediated by ventral tegmental area. J Neurochem. 1996;66(2):589598.10.1046/j.1471-4159.1996.66020589.xCrossRefGoogle ScholarPubMed
Rolland, B, Jardri, R, Amad, A, et al. Pharmacology of hallucinations: several mechanisms for one single symptom? Biomed Res Int. 2014;2014:30710610.1155/2014/307106CrossRefGoogle ScholarPubMed
Ingram, SL, Prasad, BM, Amara, SG. Dopamine transporter-mediated conductances increase excitability of midbrain dopamine neurons. Nat Neurosci. 2002;5(10):971978.CrossRefGoogle ScholarPubMed
Mahoney, JJ, 3rd., Kalechstein, AD, De La Garza, R, 2nd., Newton, TF. Presence and persistence of psychotic symptoms in cocaine- versus methamphetamine-dependent participants. Am J Addict. 2008;17(2):8398.CrossRefGoogle ScholarPubMed
McKetin, R, Baker, AL, Dawe, S, Voce, A, Lubman, DI. Differences in the symptom profile of methamphetamine-related psychosis and primary psychotic disorders. Psychiatry Res. 2017;251:349354.CrossRefGoogle ScholarPubMed
Lahti, AC, Holcomb, HH, Medoff, DR, Tamminga, CA. Ketamine activates psychosis and alters limbic blood flow in schizophrenia. Neuroreport. 1995;6(6):869872.CrossRefGoogle Scholar
Krystal, JH, Karper, LP, Seibyl, JP, et al. Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans. Psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Arch Gen Psychiatry. 1994;51(3):199214.CrossRefGoogle ScholarPubMed
Powers, AR, 3rd, Gancsos, MG, Finn, ES, Morgan, PT, Corlett, PR. Ketamine-induced hallucinations. Psychopathology. 2015;48(6):376385.CrossRefGoogle ScholarPubMed
Carhart-Harris, RL, Muthukumaraswamy, S, Roseman, L, et al. Neural correlates of the LSD experience revealed by multimodal neuroimaging. Proc Natl Acad Sci U S A. 2016;113(17):48534858.CrossRefGoogle ScholarPubMed
Griffiths, R, Richards, W, Johnson, M, McCann, U, Jesse, R. Mystical-type experiences occasioned by psilocybin mediate the attribution of personal meaning and spiritual significance 14 months later. J Psychopharmacol. 2008;22(6):621632.CrossRefGoogle ScholarPubMed
Vollenweider, FX, Vollenweider-Scherpenhuyzen, MF, Babler, A, Vogel, H, Hell, D. Psilocybin induces schizophrenia-like psychosis in humans via a serotonin-2 agonist action. Neuroreport. 1998;9(17):38973902.10.1097/00001756-199812010-00024CrossRefGoogle Scholar
Stahl, SM. Stahl’s Essential Psychopharmacology: Neuroscientific Basis and Practical Applications. 4th ed. New York, NY: Cambridge University Press; 2013.Google Scholar
Hacksell, U, Burstein, ES, McFarland, K, Mills, RG, Williams, H. On the discovery and development of pimavanserin: a novel drug candidate for Parkinson’s psychosis. Neurochem Res. 2014;39(10):20082017.CrossRefGoogle ScholarPubMed
Gardell, LR, Vanover, KE, Pounds, L, et al. ACP-103, a 5-hydroxytryptamine 2A receptor inverse agonist, improves the antipsychotic efficacy and side-effect profile of haloperidol and risperidone in experimental models. J Pharmacol Exp Ther. 2007;322(2):862870.10.1124/jpet.107.121715CrossRefGoogle ScholarPubMed
McFarland, K, Price, DL, Bonhaus, DW. Pimavanserin, a 5-HT2A inverse agonist, reverses psychosis-like behaviors in a rodent model of Parkinson’s disease. Behav Pharmacol. 2011;22(7):681692.CrossRefGoogle Scholar
Li, Z, Ichikawa, J, Huang, M, et al. ACP-103, a 5-HT2A/2C inverse agonist, potentiates haloperidol-induced dopamine release in rat medial prefrontal cortex and nucleus accumbens. Psychopharmacology (Berl). 2005;183(2):144153.10.1007/s00213-005-0170-9CrossRefGoogle ScholarPubMed
Kometer, M, Schmidt, A, Jancke, L, Vollenweider, FX. Activation of serotonin 2A receptors underlies the psilocybin-induced effects on alpha oscillations, N170 visual-evoked potentials, and visual hallucinations. J Neurosci. 2013;33(25):1054410551.CrossRefGoogle ScholarPubMed
Stahl, SM. Beyond the dopamine hypothesis of schizophrenia to three neural networks of psychosis: dopamine, serotonin, and glutamate. CNS Spectrums. 2018;23(3):187191.CrossRefGoogle ScholarPubMed
Kish, SJ, Tong, J, Hornykiewicz, O, et al. Preferential loss of serotonin markers in caudate versus putamen in Parkinson’s disease. Brain. 2008;131(Pt 1):120131.Google ScholarPubMed
Albin, RL, Koeppe, RA, Bohnen, NI, et al. Spared caudal brainstem SERT binding in early Parkinson’s disease. J Cereb Blood Flow Metab. 2008;28(3):441444.CrossRefGoogle ScholarPubMed
Joutsa, J, Johansson, J, Seppanen, M, Noponen, T, Kaasinen, V. Dorsal-to-ventral shift in midbrain dopaminergic projections and increased thalamic/raphe serotonergic function in early Parkinson disease. J Nucl Med. 2015;56(7):10361041.CrossRefGoogle ScholarPubMed
Kerenyi, L, Ricaurte, GA, Schretlen, DJ, et al. Positron emission tomography of striatal serotonin transporters in Parkinson disease. Arch Neurol. 2003;60(9):12231229.CrossRefGoogle ScholarPubMed
Ballanger, B, Strafella, AP, van Eimeren, T, et al. Serotonin 2A receptors and visual hallucinations in Parkinson disease. Arch Neurol. 2010;67(4):416421.CrossRefGoogle ScholarPubMed
Huot, P, Johnston, TH, Darr, T, et al. Increased 5-HT2A receptors in the temporal cortex of parkinsonian patients with visual hallucinations. Mov Disord. 2010;25(10):13991408.CrossRefGoogle ScholarPubMed
Cheng, AV, Ferrier, IN, Morris, CM, et al. Cortical serotonin-S2 receptor binding in Lewy body dementia, Alzheimer’s and Parkinson’s diseases. J Neurol Sci. 1991;106(1):5055.CrossRefGoogle ScholarPubMed
Perry, EK, Marshall, E, Kerwin, J, et al. Evidence of a monoaminergic-cholinergic imbalance related to visual hallucinations in Lewy body dementia. J Neurochem. 1990;55(4):14541456.10.1111/j.1471-4159.1990.tb03162.xCrossRefGoogle ScholarPubMed
Olney, JW, Wozniak, DF, Farber, NB. Excitotoxic neurodegeneration in Alzheimer disease. New hypothesis and new therapeutic strategies. Arch Neurol. 1997;54(10):12341240.CrossRefGoogle ScholarPubMed
Monastero, R, Camarda, C, Pipia, C, Camarda, R. Visual hallucinations and agitation in Alzheimer’s disease due to memantine: report of three cases. J Neurol Neurosurg Psychiatry. 2007;78(5):54610.1136/jnnp.2006.096420CrossRefGoogle ScholarPubMed
Huey, ED, Putnam, KT, Grafman, J. A systematic review of neurotransmitter deficits and treatments in frontotemporal dementia. Neurology. 2006;66(1):1722.CrossRefGoogle ScholarPubMed
Ferrer, I. Neurons and their dendrites in frontotemporal dementia. Dement Geriatr Cogn Disord. 1999;10(Suppl 1):5560.CrossRefGoogle ScholarPubMed
Elliott, MS, Ballard, CG, Kalaria, RN, et al. Increased binding to 5-HT1A and 5-HT2A receptors is associated with large vessel infarction and relative preservation of cognition. Brain. 2009;132(Pt 7):18581865.CrossRefGoogle ScholarPubMed
Govindpani, K, Calvo-Flores Guzman, B, Vinnakota, C, et al. Towards a better understanding of GABAergic remodeling in Alzheimer’s disease. Int J Mol Sci. 2017;18(8):1CrossRefGoogle ScholarPubMed
Lakhan, SE, Caro, M, Hadzimichalis, N. NMDA receptor activity in neuropsychiatric disorders. Front Psychiatry. 2013;4:52CrossRefGoogle ScholarPubMed
Braak, H, Braak, E. Frequency of stages of Alzheimer-related lesions in different age categories. Neurobiol Aging. 1997;18(4):351357.CrossRefGoogle ScholarPubMed
Braak, H, Del Tredici, K, Rub, U, et al. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197211.CrossRefGoogle ScholarPubMed
Cummings, J, Ballard, C, Tariot, P, et al. Pimavanserin: potential treatment for dementia-related psychosis. J Prev Alzheimers Dis. 2018;5(4):253258.Google ScholarPubMed
Bohnen, NI, Kaufer, DI, Ivanco, LS, et al. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: an in vivo positron emission tomographic study. Arch Neurol. 2003;60(12):17451748.CrossRefGoogle Scholar
Kuhl, DE, Minoshima, S, Fessler, JA, et al. In vivo mapping of cholinergic terminals in normal aging, Alzheimer’s disease, and Parkinson’s disease. Ann Neurol. 1996;40(3):399410.CrossRefGoogle ScholarPubMed
Fabbrini, G, Barbanti, P, Aurilia, C, et al. Donepezil in the treatment of hallucinations and delusions in Parkinson’s disease. Neurol Sci. 2002;23(1):4143.CrossRefGoogle ScholarPubMed
Lanctôt, KL, Herrmann, N, Mazzotta, P. Role of serotonin in the behavioral and psychological symptoms of dementia. J Neuropsychiatry Clin Neurosci. 2001;13(1):521.CrossRefGoogle ScholarPubMed
Rezaie, P, Cairns, NJ, Chadwick, A, Lantos, PL. Lewy bodies are located preferentially in limbic areas in diffuse Lewy body disease. Neurosci Lett. 1996;212(2):111114.CrossRefGoogle ScholarPubMed
McKeith, I, Del Ser, T, Spano, P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):20312036.CrossRefGoogle ScholarPubMed
Perry, EK, Marshall, E, Thompson, P, et al. Monoaminergic activities in Lewy body dementia: relation to hallucinosis and extrapyramidal features. J Neural Transm Park Dis Dement Sect. 1993;6(3):167177.CrossRefGoogle ScholarPubMed
Blin, J, Baron, JC, Dubois, B, et al. Loss of brain 5-HT2 receptors in Alzheimer’s disease. In vivo assessment with positron emission tomography and [18F]setoperone. Brain. 1993;116(Pt 3):497510.CrossRefGoogle Scholar
Cross, AJ, Crow, TJ, Ferrier, IN, et al. Serotonin receptor changes in dementia of the Alzheimer type. J Neurochem. 1984;43(6):15741581.CrossRefGoogle ScholarPubMed
Crow, TJ, Cross, AJ, Cooper, SJ, et al. Neurotransmitter receptors and monoamine metabolites in the brains of patients with Alzheimer-type dementia and depression, and suicides. Neuropharmacology. 1984;23(12B):15611569.CrossRefGoogle ScholarPubMed
Perry, EK, Perry, RH, Candy, JM, et al. Cortical serotonin-S2 receptor binding abnormalities in patients with Alzheimer’s disease: comparisons with Parkinson’s disease. Neurosci Lett. 1984;51(3):353357.CrossRefGoogle ScholarPubMed
Jansen, KL, Faull, RL, Dragunow, M, Synek, BL. Alzheimer’s disease: changes in hippocampal N-methyl-d-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors—an autoradiographic study. Neuroscience. 1990;39(3):613627.CrossRefGoogle ScholarPubMed
Holmes, C, Arranz, MJ, Powell, JF, Collier, DA, Lovestone, S. 5-HT2A and 5-HT2C receptor polymorphisms and psychopathology in late onset Alzheimer’s disease. Hum Mol Genet. 1998;7(9):15071509.CrossRefGoogle ScholarPubMed
Polesskaya, OO, Sokolov, BP. Differential expression of the "C" and "T" alleles of the 5-HT2A receptor gene in the temporal cortex of normal individuals and schizophrenics. J Neurosci Res. 2002;67(6):812822.CrossRefGoogle ScholarPubMed
Okada, M, Northup, JK, Ozaki, N, et al. Modification of human 5-HT(2C) receptor function by Cys23Ser, an abundant, naturally occurring amino-acid substitution. Mol Psychiatry. 2004;9(1):5564.CrossRefGoogle ScholarPubMed
Garcia-Alloza, M, Gil-Bea, FJ, Diez-Ariza, M, et al. Cholinergic-serotonergic imbalance contributes to cognitive and behavioral symptoms in Alzheimer’s disease. Neuropsychologia. 2005;43(3):442449.CrossRefGoogle ScholarPubMed
Cummings, JL, McRae, T, Zhang, R, Donepezil-Sertraline Study G. Effects of donepezil on neuropsychiatric symptoms in patients with dementia and severe behavioral disorders. Am J Geriatr Psychiatry. 2006;14(7):605612.CrossRefGoogle ScholarPubMed
Ellison, DW, Beal, MF, Mazurek, MF, Bird, ED, Martin, JB. A postmortem study of amino acid neurotransmitters in Alzheimer’s disease. Ann Neurol. 1986;20(5):616621.CrossRefGoogle ScholarPubMed
Gueli, MC, Taibi, G. Alzheimer’s disease: amino acid levels and brain metabolic status. Neurol Sci. 2013;34(9):15751579.CrossRefGoogle ScholarPubMed
Lowe, SL, Francis, PT, Procter, AW, et al. Gamma-aminobutyric acid concentration in brain tissue at two stages of Alzheimer’s disease. Brain. 1988;111(Pt 4):785799.CrossRefGoogle ScholarPubMed
Perry, TL, Yong, VW, Bergeron, C, Hansen, S, Jones, K. Amino acids, glutathione, and glutathione transferase activity in the brains of patients with Alzheimer’s disease. Ann Neurol. 1987;21(4):331336.10.1002/ana.410210403CrossRefGoogle ScholarPubMed
Sasaki, H, Muramoto, O, Kanazawa, I, et al. Regional distribution of amino acid transmitters in postmortem brains of presenile and senile dementia of Alzheimer type. Ann Neurol. 1986;19(3):263269.CrossRefGoogle ScholarPubMed
Meyer, S, Mueller, K, Stuke, K, et al. Predicting behavioral variant frontotemporal dementia with pattern classification in multi-center structural MRI data. Neuroimage Clin. 2017;14:656662.CrossRefGoogle ScholarPubMed
Boccardi, M, Sabattoli, F, Laakso, MP, et al. Frontotemporal dementia as a neural system disease. Neurobiol Aging. 2005;26(1):3744.CrossRefGoogle ScholarPubMed
Schneider, LS, Dagerman, K, Insel, PS. Efficacy and adverse effects of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatry. 2006;14(3):191210.CrossRefGoogle ScholarPubMed
US Food and Drug Administration. FDA Public Health Advisory: Deaths with Antipsychotics in Elderly Patients with Behavioral Disturbances. Silver Spring, MD: US Food and Drug Administration; 2005.Google Scholar
Figure 0

Table 1. Prevalence ranges for psychosis, delusions, and hallucinations in AD dementia, VaD, DLB, PD Dementia, and FTD

Figure 1

Figure 1. Hypothesized cortical–limbic psychosis pathway and proposed mechanism of disease for DRP. Neurobiological and pharmacological evidence suggests that hallucinations and delusions are precipitated by overactivation of the mesolimbic pathway, while visual hallucinations are mediated via overactivation of the visual cortex. Dissociative anesthetic-induced (ie, PCP, ketamine) glutamate NMDA receptor antagonism, psychedelic-induced (ie, LSD, psilocybin) serotonin 5-HT2A receptor agonism, and psychostimulant-induced (ie, amphetamine, cocaine) dopamine D2 receptor agonism/DAT antagonism have all been reported to precipitate hallucinations and delusions. In contrast, antipsychotic-mediated D2 and 5-HT2A antagonism treat both hallucinations and delusions. GABAergic interneuron or NMDA receptor dysfunction, and excess serotonin or 5-HT2A receptor upregulation in the cerebral cortex can result in sustained activation of pyramidal neurons and may lead to hyperactive glutamatergic signaling to the VTA, resulting in excess dopamine or D2 receptor upregulation in the ventral striatum, triggering hallucinations and delusions in DRP.5556,7087

Figure 2

Figure 2. Pimavanserin-mediated 5-HT2A receptor inhibition: hypothesized modulation of signaling through a variety of neurotransmitters. Through 5-HT2A antagonism/reverse agonism, pimavanserin is proposed to act as a targeted serotonergic modulator of circuits, mitigating the effects of GABAergic deficits and excess serotonergic signaling, while also reducing hyperactive glutamatergic signaling and mesolimbic pathway activation.