Abstract
Background: Approximately 15% of older adults may experience psychotic phenomena. Primary psychiatric disorders that manifest with psychosis (delusions, hallucinations, and disorganized thought or behavior) account for less than half. Up to 60% of late-life psychotic symptoms are due to systemic medical or neurological conditions, particularly neurodegenerative diseases. A thorough medical workup including laboratory tests, additional procedures if indicated, and neuroimaging studies is recommended. This narrative review summarizes current evidence regarding the epidemiology and phenomenology of psychotic symptoms encountered as part of the neurodegenerative disease continuum (including prodromal and manifest stages). Results: Prodromes are constellations of symptoms that precede the onset of overt neurodegenerative syndromes. Prodromal psychotic features, particularly delusions, have been associated with an increased likelihood of receiving a neurodegenerative disease diagnosis within several years. Prompt prodrome recognition is crucial for early intervention. The management of psychosis associated with neurodegenerative diseases includes behavioral and somatic strategies, although evidence is scarce and mostly limited to case reports, case series, or expert consensus guidelines, with few randomized controlled trials. Conclusion: The complexity of psychotic manifestations warrants management by interprofessional teams that provide coordinated, integrated care.
Keywords: neurodegeneration, movement disorder, dementia, prodrome, review, management
Introduction
Approximately 15% of older adults may experience psychotic phenomena. 1 The combined prevalence of the two most common primary psychotic disorders (schizophrenia and delusional disorder) and mood disorders with psychotic features in adults older than 65 is 5-6%, with secondary psychosis accounting for the remainder.2,3 It is estimated that about 60% of new-onset psychotic symptoms in late life reflect underlying systemic medical or neurological conditions, most often neurodegenerative diseases. 4 In contrast to primary psychotic syndromes, visual hallucinations (VH) are more common than auditory hallucinations (AH) in neurodegenerative diseases, with a variable prevalence of delusions. When patients over 40 years old present with psychosis for the first time in their life, a thorough workup is recommended. Additionally, atypical onset ages (higher than the average onset age in the general population), an insidiously progressive course, unusual clinical presentations, and treatment resistance should prompt an assessment for neurodegenerative diseases.5,6
Over the past decade, a body of work has accumulated on the previously underrecognized neurodegenerative disease prodromes. Prodromes are constellations of symptoms that occur prior to the onset of overt neurological (e.g., cognitive or motor) features and often include psychiatric manifestations. This review will cover neurodegenerative disease prodromes (also referred to as preclinical, premanifest, or premotor), as well as overt (also termed manifest or clinical) stages. Of note, in the Alzheimer’s disease (AD) literature, mild cognitive impairment (MCI) and a newer construct, mild behavioral impairment (MBI) that describes patients with behavioral symptoms who may or may not have cognitive deficits, are classified as prodromal. 7 In contrast, the earlier stage (corresponding to normal cognition, subjective cognitive decline, and MBI-preclinical) is considered preclinical. 7 For the purposes of this review, prodromes include all symptoms that precede the overt neurological symptoms of neurodegenerative diseases, focusing on psychiatric aspects. Another important distinction is that between neurodegenerative diseases and major neurocognitive disorders (NCDs). Although many patients with neurodegenerative diseases ultimately develop major NCDs, not all do. The focus of this review is on neurodegenerative diseases, not limited to major NCDs.
The purpose of this narrative review was to identify and highlight studies published in the last decade focusing on the recognition, diagnosis, and management of psychotic symptoms associated with the neurodegenerative disease continuum (including prodromal and manifest stages).
Methods
A first PubMed literature search was conducted using the terms (Alzheimer’s disease or amyotrophic lateral sclerosis or dementia or dementia with Lewy bodies or fragile X-associated tremor/ataxia syndrome or frontotemporal dementia or Huntington’s disease or Parkinson’s disease or spinocerebellar ataxia) and (psychosis or psychotic or delusions or hallucinations). A second search was then performed using the same terms and adding (prodrome or prodromal). A third search was conducted including each of the neurodegenerative diseases listed above and the term antipsychotic. In the next step, each antipsychotic medication (aripiprazole, brexpiprazole, cariprazine, clozapine, iloperidone, lurasidone, olanzapine, pimavanserin, quetiapine, risperidone, paliperidone) was used as a search term paired with each neurodegenerative disease, to identify any treatment studies that may have been missed. Additional articles were retrieved by examining the reference lists of the studies identified through the above search strategy, as well as textbook chapters. Case reports, case series, review articles, treatment guidelines, and clinical trials, including randomized clinical trials (RCTs) published in English between 2012 and 2022 were included. Primary sources were preferred over review articles, if both were available on a given topic. Articles published before 2012 were retained if newer evidence was not available or if they were “classics”, such as the Clinical Antipsychotic Trials of Intervention Effectiveness-Alzheimer’s Disease (CATIE-AD) study. 8 Publications referring only to the cognitive, autonomic, or other non-behavioral aspects of neurodegenerative diseases and those not meeting the definition of prodrome were excluded.
For the workup section, an outline of possible medical etiologies was developed based on our clinical experience and a previous comprehensive review article, 4 followed by targeted PubMed searches to identify publications that included older adults (for example, using the terms, neurosyphilis and psychosis and older). Figure 1 depicts the PRISMA flow diagram for literature search and article selection. 9
Figure 1.
PRISMA flow chart of literature search and article selection.
Results
Diagnostic Workup
The first step to elucidate the origin of new acute-onset psychotic symptoms in older adults is to seek underlying reversible causes, if any, and rule out delirium. A thorough workup, especially searching for infectious etiologies such as urinary tract infection (UTI), pneumonia, and viral encephalitis will help uncover systemic medical conditions that can cause acute mental status changes. It is also important to inquire about any recent medication changes (prescribed and over the counter), as well as alcohol and drug use. In particular, agents with anticholinergic properties, benzodiazepines, and opioids can contribute to delirium.10,11
Delirium occurs in up to 50% of hospitalized older adults and can persist for weeks after the underlying cause is treated. 12 Risk factors include age over 75 years; cognitive impairment, especially major NCD; prior history of delirium; history of cerebrovascular accident or transient ischemic attack; functional, visual, or hearing impairment; comorbidity or severity of medical illness; depression; and alcohol use disorders. 12 Delirium manifests with attention fluctuations and sleep-wake cycle disturbances; disorientation is not as common as previously thought. 13 Hallucinations (more frequently visual than auditory) can occur in 40-70% of patients, while delusions (often persecutory) have been described in 25-79% of individuals who experience delirium.4,13
Once delirium is ruled out, the workup of late-life psychotic symptoms includes routine blood tests (complete blood count, electrolytes, glucose, and hepatic and renal function tests), thyroid stimulating hormone, vitamin B12, and urine drug screen.4,14 Additional laboratory tests include erythrocyte sedimentation rate, folate, heavy metals, rapid plasma reagin, and human immunodeficiency virus and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) tests. 14 Lumbar puncture can help elucidate an infectious etiology of meningitis or encephalitis and support a diagnosis of multiple sclerosis or autoimmune encephalitis, although the latter two conditions seldom present de novo in older adults. 15 Electroencephalograms can aid in diagnosing seizure disorders, which can also be associated with psychosis. 16 Neuropsychological testing is helpful in differentiating depression from major NCDs and identifying patterns of deficits that may indicate specific neurodegenerative diseases. For example, cognitive deficits outlining a hippocampal pattern (poor recall, flat learning curve, and poor recognition) along with language and visuospatial skills impairment are pathognomonic for AD.17,18 Patients with AD forget new information rapidly and do not benefit from cues; list-learning tasks are most sensitive for detecting the memory deficits in early AD. 19 Patients with the behavioral variant of frontotemporal dementia (bvFTD) or frontal AD variant (also known as behavioral dysexecutive AD variant) show profound executive dysfunction.17,18,20 Individuals with movement disorders such as Parkinson’s disease (PD) or Huntington’s disease (HD) demonstrate a frontal-subcortical deficit pattern, with executive dysfunction and poor spontaneous recall, but generally preserved learning and intact recognition (cues help).17,21 Mixed major NCDs such as dementia with Lewy bodies (DLB) and fragile X-associated tremor/ataxia syndrome (FXTAS) combine hippocampal and frontal deficit patterns.18,22,23
Depression is often associated with neurodegenerative diseases such as AD, PD, and HD. In a study of adults aged 60-89 evaluated on 5 cognitive domains (verbal episodic memory, executive function, processing speed, constructional praxis, and language/semantic memory), patients with late-life depression scored below the 5th percentile in at least two domains, while people with mild AD had lower scores in 3 or more domains. 24 Patients with depression also tend to give more “I don’t know” answers and show impaired performance on measures of learning and free recall as well as tests that rely on attention, effort, and speed. 25 Older adults with depression perform generally better than people with AD and demonstrate retrieval difficulties on memory testing, as opposed to amnestic deficits, typical of AD. 25 In contrast, the neuropsychological profile in PD is quite similar to the one found in depression, so it is harder to differentiate. 25 Neuropsychological evaluation results should be interpreted in clinical context.
Finally, neuroimaging studies, especially brain magnetic resonance imaging (MRI), can corroborate clinical findings and support the diagnosis.26,27 Brain MRIs are particularly helpful when evaluating patients with gradual cognitive decline, acute-onset confusion, or behavioral changes and are best to identify subcortical pathology, white matter, and microvascular ischemic changes.19,26 Head computerized tomography (CT) scans help diagnose intracranial hemorrhage, acute cerebrovascular accidents (ischemic or hemorrhagic), mass-occupying lesions, normal pressure hydrocephalus, and reveal cortical atrophy with resulting ventricular enlargement. 19 Functional imaging studies such as positron emission tomography (PET) and single photon emission computed tomography (SPECT) can detect neuronal metabolic abnormalities before changes are visible on structural imaging investigations such as CT or MRI, although they are not readily available at all hospitals. 27 Patterns of cortical atrophy or hypometabolism identified through neuroimaging can help distinguish neurodegenerative diseases, although findings should always be interpreted in clinical context. A posterior (predominantly parietal and hippocampal) configuration can be found in AD, whereas an anterior (frontal and/or anterior temporal) pattern is common in bvFTD.20,26
Table 1 highlights laboratory tests and procedures that can uncover potentially reversible etiologies of late-life psychotic symptoms.
Table 1.
| Test or procedure | Clinical significance of abnormal results |
|---|---|
| Laboratory test | |
| Blood urea nitrogen, creatinine | Renal disease |
| Complete blood count | Infection |
| Lymphoproliferative disorders | |
| Electrolytes | Hyponatremia/hypernatremia |
| Hypokalemia (e.g., Cushing syndrome) | |
| Glucose | Hypoglycemia/hyperglycemia |
| Glycated hemoglobin (HbA1c) | Diabetes mellitus |
| Liver function tests | Hepatic disease |
| Ammonia | Hepatic encephalopathy |
| Antinuclear antibody | Systemic lupus erythematosus |
| Rheumatologic disease | |
| Thyroid function testsa | Hypothyroidism/hyperthyroidism |
| Urinalysis | Urinary tract infection |
| Urine drug screen | Substance intoxication/withdrawal |
| Vitamin B12 | Vitamin B12 deficiency |
| Folate | Folate deficiency |
| Vitamin D | Vitamin D deficiency |
| Erythrocyte sedimentation rate | Inflammation |
| Vasculitis | |
| Temporal arteritis | |
| SARS-CoV-2 tests | COVID-19 |
| Calciumb | Hypocalcemia/hypercalcemia (e.g., hyperparathyroidism) |
| Heavy metals | Intoxication |
| HIV | HIV-associated neurocognitive disease |
| Rapid plasma reagin; fluorescein treponema antibody | Neurosyphilis |
| Procedures | |
| Chest radiography | Pneumonia |
| Lumbar puncture | Autoimmune encephalitis |
| Meningitis | |
| Multiple sclerosis | |
| Viral encephalitis | |
| Electroencephalogram | Creutzfeldt-Jakob disease |
| HSV encephalitis | |
| Seizures | |
| Head computerized tomography | Acute cerebrovascular accident |
| Mass-occupying lesions (e.g., tumor, infection) | |
| Subdural hematoma | |
| Brain magnetic resonance imaging | Autoimmune encephalitis |
| CNS vasculitis | |
| Microvascular ischemic changes | |
| Multiple sclerosis | |
| Normal pressure hydrocephalus | |
| Neurodegenerative disease | |
CNS = central nervous system; COVID-19 = coronavirus disease 2019; HIV = human immunodeficiency virus; HSV = herpes simplex virus; SARS-CoV-2 = severe acute respiratory syndrome coronavirus-2.
a5-15 of patients with severe hypothyroidism may develop psychosis.
bBoth severe hypo- and hypercalcemia can be associated with psychosis.
Table 2 summarizes systemic medical diseases, substances, and medications that may be associated with psychotic symptoms in older adults.
Table 2.
Systemic Medical Diseases, Substances, and Medications Associated with Psychotic Symptoms in Older Adults.4,11,17,25,28-30,32-42
| Etiology | Examples |
|---|---|
| Substance intoxication | Alcohol (alcoholic hallucinosis) |
| Cannabis | |
| Cocaine | |
| Hallucinogens (ketamine, LSD, mushrooms, PCP, psilocybin) | |
| Methamphetamine | |
| MDMA | |
| Substance withdrawal | Alcohol |
| Sedative, hypnotic, anxiolytic (e.g., benzodiazepine) | |
| Opioids | |
| Medications | Anticholinergic and antihistaminic agents (benztropine, cimetidine, diphenhydramine, hydroxyzine) |
| Antiparkinsonian agents (e.g., amantadine, bromocriptine, levodopa, pramipexole, ropinirole, rotigotine, trihexyphenidyl) | |
| Antibiotic-associated encephalopathy (quinolones, macrolides, procaine penicillin) | |
| Anticonvulsants (levetiracetam, zonisamide) | |
| Corticosteroids | |
| Digoxin | |
| Interferon | |
| Opioids | |
| Psychostimulants (e.g., amphetamine, dextroamphetamine, lisdexamfetamine, methylphenidate) | |
| Metabolic and endocrine conditions | Acute intermittent porphyria |
| Adrenal disease | |
| Electrolyte imbalances (e.g., sodium, potassium, calcium) | |
| Heavy metals (e.g., lead, mercury) | |
| Hepatic failure | |
| Hypoglycemia/hyperglycemia | |
| Hypoparathyroidism/hyperparathyroidism | |
| Hypothyroidism/hyperthyroidism; Hashimoto’s thyroiditis | |
| Renal failure | |
| Vitamin deficiencies (B12, D, folate) | |
| Neurological conditions | Alzheimer’s disease |
| Amyotrophic lateral sclerosis | |
| Autoimmune encephalitis | |
| Brain tumors | |
| CADASIL | |
| Cerebral vasculitis | |
| Cerebrovascular accidents (basal ganglia, cerebellar, frontal, temporal, or parietal lobe) | |
| COVID-19 | |
| Dementia with Lewy bodies | |
| Frontotemporal dementia | |
| HIV infection | |
| Huntington’s disease | |
| Hypoxia | |
| Meningitis | |
| Multiple sclerosis | |
| Neurosyphilis | |
| Paraneoplastic syndromes | |
| Parkinson’s disease | |
| Prion disease (e.g., Creutzfekdt-Jakob) | |
| Seizure disorders | |
| Subdural hematoma | |
| Systemic lupus erythematosus | |
| Traumatic brain injury | |
| Viral encephalitis (HHV-6, HSV) |
AH = auditory hallucinations; CADASIL = cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy; COVID-19 = coronavirus-19 disease; HHV-6 = human herpesvirus-6; HIV = human immunodeficiency virus; HSV = herpes simplex virus; LSD = lysergic acid diethylamide; MDMA = 3,4-methylenedioxy-methamphetamine; PCP = phencyclidine.
Though infrequently used in clinical practice, genetic testing can identify mutations linked to neurodegenerative diseases associated with psychosis. 27 For example, hexanucleotide repeat expansions caused by chromosome 9 open reading frame 72 (C9orf72) mutations are the most common genetic abnormality in behavioral variant frontotemporal dementia (bvFTD) and familial amyotrophic lateral sclerosis (ALS) and are associated with an increased risk for psychosis.43,44 A thorough interview, inquiring about family history of neurodegenerative diseases, is also important to identify genetic causes. 27
Table 3 highlights the most common neurodegenerative diseases caused by nucleotide repeat expansions.
Table 3.
| Disease | Transmission | Chr. | Gene | Protein | Nucleotide repeat |
|---|---|---|---|---|---|
| Hexanucleotide | |||||
| FTD/ALS; bvFTD | Autosomal dominant | 9 | C9orf72 | C9orf72 protein | GGGGCC |
| Trinucleotide | |||||
| FXTAS | X-linked | X | FMR1 | FMRP | CGG |
| Friedreich’s ataxia | Autosomal recessive | 9 | FXN | Frataxin | GAA |
| Huntington’s disease | Autosomal dominant | 4 | HTT | Huntingtin | CAG |
| Spinocerebellar ataxias | |||||
| ➢ Type 1 | Autosomal dominant | 6 | ATXN1 | Ataxin-1 | CAG |
| ➢ Type 2 | Autosomal dominant | 12 | ATXN2 | Ataxin-2 | CAG |
| ➢ Type 3 (Machado-Joseph disease) | Autosomal dominant | 14 | ATXN2 | Ataxin-3 | CAG |
ALS = amyotrophic lateral sclerosis; bvFTD = behavioral variant frontotemporal dementia; chr. = chromosome; C9orf72 = chromosome 9 open reading frame 72; FMR1 = fragile X mental retardation 1; FMRP = fragile X mental retardation protein; FXTAS = fragile X-associated tremor/ataxia syndrome.
Psychotic Symptoms in Manifest Neurodegenerative Diseases
Psychotic symptoms are fairly common in manifest neurodegenerative syndromes and portend a poor prognosis, being associated with cognitive and functional decline and earlier death.51-55
AD is the most common neurodegenerative disease and major NCD worldwide, accounting for over 60% of cases.56,57 Psychotic symptoms typically occur in the middle stage and resolve in advanced disease (or are no longer communicated as patients lose verbal abilities).51,57 A large study of National Alzheimer’s Coordinating Center (NACC) data also identified delusions in 12% and hallucinations in 3% of people with mild AD. 58 Overall, delusions (mainly paranoid, of theft, or misidentification) range in prevalence from 31% to 59% across studies.57,59-61 Reeves and colleagues 60 posited that persecutory delusions appear earlier in the course of AD and are associated with dysfunction in frontostriatal circuits, whereas misidentification delusions emerge later and reflect limbic system changes. Hallucinations can occur in 16-41% of patients; simple VH predominate, although AH can also be present.3,57,62 In a sample of 1,808 adults with AD and other major NCDs characterized neuropathologically, hallucinations were more common in patients with mixed AD/Lewy body disease pathology than in those with AD or Lewy body disease alone. 63
Presence of psychotic symptoms can skew clinical diagnosis. In a study of approximately 1,000 patients with pathologically-confirmed AD, patients with psychosis were five times more likely to be misdiagnosed with DLB, whereas those without psychosis were more likely to receive a false-positive diagnosis of AD, when in fact they had vascular lesions. 64
Psychotic symptoms occur late in the course of PD and may arise in cognitively intact people, not just those with major NCD.65,66 Well-formed VH are typical, with a lifetime prevalence approaching 60%; AH are less frequent and can occur in up to 20% of people with PD.52,66,67 Delusions (mainly of infidelity, paranoid, or of misidentification) have been described in 5-10% of patients.66-68 Minor psychotic phenomena include auditory or visual illusions, passage hallucinations (brief visions of a person or animal passing in the periphery of the visual field), and extracampine hallucinations (sense of presence), which can manifest in half of patients.67,69 Psychosis may also be caused or exacerbated by treatment with levodopa or dopamine agonists, 66 in which case delusions can take on a grandiose quality (e.g., having superpowers) and be associated with elevated mood, poor sleep, dopamine dysregulation syndrome, and/or impulse control disorders.
DLB is the third most common neurodegenerative disease in the U.S., after AD and PD. 56 The major NCD occurs prior to or within 1 year of parkinsonism onset, although not all patients will develop parkinsonism. 70 Recurrent, well-formed VH are present early and constitute a core clinical feature; hallucinations in other modalities and systematized delusions can also occur and are considered supportive features. 70 Almost half of patients can harbor delusions, and up to 80% display VH.61,70-72 Delusions of misidentification, including Capgras syndrome (the false belief that a familiar person, often a close family member or caregiver, has been replaced by an identical-looking impostor) are frequent.61,73,74 Approximately 36% of patients with DLB had AH in a recent study, and 61% exhibited VH; AH often accompanied VH, like a “soundtrack”. 75 Table 4 highlights the clinical features of DLB, updated by the DLB Consortium in 2017. 70
Table 4.
Clinical Features of Dementia with Lewy Bodies. 70
| Essential feature: dementia that occurs before or concurrently (within 1 year) with parkinsonism. |
|---|
| Core clinical features (the first 3 typically occur early and persist throughout the disease course): |
| 1. Fluctuating cognition with pronounced variations in attention and alertness |
| 2. Recurrent visual hallucinations, typically well-formed and detailed |
| 3. REM sleep behavior disorder, which may precede cognitive decline |
| 4. One or more spontaneous cardinal features of parkinsonism: bradykinesia, rest tremor, or rigidity |
| Supportive clinical features: |
| 1. Severe sensitivity to antipsychotic agents |
| 2. Postural instability |
| 3. Repeated falls |
| 4. Syncope or other transient episodes of unresponsiveness |
| 5. Severe autonomic dysfunction, e.g., constipation, orthostatic hypotension, urinary incontinence |
| 6. Hypersomnia |
| 7. Hyposmia |
| 8. Hallucinations in other modalities |
| 9. Systematized delusions |
| 10. Apathy, anxiety, and depression |
| DLB is less likely: |
| • If another systemic medical or neurological illness (e.g., cerebrovascular disease) is present |
| • If parkinsonian features are the only core clinical feature and appear for the first time at a stage of severe dementia. |
DLB = dementia with Lewy bodies; REM = rapid eye movement.
FTD is the second most common form of early-onset major NCD. 27 FTD is divided into two main clinical syndromes: bvFTD and primary progressive aphasia (PPA), further subdivided into nonfluent variant (nfvPPA) and semantic variant (svPPA). 76 The newly described logopenic variant of PPA (lvPPA) is associated with AD pathology in over 80% of cases and is considered an AD variant. 77 Behavioral manifestations are most salient in bvFTD and include apathy, loss of empathy, disinhibition, hyperorality, and compulsive behaviors, along with anxiety, depression, and executive dysfunction. 20 A parallel classification describes frontotemporal lobar degeneration (FTLD) as a group of clinically and neuropathologically distinct syndromes which includes the FTD syndromes, progressive supranuclear palsy (PSP), corticobasal degeneration (CBD), and bvFTD with motor neuron disease (FTD-MND), with the exemplar FTD-ALS. The FTLD subtypes and their corresponding genetic and neuropathological underpinnings known to date are detailed in an excellent review by Elahi and Miller. 18 Neuropathologically, the two most common FTLD subtypes are FTLD-tau (characterized by misfolded tau aggregate inclusions) and FTLD-TDP (resulting from the accumulation of 43-kDa transactive response DNA binding protein, TDP-43). 18 In a series of 97 patients with autopsy-confirmed FTLD, nearly one third experienced psychotic symptoms including paranoid ideation, delusions, or hallucinations (mainly visual). 78 Gossink et al. 79 prospectively followed 22 patients with probable or definite bvFTD for 2 years; 5 (23%) developed delusions, hallucinations, or suspiciousness.
Although usually associated with slower disease progression, C9orf72 mutations confer an increased risk of psychosis in both the prodromal and manifest FTD stages.43,44,59,77,80-82 Moreover, psychosis can be the presenting symptom.43,44 In a cohort of 398 individuals with bvFTD, nfvPPA, svPPA or an overlap of these syndromes, 32 patients were found to display C9orf72 mutations, of whom 12 (32%) presented with frank psychotic symptoms (primarily delusions), and 9 others exhibited bizarre behavior. 44 Delusions can be persecutory, somatic, grandiose or of infidelity, whereas hallucinations include all modalities.81,82 Benussi and colleagues 83 explored the progression of neuropsychiatric symptoms (NPS) over a decade in a large cohort comprised of 232 patients with bvFTD, half of whom harbored C9orf72 mutations. The prevalence of hallucinations increased from 10% shortly after diagnosis, to 23% 2 years later. Delusions were also common, ranging from 7% at diagnosis to 16% after 2 years. 83 However, there was significant sample attrition over time and few mutation carriers were seen after the 2-year follow-up point; for this reason, later data should be interpreted with caution.
Neuropathologically, C9orf72 mutations align with the FTLD-TDP subtype. 18 Naasan et al. 74 conducted a retrospective chart review of 372 people with pathologically characterized neurodegenerative syndromes, of whom 111 (27%) had endorsed psychotic symptoms. Patients with FTLD-TDP pathology were significantly more likely to have delusions, particularly in the first 3 years of the disease, when compared to the AD and FTLD-tau groups. Patients with FTLD-TDP were also more likely to display paranoid ideation and grandiose or erotomanic delusions compared to those with AD or FTLD-tau changes. 74
Psychosis is exceedingly rare in PSP and CBD syndromes, although case reports have been published.59,84
Up to 22% of patients with ALS meet FTD criteria; about half remain cognitively intact, and the remainder develop milder deficits and personality changes.85,86 NPS include apathy (which can be present in up to 80% of patients), depression, anxiety, disinhibition, psychosis, and stereotyped behavior.85,87-90 Psychotic symptoms are infrequent (1-2%) in patients with ALS without FTD. 88
HD is an autosomal dominant, fully penetrant neurodegenerative disease associated with > 36 CAG repeats, with onset usually in mid-adulthood. 91 Psychiatric manifestations include apathy, depression, agitation/aggression, anxiety, obsessive-compulsive features, mania, and psychosis.92-94 The prevalence of psychotic manifestations (predominantly delusions) in HD had been previously estimated at 3-12%.92,95 However, two recent large cohort studies found rates of 13-18%.54,96 The latter two studies did not differentiate hallucinations from delusions; hence, more granular prevalence data are not available. Psychotic symptoms can range from poorly systematized paranoid ideation and isolated delusions to florid schizophrenia-like psychosis. 97
Spinocerebellar ataxias (SCAs) are autosomal dominant, adult-onset neurodegenerative diseases of the cerebellum and spinal cord, although onset age can range from childhood to late life.98,99 There are over 40 SCA types currently known, the most common being SCA3 (also known as Machado-Joseph disease). SCAs often present with pure neurological phenotypes, although several subtypes can also be associated with cognitive impairment and psychiatric symptoms. 98 In addition to psychosis, neuropsychiatric features include depression, anxiety, aggression, and personality changes.100-103 The prevalence of psychotic symptoms is difficult to establish, in part because of the heterogeneity of this group of diseases. Case reports and series have depicted individuals with SCA type 1, 2, 3, 6, 7, 10, 14, and 17 who exhibited delusions or hallucinations.101-106 The largest study involved 112 patients with Machado-Joseph disease, of whom 5 (4%) reported psychotic symptoms; all had hallucinations (either auditory or visual), along with delusional or disorganized thinking. 103 Albeit rare, SCA17 involves the basal ganglia and has a clinical picture that mimics HD (chorea with cerebellar signs); psychotic symptoms may be more frequent.99,105
Psychosis is also infrequent in FXTAS, a neurodegenerative disease that affects older carriers of fragile X mental retardation (FMR1) gene premutations, with 55-200 CGG repeats. 45 FXTAS occurs in approximately 40-75% of male premutation carriers (with penetrance increasing with age) and 16-20% of female carriers and is characterized by intention tremor, cerebellar ataxia, parkinsonism, cognitive decline, and NPS.45,107,108 Neuropsychiatric features include anxiety, depression, disinhibition, impulsivity, mood lability, irritability, apathy, and agitation.107-111 In a study of 55 carriers with FXTAS, 5 (9%) met the Diagnostic and Statistical Manual (DSM) IV-TR criteria for psychotic disorders, reporting paranoid delusions or hallucinations (visual or olfactory). 112 Accelerated cognitive decline with or without VH has infrequently been described in individuals with gray zone alleles (45-54 CGG repeats) or in rare cases of FXTAS co-occurrence with other neurodegenerative processes, such as AD.113,114
Table 5 summarizes the clinical features of common neurodegenerative diseases that can present with psychosis during the manifest stage.
Table 5.
Clinical Features of Neurodegenerative Diseases Associated with Psychosis.45,50,66,70,71,75,79-81,92,96,98,99,103,107,112,115-117
| Neurodegenerative disease | AD | DLB | bvFTD | FXTAS | HD | PD | SCA |
|---|---|---|---|---|---|---|---|
| Typical onset age | > 65 y.o. a | 65-85 y.o. | 45-65 y.o. | > 50 y.o. | 40 y.o. b | > 65 y.o. c | 35-50 y.o. d |
| Sex ratio | F:M = 3:2 | M:F = 2:1 | M > F | M >> F | M = F | M:F = 3:2 | M = F |
| Core clinical features | Early memory & learning deficits | Fluctuating cognition | Early decline in personal & social conduct | Cerebellar ataxia | Choreoathetosis | Bradykinesia | Cerebellar ataxia |
| Language deficits | Recurrent, vivid VH | Apathy | Intention tremor | Dystonia (esp. juvenile form) | Resting tremor or rigidity | Pyramidal signs | |
| Visuo-spatial deficits | Parkinsonism | Poor insight | Parkinsonism | Bradykinesia | Postural instability Falls | Dysarthria | |
| REM sleep behavior disorder | Stereotyped/ compulsive behaviors | Nystagmus | |||||
| Other clinical features | Executive dysfunction (in frontal variant) | Neuroleptic sensitivity | Executive dysfunction | Primary ovarian insufficiency (F) | Apathy | Dystonia | Ophthalmoplegia |
| Carbohydrate craving | Migraines | Depression | Dyskinesia | Extrapyramidal signs | |||
| Thyroid disease | Suicidality | Autonomic dysfunction | Seizures | ||||
| Psychotic symptoms | |||||||
| Delusions | 9-59% | 50% | 2-23% (57% in C9orf72 mutation carriers) | 5% e | 12% f | 7-10% | 5% g |
| Hallucinations h | 16% (6-41%) | 80% | 18% (36% in C9orf72 mutation carriers) | 3% | 2% | 53-60% | 5% g |
| • Auditory | 8-12% | 36% | 9-10% | ─ | ─ | 22% | Case reports |
| • Visual | 7-23% | 60-80% | 7-9% | 1% | 2% | 16-38% | Case reports |
AD = Alzheimer’s disease; DLB = dementia with Lewy bodies; bvFTD = behavioral variant frontotemporal dementia; FXTAS = fragile X-associated tremor/ataxia syndrome; HD = Huntington’s disease; PD = Parkinson’s disease; SCA = spinocerebellar ataxia; F = female; M = male; VH = visual hallucinations; REM = rapid eye movement; C9orf72 = chromosome 9 open reading frame 72.
a5-6% of patients have early onset AD (before age 65).
b5-10% of patients develop a juvenile HD form, with onset before age 20.
c3-5% of patients have young onset PD (before age 50); there is a very rare juvenile form, with onset before age 21.
dOnset ages vary for different SCA types and can range from 3 to 75 years old.
eThis estimate is based on a single study; results need to be replicated.
fNewer studies in larger cohorts found 13-18% prevalence for psychosis (hallucinations not separated from delusions).
gPrevalence data based on a single study of SCA3 (Machado-Joseph disease).
hIncludes auditory, visual, olfactory, and minor phenomena, where present.
Psychotic Symptoms as Part of Neurodegenerative Disease Prodromes
This section covers common psychiatric prodromes of neurodegenerative diseases. It is important to note that subtle cognitive deficits can also occur in preclinical stages; however, a comprehensive review of cognitive prodromal manifestations is beyond the scope of this review. Several neurodegenerative disease prodromes are well characterized. For example, anosmia, constipation, and rapid eye movement (REM) sleep behavior disorder are part of a well-known prodrome encountered in synucleinopathies such as PD, DLB, and, less often, multiple system atrophy.118-124 To date, prodromes with psychiatric symptoms have been described in AD,125-132 ALS,88,89,133-135 bvFTD,5,136-139 DLB,121,124,126,127,140-145 FXTAS, 146 HD,91,93,96,147,148 PD,6,118-120,123 and SCAs.101,149 The most common prodromal NPS are apathy, anxiety, depression, and REM sleep behavior disorder. Albeit less frequent, psychosis can also occur in AD, ALS, bvFTD (especially among C9orf72 mutation carriers), DLB, and HD prodromes.93,96,125,128,133-138,143,145,148
The MBI construct, initially proposed by Taragano et al. 139 as a prodrome of FTD, was later expanded to describe an at-risk stage for all major NCDs. MBI has been extensively studied in recent years as part the AD prodrome, along with MCI. MBI is subdivided into 5 domains: reduced motivation/apathy, affective dysregulation, impulse dyscontrol, social inappropriateness, and abnormal perception or thought content (e.g., delusions, hallucinations) and can co-occur with or even precede MCI. 150 MBI prevalence varies depending on participant age, setting, and instrument used, ranging from 3.5% among outpatients aged ≥ 50 seen in a psychiatry clinic 151 to 14% of patients with MCI in a primary care practice 152 and 34% in a sample of 1,377 community-dwelling adults aged 72-79 years (49% in the MCI subgroup among them). 153 Patients with MBI were found to have a higher risk (hazard ratio, HR, 8.07) of converting to major NCD (mainly AD) over a follow-up period of up to 104 months compared to those with MCI without MBI (HR 7.05). 151 The most common MBI domain was affective dysregulation (64%), followed by abnormal perception or thought content (21%). 151 In a recent web-based longitudinal study of 8,181 older adults (median age, 63 years), 11% of women and 14% of men met MBI criteria. 154 All MBI domains were associated with cognitive decline; the association with psychosis had the largest effect size, but only in men, underscoring the importance of exploring sex differences. 154
In the NACC database analysis conducted by Apostolova et al., 3% of participants with amnestic MCI (often an AD precursor) and 4% of those with nonamnestic MCI had delusions, while 1% in each group exhibited hallucinations. 58 Ismail et al. 155 reported a 3.1-10.5% pooled prevalence of delusions and 1.3-2.6% frequency of hallucinations among individuals with MCI.
The prodrome that is perhaps best known to neurologists and psychiatrists is that of DLB, consisting of anxiety, apathy, depression, or REM sleep behavior disorder.121,122,124,126,127,140-144 Psychotic symptoms are also prominent.126,140,143,145 In a large cohort study including 148 patients with prodromal DLB, 64% had hallucinations and 13% displayed delusions. 140 Utsumi and colleagues 145 followed longitudinally 21 patients who presented with recurrent catatonia, delusions, or hallucinations and were diagnosed with DLB a decade later, on average. Wyman-Chick et al. 143 conducted a retrospective chart review of 116 NACC Uniform Data Set database participants who did not have a major NCD diagnosis initially and converted to DLB at a subsequent visit. The authors investigated NPS prevalence for up to 5 years prior to diagnosis. They found that 19% of participants had hallucinations and 14% endorsed delusions 4 years prior to DLB diagnosis, with VH incidence rising to 20-25% 2 years later. 143
Anxiety, apathy, depression, and irritability are common NPS which can occur as part of the AD prodrome, along with psychosis.58,129-132,156 Presence of psychotic symptoms increases risk for AD.125,128,156 In a large Italian nested case-control cohort study, medical records of 1,889 primary care patients who developed AD were examined retrospectively for 10 years prior to diagnosis. 128 Patients with a history of hallucinations had a four-times higher likelihood of developing AD; anxiety, agitation, aberrant motor behavior, memory deficits, and depression also increased risk. 128 In a Japanese study of 234 individuals with MCI (mean age, 73 years) followed for up to 3 years, 3% had psychotic symptoms. 156 Delusions were the only factor significantly associated with conversion to AD (unadjusted HR, 2.9). 156 Apolipoprotein E ε4 carriers with NPS have an even higher risk of progressing to AD.125,155 In an analysis of 11,453 cognitively intact NACC database participants, ε4 carriers who experienced delusions or hallucinations had a significantly higher chance of developing AD within several years compared to those without NPS; specifically, presence of delusions amplified the risk thirteen-fold. 125
Among all neurodegenerative diseases, bvFTD is the best mimic of psychiatric disorders. One third to half of patients are initially diagnosed with psychiatric conditions, and the bvFTD diagnosis can be delayed by 5-6 years.27,136,137 In a cross-sectional study based on standardized interviews with the caregivers of 46 people with bvFTD, 37% of patients had received a diagnosis of bipolar disorder (most often), schizophrenia, or schizoaffective disorder. 137 The association of FTD with ALS is also salient. Lillo et al. 133 examined a series of 18 patients with FTD-ALS who presented with behavioral changes. Patients with concurrent onset of behavioral and motor symptoms and those initially diagnosed with ALS who developed behavioral changes later were excluded. Nine (50%) of the 18 patients had delusions and 5 (28%) endorsed VH. 133 Delusions were persecutory, of theft, and erotomanic in nature. Presence of delusions predicted a subsequent diagnosis of ALS. 133
Turner et al. 134 found that hospital admissions for diagnoses of schizophrenia, bipolar disorder, depression, or anxiety were significantly associated with a new diagnosis of ALS in the following year. The association remained significant only for depression hospitalizations up to 5 years preceding ALS diagnosis. Similarly, Longinetti et al. 135 identified a significant association of depression, neurotic (anxiety, obsessive-compulsive, dissociative, and somatoform) disorders, and drug abuse/dependence, as well as other neurodegenerative diseases (FD, AD, PD, or other dementia) with risk of developing ALS. This association was strongest in the year preceding the ALS diagnosis but extended for the previous 2-5 years. 135
Psychosis is also part of the HD prodrome. In the European Registry of HD mutation carriers, 48% of participants had a motor onset, 20% first experienced psychiatric symptoms, and 8% had initial cognitive deficits, whereas 13% had a mixed presentation. 148 Almost 40% had a lifetime history of severe psychiatric disease, including psychosis, aggression, and suicidality. 148 In another large cohort study using the Enroll-HD international database, 3% of individuals with pre-manifest HD reported psychosis. 96 Other neuropsychiatric features of prodromal HD are apathy, depression, irritability, and anxiety. 93
Prodromal psychiatric manifestations are rare in SCAs, besides REM sleep behavior disturbances. 149 A case report depicted one man initially diagnosed with schizophrenia at age 22, who had persistent psychotic symptoms, progressively declined over time, and was ultimately found to have SCA2 ten years later. 101
Minor hallucinations can predate the onset of motor symptoms in PD. 157 A recent study also found an association between late-life psychosis and probability of having prodromal PD, calculated based on depression, constipation, and subthreshold parkinsonism. 158
Table 6 summarizes psychiatric prodromes of neurodegenerative diseases known to date.
Table 6.
| Neurodegenerative disease | Psychiatric prodrome |
|---|---|
| Alzheimer’s disease | Anxiety, apathy, depression, irritability, psychosis (especially in apoE ε4 carriers) |
| Amyotrophic lateral sclerosis | Anxiety, mood disorders (depression, bipolar), psychosis, substance use |
| Behavioral variant frontotemporal dementia | Anxiety, depression, psychosis (especially in C9orf72 carriers) |
| Dementia with Lewy bodies | Anxiety, apathy, depression, REM sleep behavior disorder, psychosis |
| FXTAS | Anxiety, depression |
| Huntington’s disease | Apathy, anxiety, depression, psychosis, aggression, suicidality |
| Parkinson’s disease | Anxiety, depression, REM sleep behavior disorder |
| Spinocerebellar ataxia | Psychosis (rare), REM sleep behavior disorder |
ApoE = apolipoprotein E; C9orf72 = chromosome 9 open reading frame 72; FXTAS = fragile X-associated tremor/ataxia syndrome; REM = rapid eye movement.
Diagnostic Criteria
Psychosis in Manifest Neurodegenerative Diseases
Jeste and Finkel 57 were the first to attempt to differentiate the psychotic symptoms associated with AD from those occurring in late-life primary psychotic disorders. They formulated separate diagnostic criteria for psychosis of AD. 57 Later, a National Institute of Neurological Disorders and Stroke – National Institute of Mental Health work group developed PD psychosis criteria. 159 Recently, an International Psychogeriatric Association (IPA) expert panel proposed clinical and research criteria for psychosis in major, as well as mild NCD, taking into consideration the fact that psychotic symptoms can occur earlier in the course of NCDs. 160 The International Society to Advance Alzheimer’s Research and Treatment (ISTAART) Professional Interest Area psychosis subgroup led by Fischer et al. 7 reviewed the previous criteria sets for psychosis of AD and other major NCDs, including those by Jeste and Finkel, 57 Lyketsos et al., 161 and DSM-5. In revising the AD psychosis criteria, Fischer and colleagues 7 advanced a new major NCD framework that includes neuroimaging and other biomarkers alongside clinical elements, thus mirroring current diagnostic criteria for other neurodegenerative diseases such as DLB and PSP.70,162 Of note, all the diagnostic criteria mentioned above, except for the ones regarding PD psychosis, imply the presence or later development of a major NCD.
Neurodegenerative Disease Prodromes
Research criteria have been proposed for prodromal PD and DLB.120,124,163 Psychiatric symptoms (depression ± anxiety) are included in the PD prodrome, although the emphasis is on other clinical aspects and biomarkers. 120 The DLB prodrome also includes clinical features and evidence-based biomarkers. 124 However, given the prominent psychiatric symptoms, the DLB prodrome has been classified in 3 subtypes: DLB-MCI, delirium onset-DLB (with provoked or spontaneous delirium), and psychiatric disorder-DLB (manifested primarily as late-onset mood or psychotic disorder).124,163 These presentations differ markedly from MBI symptoms and may be severe enough to require hospitalization. 124
ISTAART introduced diagnostic criteria for MBI in 2016. 150 MBI is defined as changes in behavior or personality reflected in the aforementioned 5 domains (reduced motivation, affective dysregulation, impulse dyscontrol, social inappropriateness, and abnormal perception or thought content) that start at or after age 50, are observed by patients, informants, or clinicians, and persist at least intermittently for 6 months or longer. 150 Symptoms have to be of sufficient severity to cause impairment in interpersonal, social, or occupational functioning. Having a primary psychiatric disorder or a major NCD are exclusionary criteria. MBI can be diagnosed concurrently with MCI, but co-occurrence is not necessary. A 34-item MBI-Checklist was also developed and validated to aid in clinical diagnosis and research studies.164,165
Management of Psychotic Symptoms in Neurodegenerative Diseases
Behavioral strategies are always recommended as first step in the management of NPS in neurodegenerative diseases. The American Psychiatric Association Guidelines for treatment of psychosis and agitation in AD suggest using antipsychotic medications only after behavioral approaches have failed, and if there is risk of self-harm or harm to others. 166 An international panel of experts recommended the DICE (Describe, Investigate, Create, and Evaluate) 167 model and music therapy to address overall NPS, agitation, and psychosis. 168 No RCTs of behavioral interventions for psychosis associated with neurodegenerative diseases have been conducted to date. Diederich et al. 169 studied 46 patients with PD and classified the strategies they used to cope with VH into 3 categories: visual (focusing better on the hallucinatory object, looking away from it, or focusing on another object), cognitive (turning the light on or telling themselves these phenomena are not real and will resolve shortly), and interactive (discussing with family and caregivers for reality testing and reassurance). Regardless of cognitive status, patients used cognitive strategies most often (69%), followed by interactive (62%), then visual (33%) techniques. 169
There are few RCTs for the pharmacological management of psychosis in major NCDs, and even fewer published in the last decade.170,171 Most treatment recommendations are based on anecdotal evidence and expert consensus.166,168,170-172 A recent network meta-analysis suggested that aripiprazole might be the most effective and safe antipsychotic for treating the behavioral and psychological symptoms of major NCDs; however, psychosis outcomes were not analyzed separately. 173 Of note, all antipsychotics have a boxed warning regarding the increased risk of death for older adults with major NCD-related psychosis. 174 There are no data on the newer antipsychotics brexpiprazole, cariprazine, iloperidone, or lurasidone.
Alzheimer’s Disease
The CATIE-AD study examined the effectiveness and safety of second-generation antipsychotics for AD-associated agitation or psychosis. In this multi-site, double blind RCT, 421 outpatients received olanzapine (mean dose, 5.5 mg/day), quetiapine (mean dose, 56.5 mg/day), risperidone (mean dose, 1 mg/day), or placebo. 8 There were no differences across groups regarding effectiveness or time to discontinuation, 8 but there was a modest advantage for olanzapine and risperidone in improving NPS. 175 In an earlier systematic review by Sink et al., 176 olanzapine 5-10 mg/day and risperidone 1mg/day also showed significant, but modest effects in reducing delusions, hallucinations, and aggression associated with AD or vascular major NCD.
Aripiprazole, a partial D2 receptor agonist, showed benefit in an RCT of 487 nursing home residents with AD psychosis who were randomized to placebo or aripiprazole 2, 5 or 10 mg/day. Aripiprazole 10 mg daily dose resulted in a significant improvement in the Neuropsychiatric Inventory-Nursing Home (NPI-NH) Psychosis subscale at ten weeks, in contrast to placebo. 177
There have been few newer RCTs for treatment of psychosis in AD. The expert panel listed risperidone as first line and pimavanserin, discussed in detail below, and citalopram as promising alternatives. 168 A phase II RCT including 181 nursing home residents with psychosis associated with AD showed benefit for pimavanserin 34 mg/day over placebo at 6 weeks, but differences were not maintained at 12 weeks. 178 In a phase III trial that was stopped early for efficacy, patients with psychosis related to AD, PD, DLB, FTD, or vascular major NCD received open-label pimavanserin 34 mg daily for 12 weeks. 179 Half of the responders were assigned to continue pimavanserin, and half to placebo. Responders had a lower risk of psychosis relapse if they continued pimavanserin, as opposed to stopping it. 179
The Citalopram for Agitation in Alzheimer's Disease (CitAD) trial demonstrated a significant improvement in agitation associated with AD with citalopram up to 30 mg daily. 180 A secondary analysis also found a reduction in delusions and hallucinations, with the best response noted in patients with concurrent agitation and psychosis. 181 Of note, the maximum FDA-recommended citalopram daily dose for adults over age 60 is 20 mg. 182
A 12-week double-blind RCT of low-dose lithium (150-600 mg daily) for agitation in AD was negative with regard to the primary outcome. 183 Exploratory analyses yielded a statistically significant superior improvement in delusions, but not hallucinations, with lithium vs. placebo.
Secondary analyses of data from cholinesterase inhibitors (ChEIs) trials in AD have revealed potential benefit or reduced emergence of psychosis, although there is no RCT demonstrating specific benefit of ChEIs in the management of AD psychosis. 171 In a study of over 17,000 individuals without prior psychotropic use from the Swedish Dementia Registry Study, use of ChEIs, particularly at higher doses, was associated with a lower likelihood of starting treatment with antipsychotics in patients with AD, but not DLB. 184
Parkinson’s Disease
Since psychotic symptoms can be induced or exacerbated by dopaminergic medications, an important first step is to attempt to reduce these agents, as tolerated without worsening of motor symptoms. This should be done in collaboration with treating neurologists. Experts recommend reducing or stopping PD medications in the following order: anticholinergic agents, amantadine, dopamine agonists, monoamine oxidase inhibitors, catechol-O-methyltransferase inhibitors, and lastly, levodopa. 185 If this strategy is ineffective, second-generation antipsychotics (never first-generation) can be used cautiously.
A recent systematic review and meta-analysis summarized all RCTs of antipsychotics used for PD psychosis. 186 There have been no new RCTs since 2012, with the exception of the pimavanserin trials described below. Pimavanserin is the only FDA-approved agent for psychosis in a neurodegenerative disease (specifically, for delusions and hallucinations of PD) and was recommended by an expert panel as first line for PD psychosis. 172 Pimavanserin has a novel mechanism of action, acting as a selective serotonin 5HT2A receptor inverse agonist. Cummings et al. 187 reported significant improvements in psychotic symptom scores in a phase III 6-week long RCT comprising 199 adults (mean age, 72.4 years) with PD psychosis. This trial was followed by a 4-week open label extension, during which all participants received daily pimavanserin 34 mg and the Scale for the Assessment of Positive Symptoms for Parkinson's Disease Psychosis (SAPS-PD) scores continued to drop. 188 Almost half of patients experienced side effects, leading to study discontinuation in 5.9% of cases. 188 Pimavanserin takes 10-12 days to reach steady state and may need up to 6 weeks to achieve effect; the experts recommended discontinuing it after 8 weeks, if no benefit is noted. 172 Further safety data, particularly in older adults, need to be collected. In 2018, the Food and Drug Administration did not find any new or unexpected safety risks in an analysis of all postmarketing reports of death and serious adverse events reported. 189 However, in a retrospective cohort study of adults aged 65 or older with PD (of whom 2,186 were taking pimavanserin and 18,212 were not taking it), pimavanserin users had a significantly higher risk of hospitalization at 30 days and higher risk of death for up to 1 year after starting treatment, compared to non-users. 190
Clozapine and quetiapine have low D2 receptor binding affinity; as such, they are least likely to cause motor symptom worsening. Clozapine is the only agent with sufficient evidence, based on two earlier RCTs and one meta-analysis that supported its efficacy in improving PD psychosis, with questionable benefit for motor symptoms.172,186,191 Clozapine requires frequent monitoring and can potentially have severe adverse effects such as agranulocytosis, albeit rare. 191
Quetiapine is preferred in practice, although there is mixed evidence for its use.186,191,192 In a chart review of 5,297 Veterans Affairs patients with PD psychosis, quetiapine accounted for two thirds of antipsychotic prescriptions. 191 Horn et al. 193 conducted a retrospective study comparing pimavanserin to quetiapine for psychosis in patients 41-97 years old (mean age, 73 ± 8 years) with PD or DLB. Patients in the pimavanserin group were more likely to have a diagnosis of DLB and to have tried an antipsychotic medication previously. Pimavanserin was more often discontinued due to refractory psychosis (i.e., lack of efficacy), while quetiapine was more likely to be discontinued due to side effects. 193
Evidence regarding the efficacy of ziprasidone in psychosis or acute agitation associated with PD is mixed, consisting mostly of case reports and a case series.194,195 Although generally well tolerated in PD, ziprasidone caused worsening of motor symptoms in patients with DLB. A 4-week, randomized, single-blind, open-label study including 14 patients compared ziprasidone (final mean dose, 35 mg/day) to clozapine (final mean dose, 32 mg/day) and found ziprasidone to be at least as effective as clozapine for PD psychosis. 195
In a recent open-label trial, aripiprazole at 3 mg/day appeared effective, however 8 of the 24 participants reported worsening of motor symptoms. 196 Olanzapine, risperidone, and aripiprazole should be avoided, based on evidence to date and expert recommendations.172,186
The ChEIs donepezil, galantamine, and rivastigmine have also been used for PD psychosis. However, the evidence is scarce and the results, unsatisfactory. Studies performed prior to 2012 were summarized in a Cochrane database systematic review. 197 Of note, most studies were designed to evaluate cognitive functioning in patients with major NCD as primary outcome, with psychotic symptoms as secondary outcomes. There are few studies in cognitively intact people with PD and psychosis. 198 One recent RCT randomized patients with PD and minor VH to rivastigmine 3 or 6 mg twice daily or placebo but was stopped early due to slow recruitment. 199 The study was not adequately powered to evaluate primary outcomes, but the authors reported no group differences in progression to frank psychosis or major NCD during the 2-year follow up period. 199
Anecdotal reports describe the benefits of antidepressants (citalopram, escitalopram, clomipramine, mianserin, mirtazapine, and venlafaxine) for the treatment of psychotic symptoms in patients with PD with or without comorbid depression, including one case report of a 67-year-old man whose VH were refractory to quetiapine, then clozapine, but improved with mirtazapine 30 mg at bedtime. 200 The evidence regarding antidepressant use for PD psychosis is weak, limited to case reports or series.200-203
Electroconvulsive therapy (ECT) was shown to improve depression, psychosis, and motor symptoms in patients with PD, although the motor improvement was short-lasting.204,205
Dementia with Lewy Bodies
Patients with DLB are highly sensitive to antipsychotic parkinsonian side effects and may experience motor symptom exacerbation.70,172 Severe sensitivity to neuroleptics is a supportive clinical feature for a probable DLB diagnosis. 70 For this reason, less potent D2 receptor blockers like quetiapine or clozapine are preferred. Quetiapine is favored in practice, although there is insufficient evidence for its efficacy.193,206,207 Several case reports have shown mixed results for clozapine, but trial data are lacking.206-208 Due to its novel mechanism of action, pimavanserin shows promise, yet its efficacy has not been proven to date. 193 Similar to PD, olanzapine and risperidone can cause motor worsening and have unclear benefit, so they are best avoided.206,207 One case report revealed benefit of low-dose aripiprazole without exacerbation of parkinsonism but more data are needed. 72
ChEIs have also been used for treating psychosis in mild to moderate DLB. The evidence is stronger than in PD. In an RCT of 140 patients with DLB randomized to receive donepezil 3, 5 or 10 mg daily or placebo for 12 weeks, there was significant improvement in hallucinations and delusions with donepezil 5 and 10 mg doses. 209 The benefit was maintained over a 52-week open label phase. 210 The same group conducted a 16-week phase III trial, followed by a 36-week open label arm using donepezil 10 mg daily. 211 Among the 100 patients who completed the study, there was no difference compared to placebo. 211 In a small study including 8 patients with DLB and VH, donepezil 10 mg daily was effective in treating symptom recurrence after initial resolution with 5 mg daily. 212
ECT has also been tried for patients with DLB, but benefits are less robust compared to PD.204,213
Behavioral Variant Frontotemporal Dementia
Studies suggest a dopaminergic deficit in bvFTD. 214 As such, quetiapine or aripiprazole may be preferred.215,216 There have been several case reports and case series using olanzapine, quetiapine, and risperidone for psychosis in bvFTD, but no RCTs.82,86,217,218 Clozapine 400 mg daily was helpful for a 26-year-old man with an initial diagnosis of schizoaffective disorder (later changed to FTD) with treatment-refractory psychosis and severe aggression. 219 As discussed above, there is insufficient evidence for pimavanserin. 179 The cholinergic system is only mildly affected in FTD.18,214 As such, ChEIs are not beneficial for cognition and may actually worsen behavioral symptoms.86,216
Huntington’s Disease
There are no RCTs of antipsychotics in HD; most treatment recommendations come from anecdotal experience and expert consensus.50,220 In practice, strong D2 blockers like haloperidol, risperidone, or olanzapine or the partial D2 agonist aripiprazole have been used to manage both chorea and psychosis.148,221-223 Aripiprazole is also recommended for treating psychosis with prominent negative symptoms. 50 Less potent D2 blockers such as quetiapine and clozapine can also be helpful. Quetiapine showed positive results in case reports and a series comprised of 5 patients,97,223,224 whereas clozapine was found to be effective in two middle-aged adults with treatment-refractory HD psychosis.225,226 The International Guidelines for the Treatment of Huntington’s Disease suggest clozapine as first line for psychosis in patients with akinetic HD and disabling parkinsonian symptoms. 220 HD psychosis can be severe and relentless; in some cases, combinations of two antipsychotics are necessary, although this should be avoided as much as possible due to risk of tardive dyskinesia and worsening parkinsonism, which often emerges later in the course of the disease.223,227
ECT has been used for medication-refractory cases, with mixed results.220,225
Spinocerebellar Ataxias
Given the low frequency of psychotic symptoms in SCAs, there is scarce evidence on the use of antipsychotics. Okamoto and colleagues 106 described a 43-year-old man with a 15-year history of SCA3, who was admitted for depression and somatic and grandiose delusions. His depression and grandiosity responded to escitalopram 10 mg and olanzapine 2.5 mg daily; however, the somatic delusions persisted. 106 Two other patients in the case series by Turk et al. 105 were treated with olanzapine, and one with risperidone (doses not mentioned). Wexler and Fogel described a 37-year-old man with SCA type 10 and new-onset psychosis and violent behavior, whose symptoms responded to risperidone 2 mg twice a day. 102
There are anecdotal reports on the use of quetiapine for psychosis in FXTAS , given that parkinsonian features may be part of the clinical picture. 228 Larger studies are needed. Table 7 summarizes the evidence discussed above.
Table 7.
Antipsychotic Medications Used for Treatment of Psychotic Symptoms in Neurodegenerative Diseases.8,72,82,92,97,168,179,187-189,192,194,207,217-219,221-225,229,230
| Medication (starting/target daily dose) | Strength of evidence/Other information |
|---|---|
| Alzheimer’s disease | |
| Risperidone 0.25 mg/1 mg | Good RCTs; modest effect in CATIE-AD |
| Olanzapine 2.5 mg/5-10 mg | Few RCTs; modest effect in CATIE-AD |
| Quetiapine 12.5 mg/200 mg | Negative RCTs; positive small case series |
| Aripiprazole 2 mg/10 mg | One positive RCT in nursing home patients |
| Pimavanserin 34 mg/34 mg | Insufficient evidence |
| Parkinson’s disease | |
| Clozapine 6.25 mg/50-75 mg | Few early RCTs; maximum dose reported 225 mg daily |
| Quetiapine 12.5 mg/125-225 mg | Mixed evidence; maximum dose reported 400 mg daily |
| Ziprasidone 20 mg/40 mg | Case reports/series, one open-label randomized trial |
| Pimavanserin 34 mg/34 mg | RCTs; mixed long-term and safety data to date |
| Dementia with Lewy bodies | |
| Quetiapine 12.5 mg/175 mg | 1 RCT, 1 case series; mixed results |
| Clozapine 6.25 mg/50 mg | No studies |
| Aripiprazole 1.5 mg/3 mg | Case report |
| Pimavanserin 34 mg/34 mg | Insufficient evidence |
| BvFTD | |
| Risperidone 1 mg/6 mg | Case reports |
| Olanzapine 2.5mg/10 mg | Case series |
| Quetiapine 25 mg/150-800 mg | Case reports |
| Clozapine 12.5 mg/400 mg | Case report |
| Pimavanserin 34 mg/34 mg | Insufficient evidence |
| Huntington’s disease | |
| Risperidone 0.5-1 mg/3-6 mg | Case reports |
| Olanzapine 2.5 mg/10-20 mg | Case reports |
| Aripiprazole 2.5 mg/10-30 mg | Case reports |
| Quetiapine 25 mg/300-600 mg | Case reports; case series |
| Clozapine 12.5 mg/450-500 mg | Case reports |
CATIE-AD = Clinical Antipsychotic Trials of Intervention Effectiveness–Alzheimer’s Disease; RCT = randomized controlled trial; bvFTD = behavioral variant frontotemporal dementia.
Discussion
Psychotic symptoms associated with neurodegenerative diseases are multifaceted and can occur as part of both prodromal and manifest disease stages. Psychiatric prodromes constitute a newer area of interest, which has yet to be fully elucidated. Knowledge is evolving, and studies of increasing quality have been published over the last decade. This narrative review summarized the extant body of knowledge. The current terminology is heterogenous. Most authors agree on the term “prodrome”, while others used the terms “preclinical”, “premanifest”, “premotor”, or “predementia”. The ISTAART AD psychosis criteria distinguish the “preclinical” from the “prodromal” stage (the latter encompasses MBI and MCI). 7
The neurodegenerative diseases included in this review were selected based on their prevalence in the general population (in descending order): AD, PD, DLB, FTD, HD, ALS, and SCAs. 56 Conditions that have a described psychiatric prodrome, such as FXTAS, were also of interest. The strength of the evidence varied. Studies ranged from case reports,101,102,106,121,138,141 case series,5,79,82,91,122,133 retrospective chart or database reviews,58,96,128,133-136,140,143,146-148 and cross-sectional analyses 137 to prospective longitudinal studies.83,145 Including case reports diluted the strength of the evidence summarized; however, a higher level of evidence, particularly with regard to treatment, is lacking at this time for less common neurodegenerative diseases (e.g., HD, SCAs, even bvFTD – see Table 7).
Psychiatric prodromes can predate overt neurodegenerative disease manifestations by 1-5 years in AD and ALS, 5-10 years in DLB, and two-to-three decades in bvFTD, FXTAS, HD, PD, and certain SCAs.6,93,96,101,119-124,134,137,138 This is clinically relevant, because patients may present with psychiatric symptoms long before they develop full-blown neurological syndromes. Hence, accurate recognition of psychiatric prodromes is paramount. Hallucinations are more common in the DLB prodromal stage,140,143 whereas delusions are more frequent during the AD and bvFTD prodromes.58,125,133,154-156 Of note, prodromal psychotic symptoms, particularly delusions, have been associated with an increased likelihood of receiving a neurodegenerative disease diagnosis within several years.125,133,154,156 In PD, delusions are considered to represent a deterioration of hallucinations, as distorted thought processes crystallize around misperceptions. 66 As such, delusions may indicate more advanced disease, being succeeded by overt cognitive or motor manifestations shortly thereafter.
Accurate identification of the neurodegenerative disease prodromes will also allow early intervention with disease-modifying treatments, as these become available. The ISTAART MBI criteria 150 are an excellent, albeit not disease-specific, starting point for a broader definition of major NCD prodromes. Nevertheless, the MBI syndrome does not entirely overlap with neurodegenerative disease prodromes. In the AD literature (and, to some extent, other major NCDs), the prodromal stage only includes MCI and MBI (much closer in time to the onset of clinically relevant cognitive deficits), while earlier manifestations are deemed preclinical. 7 However, in the movement disorders literature (PD, DLB, ALS), prodromes are a broader concept, extending decades before motor symptoms occur and including autonomic and other clinical features, along with neuroimaging, genetic, and other biomarkers.120,124,163 While MBI is considered a pre-dementia stage, not all patients with neurodegenerative diseases and psychiatric prodromes (e.g., ALS, SCAs) will develop major NCDs.85,98 The MBI onset age is ≥ 50 years, yet psychiatric prodromal symptoms can occur in people as young as their 20’s.6,101,137,138,146 Lastly, presence of a psychiatric disorder precludes an MBI diagnosis. 150 However, the majority of the studies focusing on prodromes reviewed here indicate that patients are often diagnosed with various psychiatric disorders prior to receiving a neurodegenerative disease diagnosis. These distinctions will be important to keep in mind as research and clinical knowledge advance and the neuropsychiatric field moves toward a unified prodrome definition and nomenclature. One possible expanded prodrome definition involves the emergence of NPS that precede the cognitive or motor manifestations of neurodegenerative diseases, as early as several decades in advance. Further research is needed to operationalize and validate a broader prodrome construct.
Mild psychotic symptoms can occur in cognitively intact, community-dwelling older adults.7,155 Two recent studies revealed a prevalence of 0.7-0.8% for delusions and 0.2-0.3% for hallucinations among cognitively intact older adults.153,231 These may represent prodromal symptoms that have not yet reached clinical significance, although late- or very late-onset primary psychotic disorders should enter the differential as well. 232
There is a debate in the field whether psychiatric symptoms represent a prodrome or a risk factor for neurodegenerative diseases. 53 Presence of NPS, especially apathy and depression, predicts MCI conversion to major NCD, specifically in AD and vascular major NCD.55,131,233-237 Although an association between mood disorders and risk of major NCD had previously been reported in large epidemiological studies,235,238 a recent systematic review and meta-analysis was the first to reveal a significant association of psychotic disorders with subsequent development of AD or vascular major NCD. 239 It is important to note that the association of NPS with major NCDs does not imply causality, and the statistical notion of risk (the probability that an event will occur) is not equivalent to biological vulnerability. 240 Therefore, it is more appropriate to substitute “probability” for “risk” when we interpret the results of these epidemiological studies.
As discussed above, presence of NPS, particularly delusions, may indicate a high likelihood that the overt stage of the disease will soon follow. The present review summarized a rich body of evidence supporting the idea that NPS are early manifestations of neurodegenerative diseases, particularly when they arise later than the typical onset age for primary psychiatric conditions in the general population.5,6,146 The NPS phenomenology also appears to be different in neurodegenerative prodromes, although more research is needed to state this with confidence. For example, sudden onset of severe anxiety around trivial stressors in a previously highly-functioning, well-adjusted individual in their mid-40’s who had never experienced such challenges should prompt consideration of a non-primary psychiatric etiology. 6 Ghahremani et al. found correlations between levels of a well-validated AD biomarker, plasma phosphorylated tau at threonine 181 (p-tau181) and MBI, although not individual NPS. 241 Over a 1-year period, MBI was associated with higher p-tau181 levels as well as decline in memory and executive function. These results highlight the significance of MBI as a neurodegenerative disease marker.
It is not entirely clear why NPS occur decades prior to the onset of neurological changes. One hypothesis concerns the selective vulnerability of certain brain areas such as the amygdala, which may be more susceptible to β-amyloid, tau, or α-synuclein deposits.242,243 Anatomical mapping studies have shown the amygdala is affected only later in the course of Lewy body spectrum diseases.244-246 Nevertheless, subtle changes in brainstem neuronal projections may be present early, leading to limbic circuit dysfunction.6,121,247-249 This hypothesis is supported by functional imaging studies performed in patients with prodromal PD.250,251
Regarding psychotic symptoms as part of manifest neurodegenerative diseases, major advances in the last decade consist in the clarification of genetic underpinnings, including the discovery of C9orf72 gene mutations in 2011, now recognized as the most common genetic abnormality associated with bvFTD and familial ALS.43,44 This is even more fascinating when juxtaposed with previous studies of bvFTD that did not find psychosis as part of FTD picture. 252 As detailed in this review, several studies of high quality revealed a heightened risk of psychosis for carriers of C9orf72 mutations with FTLD spectrum disorders, with some authors reporting increased frequency of delusions early in the course of disease in the FTLD-TDP subtype (often associated with C9orf72 mutations), compared to other pathological subgroups.18,43,44,59,80-83 Better characterization of biomarkers will guide the development of disease-specific diagnostic algorithms and treatment.155,171
There is little novelty regarding the management of psychosis of neurodegenerative diseases. No RCTs exist to evaluate nonpharmacological strategies; the lack of evidence may also be due to the fact that behavioral approaches focus on broader NPS, not just psychosis. 167 Also, psychosis is often associated with agitation, making it difficult to assess distinct study outcomes. 155 There have been only few RCTs conducted in the past decade to investigate the effectiveness of medications in the treatment of psychosis associated with major NCDs or neurodegenerative diseases, mirroring the dearth of available new psychotropic agents. The exception is pimavanserin, which was met with a great deal of enthusiasm due to its novel mechanism of action. The evidence for pimavanserin has been mixed to date and additional postmarketing data will be helpful to fully elucidate its safety for older adults. Furthermore, there is still very limited evidence pertaining to the management of neurobehavioral aspects of bvFTD and HD, despite the prevalence of severe psychiatric manifestations, including psychosis.
In summary, recent advances in the understanding of psychotic symptoms associated with neurodegenerative diseases center on psychiatric prodromes, including the MBI construct, and their significance as harbingers of full-blown neurobehavioral syndromes. Prompt prodrome recognition is crucial and will allow early intervention with disease-modifying treatments, hopefully in the near future. The symptomatic management of psychosis associated with neurodegenerative diseases, both in the prodromal and manifest stages, remains as much art as it is science, but stronger evidence is needed to support clinical decisions. The complexity of psychotic manifestations warrants management by interprofessional teams that provide coordinated care, integrating nonpharmacological and somatic treatment approaches.
Footnotes
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD
Andreea L. Seritan https://orcid.org/0000-0001-9287-9056
References
- 1.McGrath JJ, Saha S, Al-Hamzawi A, et al. Psychotic experiences in the general population: A cross-national analysis based on 31,261 respondents from 18 countries. JAMA Psychiatry. 2015;72:697-705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Meesters PD, de Haan L, Comijs HC, et al. Schizophrenia spectrum disorders in later life: Prevalence and distribution of age at onset and sex in a dutch catchment area. Am J Geriatr Psychiatry. 2012;20:18-28. [DOI] [PubMed] [Google Scholar]
- 3.González-Rodríguez A, Seeman MV, Izquierdo E, et al. Delusional disorder in old age: A hypothesis-driven review of recent work focusing on epidemiology, clinical aspects, and outcomes. Int J Environ Res Public Health. 2022;19:7911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Reinhardt MM, Cohen CI. Late-life psychosis: Diagnosis and treatment. Curr Psychiatry Rep. 2015;17:1. [DOI] [PubMed] [Google Scholar]
- 5.Block NR, Sha SJ, Karydas AM, et al. Frontotemporal dementia and psychiatric illness: Emerging clinical and biological links in gene carriers. Am J Geriatr Psychiatry. 2016;24;107-116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Seritan AL, Rienas C, Duong T, Delucchi K, Ostrem JL. Ages at onset of anxiety and depressive disorders in parkinson’s disease. J Neuropsychiatry Clin Neurosci. 2019;31:346-352. [DOI] [PubMed] [Google Scholar]
- 7.Fischer CE, Ismail Z, Youakim JM, et al. Revisiting criteria for psychosis in Alzheimer’s disease and related dementias: Toward better phenotypic classification and biomarker research. J Alzheimers Dis. 2020;73:1143-1156. [DOI] [PubMed] [Google Scholar]
- 8.Schneider LS, Tariot PN, Dagerman KS, et al. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med. 2006;355:1525-1538. [DOI] [PubMed] [Google Scholar]
- 9.Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group . Preferred reporting items for systematic reviews and meta analyses: The PRISMA statement. PLoS Med 2009;6:e1000097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.By the 2019 American Geriatrics Society Beers Criteria® Update Expert Panel . American geriatrics society 2019 updated AGS beers criteria® for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2019;67:674-694. [DOI] [PubMed] [Google Scholar]
- 11.López-Álvarez J, Sevilla-Llewellyn-Jones J, Agüera-Ortiz L. Anticholinergic drugs in geriatric psychopharmacology. Front Neurosci. 2019;13:1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Inouye SK, Westendorp RGJ, Saczynski JS. Delirium in elderly people. Lancet. 2014;383:911-922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Meagher DJ, Moran M, Raju B, et al. Phenomenology of delirium. Assessment of 100 adult cases using standardised measures. Br J Psychiatry. 2007;190:135–141. [DOI] [PubMed] [Google Scholar]
- 14.Hathaway E, Wang S. Use of the laboratory in the diagnostic workup of older adults. In: Steffens DC, Zdanys KF, eds. The American Psychiatric Association Publishing Textbook of Geriatric Psychiatry. 6th Ed. Washington, DC: American Psychiatric Association Publishing; 2023:95-118. [Google Scholar]
- 15.Titulaer MJ, McCracken L, Gabilondo I, et al. Late-onset anti-NMDA receptor encephalitis. Neurology. 2013;81:1058-1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Maguire M, Singh J, Marson A. Epilepsy and psychosis: A practical approach. Pract Neurol. 2018;18:106-114. [DOI] [PubMed] [Google Scholar]
- 17.Racine-Belkoura C, Geschwind M, eds. The multidisciplinary evaluation of the atypical dementia patient. In: Non-Alzheimer’s and Atypical Dementia. Oxford, UK: Wiley Publishing; 2016:6-16. [Google Scholar]
- 18.Elahi FM, Miller BL. A clinicopathological approach to the diagnosis of dementia. Nat Rev Neurol. 2017;13:457-476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Spar J, LaRue A, eds. Dementia and Alzheimer’s disease. In: Clinical Manual of Geriatric Psychiatry. Washington, DC: American Psychiatric Publishing, Inc; 2006:173-228. [Google Scholar]
- 20.Rascovsky K, Hodges JR, Knopman D, et al. Sensitivity of revised diagnostic criteria for the behavioural variant of frontotemporal dementia. Brain. 2011;134(Pt 9):2456-2477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bonelli RM, Cummings JL. Frontal-subcortical dementias. Neurologist. 2008;14:100-107. [DOI] [PubMed] [Google Scholar]
- 22.Aldridge GM, Birnschein A, Denburg NL, Narayanan NS. Parkinson’s disease dementia and dementia with lewy bodies have similar neuropsychological profiles. Front Neurol. 2018;9:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Seritan AL, Kim K, Benjamin I, Seritan I, Hagerman RJ. Risk factors for cognitive impairment in fragile X-associated tremor/ataxia syndrome. J Geriatr Psychiatry Neurol. 2016;29:328-337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Masse C, Vandel P, Sylvestre G, et al. Cognitive impairment in late-life depression: A comparative study of healthy older people, late-life depression, and mild Alzheimer’s disease using multivariate base rates of low scores. Front Psychol. 2021;12:724731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mendez MF, Cummings JL, eds. Psychiatric dementias associated with psychiatric disorders. In Dementia: A Clinical Approach. 3rd Ed. Philadelphia, PA: Butterworth Heinemann; 2003:477-502. [Google Scholar]
- 26.Blamire AM. MR approaches in neurodegenerative disorders. Prog Nucl Magn Reson Spectrosc. 2018;108:1-16. [DOI] [PubMed] [Google Scholar]
- 27.Ducharme S, Dols A, Laforce R, et al. Recommendations to distinguish behavioural variant frontotemporal dementia from psychiatric disorders. Brain. 2020;143:1632-1650. Erratum in: Brain. 2020;143:e62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Levenson JL. Psychiatric issues in endocrinology. Prim Psychiatry. 2006;13:27-30. [Google Scholar]
- 29.Hines A, Stewart JT, Catalano G. A case of capgras syndrome related to hypothyroidism. J Psychiatr Pract. 2015;21:445-448. [DOI] [PubMed] [Google Scholar]
- 30.Belvederi Murri M, Respino M, Masotti M, et al. Vitamin D and psychosis: Mini meta-analysis. Schizophr Res. 2013;150:235-239. [DOI] [PubMed] [Google Scholar]
- 31.Fogel BL, Perlman S. An approach to the patient with late-onset cerebellar ataxia. Nat Clin Pract Neurol. 2006;2:629-635. [DOI] [PubMed] [Google Scholar]
- 32.Mendez MF, Cummings JL, eds. Toxic-metabolic causes of dementia. In Dementia: A Clinical Approach. 3rd Ed. Philadelphia, PA: Butterworth Heinemann; 2003:421-476. [Google Scholar]
- 33.Parks KA, Parks CG, Onwuameze OE, Shrestha S. Psychiatric complications of primary hyperparathyroidism and mild hypercalcemia. Am J Psychiatry. 2017;174:620-622. [DOI] [PubMed] [Google Scholar]
- 34.Friedrich F, Aigner M, Fearns N, Friedrich ME, FRey R, GeusAu A. Psychosis in neurosyphilis -- clinical aspects and implications. Psychopathology. 2014;47:3-9. [DOI] [PubMed] [Google Scholar]
- 35.Rogers JP, Chesney E, Oliver D, et al. Psychiatric and neuropsychiatric presentations associated with severe coronavirus infections: A systematic review and meta-analysis with comparison to the COVID-19 pandemic. Lancet Psychiatry. 2020;7:611-627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bhat PS, Ryali V, Srivastava K, Kumar SR, Prakash J, Singal A. Alcoholic hallucinosis. Ind Psychiatry J. 2012;21:155-157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Isaac ML, Larson EB. Medical conditions with neuropsychiatric manifestations. Med Clin North Am. 2014;98:1193-1208. [DOI] [PubMed] [Google Scholar]
- 38.Bhattacharyya S, Darby RR, Raibagkar P, Gonzalez Castro LN, Berkowitz AL. Antibiotic-associated encephalopathy. Neurology. 2016; 86: 963-971. [DOI] [PubMed] [Google Scholar]
- 39.Borowoy AM, Pope JE, Silverman E, et al. Neuropsychiatric lupus: The prevalence and autoantibody associations depend on the definition: results from the 1000 faces of lupus cohort. Semin Arthritis Rheum. 2012;42:179-185. [DOI] [PubMed] [Google Scholar]
- 40.Drazyk AM, Tan RYY, Tay J, Traylor M, Das T, Markus HS. Encephalopathy in a large cohort of British cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy patients. Stroke. 2019; 50: 283-290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kaeley N, Bansal S, Bhatia R, Ahmad S. Herpes simplex encephalitis: An uncommon presentation. J Clin Diagn Res. 2016;10:OD25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pollak TA, Lennox BR, Müller S, et al. Autoimmune psychosis: An international consensus on an approach to the diagnosis and management of psychosis of suspected autoimmune origin. Lancet Psychiatry. 2020;7:93-108. [DOI] [PubMed] [Google Scholar]
- 43.Sha SJ, Takada LT, Rankin KP, et al. Frontotemporal dementia due to C9ORF72 mutations: Clinical and imaging features. Neurology. 2012;79:1002-1011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Snowden JS, Rollinson S, Thompson JC, et al. Distinct clinical and pathological characteristics of frontotemporal dementia associated with C9ORF72 mutations. Brain. 2012;135(Pt 3):693-708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hagerman RJ, Hagerman P. Fragile X-associated tremor/ataxia syndrome - features, mechanisms and management. Nat Rev Neurol. 2016;12:403-412. [DOI] [PubMed] [Google Scholar]
- 46.Kaufman DM, Geyer HL, Milstein MJ, eds. Diseases transmitted by chromosome or mitochondria abnormalities. In: Kaufman’s Clinical Neurology for Psychiatrists. 8th ed. Philadelphia, PA: Elsevier; 2017:540-541. [Google Scholar]
- 47.NIH National Library of Medicine . Medline Plus. Spinocerebellar ataxia type 1. National Institutes of Health. https://ghr.nlm.nih.gov/condition/spinocerebellar-ataxia-type-1. Published February 1, 2011. Accessed September 24, 2022. [Google Scholar]
- 48.NIH National Library of Medicine . Medline Plus. Spinocerebellar ataxia type 2. National Institutes of Health. https://ghr.nlm.nih.gov/condition/spinocerebellar-ataxia-type-2. Published December 1, 2019. Accessed September 24, 2022. [Google Scholar]
- 49.NIH National Library of Medicine . Medline Plus. Spinocerebellar ataxia type 3. National Institutes of Health. https://ghr.nlm.nih.gov/condition/spinocerebellar-ataxia-type-3. Published February 1, 2019. Accessed September 24, 2022. [Google Scholar]
- 50.Ross CA, Tabrizi SJ. Huntington’s disease: From molecular pathogenesis to clinical treatment. Lancet Neurol. 2011;10:83-98. [DOI] [PubMed] [Google Scholar]
- 51.Scarmeas N, Brandt J, Albert M, et al. Delusions and hallucinations are associated with worse outcome in Alzheimer disease. Arch Neurol. 2005;62:1601-1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Forsaa EB, Larsen JP, Wentzel-Larsen T, et al. A 12-year population-based study of psychosis in Parkinson disease. Arch Neurol. 2010;67:996-1001. [DOI] [PubMed] [Google Scholar]
- 53.Fischer CE, Agüera-Ortiz L. Psychosis and dementia: Risk factor, prodrome, or cause? Int Psychogeriatr. 2018;30:209-219. [DOI] [PubMed] [Google Scholar]
- 54.Connors MH, Teixeira-Pinto A, Loy CT. Psychosis and longitudinal outcomes in Huntington disease: The COHORT Study. J Neurol Neurosurg Psychiatry. 2020;91:15-20. [DOI] [PubMed] [Google Scholar]
- 55.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;172:460-465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Mason AR, Ziemann A, Finkbeiner S. Targeting the low-hanging fruit of neurodegeneration. Neurology. 2014;83:1470-1473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Jeste DV, Finkel SI. Psychosis of Alzheimer’s disease and related dementias. Diagnostic criteria for a distinct syndrome. Am J Geriatr Psychiatry. 2000;8:29-34. [DOI] [PubMed] [Google Scholar]
- 58.Apostolova LG, Di LJ, Duffy EL, et al. Risk factors for behavioral abnormalities in mild cognitive impairment and mild Alzheimer’s disease. Dement Geriatr Cogn Disord. 2014;37:315-326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kumfor F, Liang CT, Hazelton JL, et al. Examining the presence and nature of delusions in Alzheimer’s disease and frontotemporal dementia syndromes. Int J Geriatr Psychiatry. 2022;37(3). doi: 10.1002/gps.5692. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Reeves SJ, Gould RL, Powell JF, Howard RJ. Origins of delusions in Alzheimer’s disease. Neurosci Biobehav Rev. 2012;36:2274-2287. [DOI] [PubMed] [Google Scholar]
- 61.Perini G, Carlini A, Pomati S, et al. Misidentification delusions: Prevalence in different types of dementia and validation of a structured questionnaire. Alzheimer Dis Assoc Disord. 2016;30:331-337. [DOI] [PubMed] [Google Scholar]
- 62.Ruiz M, Arias A, Sánchez-Llanos E, et al. Minor hallucinations in Alzheimer’s disease. J Alzheimers Dis. 2018;64:543-549. [DOI] [PubMed] [Google Scholar]
- 63.Devanand DP, Lee S, Huey ED, Goldberg TE. Associations between neuropsychiatric symptoms and neuropathological diagnoses of Alzheimer disease and related dementias. JAMA Psychiatry. 2022;79:359-367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Fischer CE, Qian W, Schweizer TA, et al. Determining the impact of psychosis on rates of false-positive and false-negative diagnosis in Alzheimer’s disease. Alzheimers Dement 2017;3:385-392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lee AH, Weintraub D. Psychosis in Parkinson’s disease without dementia: Common and comorbid with other non-motor symptoms. Mov Disord. 2012;27:858-863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fénelon G, Alves G. Epidemiology of psychosis in Parkinson’s disease. J Neurol Sci. 2010;289:12-17. [DOI] [PubMed] [Google Scholar]
- 67.Kulick CV, Montgomery KM, Nirenberg MJ. Comprehensive identification of delusions and olfactory, tactile, gustatory, and minor hallucinations in Parkinson’s disease psychosis. Parkinsonism Relat Disord. 2018;54:40-45. [DOI] [PubMed] [Google Scholar]
- 68.Warren N, O'Gorman C, Hume Z, Kisely S, Siskind D. Delusions in Parkinson’s disease: A systematic review of published cases. Neuropsychol Rev. 2018;28:310-316. [DOI] [PubMed] [Google Scholar]
- 69.Wood RA, Hopkins SA, Moodley KK, Chan D. Fifty percent prevalence of extracampine hallucinations in Parkinson’s disease patients. Front Neurol. 2015;6:263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.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;89:88-100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chiba Y, Fujishiro H, Ota K, et al. Clinical profiles of dementia with Lewy bodies with and without Alzheimer’s disease-like hypometabolism. Int J Geriatr Psychiatry. 2015;30:316-323. [DOI] [PubMed] [Google Scholar]
- 72.Ochiai S, Sugawara H, Kajio Y, et al. Delusional parasitosis in dementia with Lewy bodies: A case report. Ann Gen Psychiatry. 2019;18:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Josephs KA. Capgras syndrome and its relationship to neurodegenerative disease. Arch Neurol. 2007;64:1762-1766. [DOI] [PubMed] [Google Scholar]
- 74.Naasan G, Shdo SM, Rodriguez EM, et al. Psychosis in neurodegenerative disease: Differential patterns of hallucination and delusion symptoms. Brain. 2021;144:999-1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tsunoda N, Hashimoto M, Ishikawa T, et al. Clinical features of auditory hallucinations in patients with dementia with lewy bodies: A soundtrack of visual hallucinations. J Clin Psychiatry. 2018;79:17m11623. [DOI] [PubMed] [Google Scholar]
- 76.Gorno-Tempini ML, Hillis AE, Weintraub S, et al. Classification of primary progressive aphasia and its variants. Neurology. 2011;76:1006-1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ramanan S, Irish M, Patterson K, Rowe JB, Gorno-Tempini ML, Lambon Ralph MA. Understanding the multidimensional cognitive deficits of logopenic variant primary progressive aphasia. Brain. 2022;145:2955-2966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Landqvist Waldö M, Gustafson L, Passant U, Englund E. Psychotic symptoms in frontotemporal dementia: A diagnostic dilemma? Int Psychogeriatr. 2015;27:531-539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gossink FT, Vijverberg EG, Krudop W, et al. Psychosis in behavioral variant frontotemporal dementia. Neuropsychiatr Dis Treat. 2017;13:1099-1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Devenney E, Hornberger M, Irish M, et al. Frontotemporal dementia associated with the C9ORF72 mutation: a unique clinical profile. JAMA Neurol. 2014;71:331-339. [DOI] [PubMed] [Google Scholar]
- 81.Devenney EM, Landin-Romero R, Irish M, et al. The neural correlates and clinical characteristics of psychosis in the frontotemporal dementia continuum and the C9orf72 expansion. Neuroimage Clin. 2017;13:439-445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Shinagawa S, Naasan G, Karydas AM, et al. Clinicopathological study of patients with C9ORF72-associated frontotemporal dementia presenting with delusions. J Geriatr Psychiatry Neurol. 2015;28:99-107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Benussi A, Premi E, Gazzina S, et al. Progression of behavioral disturbances and neuropsychiatric symptoms in patients with genetic frontotemporal dementia. JAMA Netw Open. 2021;4:e2030194. Erratum in: JAMA Netw Open. 2021;4:e217664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Fabbrini G, Fabbrini A, Suppa A. Progressive supranuclear palsy, multiple system atrophy and corticobasal degeneration. Handb Clin Neurol. 2019;165:155-177. [DOI] [PubMed] [Google Scholar]
- 85.Strong MJ, Abrahams S, Goldstein LH, et al. Amyotrophic lateral sclerosis - frontotemporal spectrum disorder (ALS-FTSD): Revised diagnostic criteria. Amyotroph Lateral Scler Frontotemporal Degener. 2017;18:153-174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tsai RM, Boxer AL. Therapy and clinical trials in frontotemporal dementia: Past, present, and future. J Neurochem. 2016;138(Suppl 1):211-221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lillo P, Mioshi E, Zoing MC, Kiernan MC, Hodges JR. How common are behavioural changes in amyotrophic lateral sclerosis? Amyotroph Lateral Scler. 2011;12:45-51. [DOI] [PubMed] [Google Scholar]
- 88.McHutchison CA, Leighton DJ, McIntosh A, et al. Relationship between neuropsychiatric disorders and cognitive and behavioural change in MND. J Neurol Neurosurg Psychiatry. 2020;91:245-253. [DOI] [PubMed] [Google Scholar]
- 89.Mioshi E, Caga J, Lillo P, et al. Neuropsychiatric changes precede classic motor symptoms in ALS and do not affect survival. Neurology. 2014;82:149-155. [DOI] [PubMed] [Google Scholar]
- 90.Zucchi E, Ticozzi N, Mandrioli J. Psychiatric symptoms in amyotrophic lateral sclerosis: Beyond a motor neuron disorder. Front Neurosci. 2019;13:175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Epping EA, Kim JI, Craufurd D, et al. Longitudinal psychiatric symptoms in prodromal Huntington’s disease: A decade of data. Am J Psychiatry. 2016;173:184–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Paulsen JS, Ready RE, Hamilton JM, Mega MS, Cummings JL. Neuropsychiatric aspects of Huntington’s disease. J Neurol Neurosurg Psychiatry. 2001;71:310-314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Paulsen JS, Miller AC, Hayes T, Shaw E. Cognitive and behavioral changes in Huntington disease before diagnosis. Handb Clin Neurol. 2017;144: 69–91. [DOI] [PubMed] [Google Scholar]
- 94.Reedeker W, van der Mast RC, Giltay EJ, Kooistra TAD, Roos RAC, van Duijn E. Psychiatric disorders in Huntington’s disease: A 2-year follow-up study. Psychosomatics. 2012;53:220-229. [DOI] [PubMed] [Google Scholar]
- 95.van Duijn E, Craufurd D, Hubers AAM, et al. Neuropsychiatric symptoms in a European huntington’s disease cohort (REGISTRY). J Neurol Neurosurg Psychiatry. 2014;85:1411-1418. [DOI] [PubMed] [Google Scholar]
- 96.Jaini A, Yomtoob J, Yeh C, Bega D. Understanding HD psychosis: An analysis from the ENROLL-HD database. Tremor Other Hyperkinet Mov (N Y). 2020;10:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Alpay M, Koroshetz WJ. Quetiapine in the treatment of behavioral disturbances in patients with Huntington’s disease. Psychosomatics. 2006;47:70-72. [DOI] [PubMed] [Google Scholar]
- 98.Shakkottai VG, Fogel BL. Clinical neurogenetics: Autosomal dominant spinocerebellar ataxia. Neurol Clin. 2013;31:987-1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Stevanin G, Brice A. Spinocerebellar ataxia 17 (SCA17) and Huntington’s disease-like 4 (HDL4). Cerebellum. 2008;7(2):170-178. [DOI] [PubMed] [Google Scholar]
- 100.Liszewski CM, O’Hearn E, Leroi I, Gourley L, Ross CA, Margolis RL. Cognitive impairment and psychiatric symptoms in 133 patients with diseases associated with cerebellar degeneration. J Neuropsychiatry Clin Neurosci. 2004;16:109-112. [DOI] [PubMed] [Google Scholar]
- 101.Rottnek M, Riggio S, Byne W, Sano M, Margolis RL, Walker RH. Schizophrenia in a patient with spinocerebellar ataxia 2: Coincidence of two disorders or a neurodegenerative disease presenting with psychosis? Am J Psychiatry. 2008;165:964-967. [DOI] [PubMed] [Google Scholar]
- 102.Wexler E, Fogel BL. New-onset psychosis in a patient with spinocerebellar ataxia type 10. Am J Psychiatry. 2011;168:1339-1340. [DOI] [PubMed] [Google Scholar]
- 103.Braga-Neto P, Pedroso JL, Gadelha A, et al. Psychosis in machado-joseph disease: Clinical correlates, pathophysiological discussion, and functional brain imaging. Expanding the cerebellar cognitive affective syndrome. Cerebellum. 2016;15:483-490. [DOI] [PubMed] [Google Scholar]
- 104.Nolte D, Klebe S, Baron R, Deuschl G, Muller U. Codon 101 of PRKCG, a preferential mutation site in SCA14. Mov Disord. 2007;22:1831-1832. [DOI] [PubMed] [Google Scholar]
- 105.Turk KW, Flanagan ME, Josephson S, Keene CD, Jayadev S, Bird TD. Psychosis in spinocerebellar ataxias: A case series and study of tyrosine hydroxylase in substantia Nigra. Cerebellum. 2018;17:143-151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Okamoto N, Ikenouchi A, Ide S, Hashimoto Y, Yoshimura R. A young Japanese patient with spinocerebellar ataxia type 3 presenting depressive state with cenesthopathy and delusion: A case report. Cerebellum. 2022;21:1135-1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Jacquemont S, Farzin F, Hall D, et al. Aging in individuals with the FMR1 mutation. Am J Ment Retard. 2004;109:154-164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Jacquemont S, Hagerman RJ, Leehey MA, et al. Penetrance of the fragile X-associated tremor/ataxia syndrome in a premutation carrier population. JAMA. 2004;291:460-469. [DOI] [PubMed] [Google Scholar]
- 109.Bacalman S, Farzin F, Bourgeois JA, et al. Psychiatric phenotype of the fragile X-associated tremor/ataxia syndrome (FXTAS) in males: Newly described fronto-subcortical dementia. J Clin Psychiatry. 2006;67:87-94. [DOI] [PubMed] [Google Scholar]
- 110.Bourgeois JA, Seritan AL, Casillas EM, et al. Lifetime prevalence of mood and anxiety disorders in fragile X premutation carriers. J Clin Psychiatry. 2011;72:175-182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Flavell J, Franklin C, Nestor PJ. A systematic review of fragile X-associated neuropsychiatric disorders. J Neuropsychiatry Clin Neurosci. 2022;appineuropsych21110282. doi: 10.1176/appi.neuropsych.21110282 [DOI] [PubMed] [Google Scholar]
- 112.Seritan AL, Ortigas M, Seritan S, Bourgeois JA, Hagerman RJ. Psychiatric disorders associated with FXTAS. Curr Psychiatry Rev. 2013;9:59-64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kurz MW, Schlitter AM, Klenk Y, et al. FMR1 alleles in Parkinson’s disease: Relation to cognitive decline and hallucinations, a longitudinal study. J Geriatr Psychiatry Neurol. 2007;20:89-92. [DOI] [PubMed] [Google Scholar]
- 114.Salcedo-Arellano MJ, Sanchez D, Wang JY, et al. Case report: Coexistence of Alzheimer-type neuropathology in fragile X-associated tremor ataxia syndrome. Front Neurosci. 2021;15:720253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Schrag A, Schott JM. Epidemiological, clinical, and genetic characteristics of early-onset parkinsonism. Lancet Neurol. 2006;5:355-363. [DOI] [PubMed] [Google Scholar]
- 116.Seritan AL, Cogswell J, Grigsby J. Cognitive dysfunction in FMR1 premutation carriers. Curr Psychiatry Rev. 2013;9:78-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.NIH National Center for Advancing Translational Sciences . Juvenile Huntington Disease. juvenile/Huntington/disease-about/the/Disease-Genetic/and/Rare/Diseases/Information/Center/(nih.gov). Published November 8, 2021. Accessed October 2, 2022. [Google Scholar]
- 118.Postuma RB, Aarsland D, Barone P, et al. Identifying prodromal Parkinson’s disease: Pre-motor disorders in Parkinson’s disease. Mov Disord. 2012;27:617–626. [DOI] [PubMed] [Google Scholar]
- 119.Durcan R, Wiblin L, Lawson RA, et al. Prevalence and duration of non-motor symptoms in prodromal Parkinson’s disease. Eur J Neurol. 2019;26:979-985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Berg D, Postuma RB, Adler CH, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30:1600-1611. [DOI] [PubMed] [Google Scholar]
- 121.Iranzo A, Gelpi E, Tolosa E, et al. Neuropathology of prodromal Lewy body disease. Mov Disord. 2014;29:410-415. [DOI] [PubMed] [Google Scholar]
- 122.Iranzo A, Fernández-Arcos A, Tolosa E, et al. Neurodegenerative disorder risk in idiopathic REM sleep behavior disorder: Study in 174 patients. PLoS One. 2014;9: e89741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Mahlknecht P, Seppi K, Poewe W. The concept of prodromal Parkinson’s disease. J Parkinsons Dis. 2015;5:681–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.McKeith IG, Ferman TJ, Thomas AJ, et al. Research criteria for the diagnosis of prodromal dementia with Lewy bodies. Neurology. 2020;94:743-755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Burke SL, Maramaldi P, Cadet T, Kukull W. Neuropsychiatric symptoms and apolipoprotein E: Associations with eventual Alzheimer’s disease development. Arch Gerontol Geriatr. 2016;65:231-238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Sadiq D, Whitfield T, Lee L, Stevens T, Costafreda S, Walker Z. Prodromal dementia with lewy bodies and prodromal Alzheimer’s disease: A comparison of the cognitive and clinical profiles. J Alzheimers Dis. 2017;58:463-470. [DOI] [PubMed] [Google Scholar]
- 127.Segers K, Benoit F, Meyts JM, Surquin M. Anxiety symptoms are quantitatively and qualitatively different in dementia with Lewy bodies than in Alzheimer’s disease in the years preceding clinical diagnosis. Psychogeriatrics. 2020;20:242-246. [DOI] [PubMed] [Google Scholar]
- 128.Grande G, Vetrano DL, Mazzoleni F, et al. Detection and prediction of incident Alzheimer dementia over a 10-year or longer medical history: a population-based study in primary care. Dement Geriatr Cogn Disord. 2020;49:384-389. [DOI] [PubMed] [Google Scholar]
- 129.Gallagher D, Coen R, Kilroy D, et al. Anxiety and behavioural disturbance as markers of prodromal Alzheimer’s disease in patients with mild cognitive impairment. Int J Geriatr Psychiatry. 2011;26:166-172. [DOI] [PubMed] [Google Scholar]
- 130.Lee GJ, Lu PH, Hua X, et al. Depressive symptoms in mild cognitive impairment predict greater atrophy in Alzheimer’s disease-related regions. Biol Psychiatry. 2012;71:814-821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Vellone D, Ghahremani M, Goodarzi Z, Forkert ND, Smith EE, Ismail Z. Apathy and APOE in mild behavioral impairment, and risk for incident dementia. Alzheimers Dement 2022;8:e12370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wise EA, Rosenberg PB, Lyketsos CG, Leoutsakos JM. Time course of neuropsychiatric symptoms and cognitive diagnosis in National Alzheimer’s coordinating centers volunteers. Alzheimers Dement (Amst). 2019;11:333-339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Lillo P, Garcin B, Hornberger M, Bak TH, Hodges JR. Neurobehavioral features in frontotemporal dementia with amyotrophic lateral sclerosis. Arch Neurol. 2010;67:826-830. [DOI] [PubMed] [Google Scholar]
- 134.Turner MR, Goldacre R, Talbot K, Goldacre MJ. Psychiatric disorders prior to amyotrophic lateral sclerosis. Ann Neurol. 2016;80:935-938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Longinetti E, Mariosa D, Larsson H, et al. Neurodegenerative and psychiatric diseases among families with amyotrophic lateral sclerosis. Neurology. 2017;89:578-585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Woolley JD, Khan BK, Murthy NK, Miller BL, Rankin KP. The diagnostic challenge of psychiatric symptoms in neurodegenerative disease: Rates of and risk factors for prior psychiatric diagnosis in patients with early neurodegenerative disease. J Clin Psychiatry. 2011;72:126-133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Gambogi LB, Guimarães HC, de Souza LC, Caramelli P. Long-term severe mental disorders preceding behavioral variant fronto-temporal dementia: Frequency and clinical correlates in an outpatient sample. J Alzheimers Dis. 2018;66:1577-1585. [DOI] [PubMed] [Google Scholar]
- 138.Sellami L, St-Onge F, Poulin S, Laforce R, Jr. Schizophrenia phenotype preceding behavioral variant frontotemporal dementia related to C9orf72 repeat expansion. Cogn Behav Neurol. 2019;32:120-123. [DOI] [PubMed] [Google Scholar]
- 139.Taragano FE, Allegri RF, Krupitzki H, et al. Mild behavioral impairment and risk of dementia: A prospective cohort study of 358 patients. J Clin Psychiatry. 2009;70:584-592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Blanc F, Bouteloup V, Paquet C, et al. Prodromal characteristics of dementia with Lewy bodies: Baseline results of the MEMENTO memory clinics nationwide cohort. Alzheimers Res Ther. 2022;14:96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Jaramillo-Jimenez A, Ying Y, Ren P, et al. Prodromal dementia with lewy bodies and recurrent panic attacks as the first symptom: A case report. Front Neurol. 2022;13:839539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Urso D, Gnoni V, De Blasi R, et al. Neuroimaging biomarkers in a patient with probable psychiatric-onset prodromal dementia with lewy bodies. Neurology. 2022;99:654-657. doi: 10.1212/WNL.0000000000201166. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 143.Wyman-Chick KA, O'Keefe LR, Weintraub D, et al. Prodromal dementia with lewy bodies: Evolution of symptoms and predictors of dementia onset. J Geriatr Psychiatry Neurol. 2022;35:527-534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Kobayashi K, Nakano H, Akiyama N, Maeda T, Yamamori S. Pure psychiatric presentation of the Lewy body disease is depression--an analysis of 60 cases verified with myocardial meta-iodobenzylguanidine study. Int J Geriatr Psychiatry. 2015;30:663-668. [DOI] [PubMed] [Google Scholar]
- 145.Utsumi K, Fukatsu R, Hara Y, Takamaru Y, Yasumura S. Psychotic features among patients in the prodromal stage of dementia with lewy bodies during longitudinal observation. J Alzheimers Dis. 2021;83:1917-1927. [DOI] [PubMed] [Google Scholar]
- 146.Seritan AL, Bourgeois JA, Schneider A, Mu Y, Hagerman RJ, Nguyen DV. Ages of onset of mood and anxiety disorders in fragile X premutation carriers. Curr Psychiatry Rev. 2013;9:65–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Epping EA, Mills JA, Beglinger LJ, et al. Characterization of depression in prodromal Huntington disease in the neurobiological predictors of HD (PREDICT-HD) study. J Psychiatr Res. 2013;47:1423-1431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Orth M, Handley OJ, Schwenke C, et al. Observing Huntington’s disease: The European huntington’s disease network’s REGISTRY. PLoS Curr. 2010;2:RRN1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Pedroso JL, Bor-Seng-Shu E, Braga-Neto P, et al. Neurophysiological studies and non-motor symptoms prior to ataxia in a patient with Machado-Joseph disease: Trying to understand the natural history of brain degeneration. Cerebellum. 2014;13:447-451. [DOI] [PubMed] [Google Scholar]
- 150.Ismail Z, Smith EE, Geda Y, et al. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement. 2016;12:195-202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Matsuoka T, Ismail Z, Narumoto J. Prevalence of mild behavioral impairment and risk of dementia in a psychiatric outpatient clinic. J Alzheimers Dis. 2019;70:505-513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Mallo SC, Ismail Z, Pereiro AX, et al. Assessing mild behavioral impairment with the mild behavioral impairment-checklist in people with mild cognitive impairment. J Alzheimers Dis. 2018;66:83-95. [DOI] [PubMed] [Google Scholar]
- 153.Mortby ME, Ismail Z, Anstey KJ. Prevalence estimates of mild behavioral impairment in a population-based sample of pre-dementia states and cognitively healthy older adults. Int Psychogeriatr. 2018;30:221-232. [DOI] [PubMed] [Google Scholar]
- 154.Wolfova K, Creese B, Aarsland D, et al. Gender/sex differences in the association of mild behavioral impairment with cognitive aging. J Alzheimers Dis. 2022;88:345-355. [DOI] [PubMed] [Google Scholar]
- 155.Ismail Z, Creese B, Aarsland D, et al. Psychosis in Alzheimer disease - mechanisms, genetics and therapeutic opportunities. Nat Rev Neurol. 2022;18:131-144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Yokoi Y, Takano H, Sakata M, Maruo K, Nakagome K, Matsuda H. Discrete effect of each mild behavioural impairment category on dementia conversion or cognitive decline in patients with mild cognitive impairment. Psychogeriatrics. 2019;19:591-600. [DOI] [PubMed] [Google Scholar]
- 157.Pagonabarraga J, Martinez-Horta S, Fernández de Bobadilla R, et al. Minor hallucinations occur in drug-naive Parkinson’s disease patients, even from the premotor phase. Mov Disord. 2016;31:45-52. Erratum in: Mov Disord. 2016 Mar;31(3):433. [DOI] [PubMed] [Google Scholar]
- 158.Pachi I, Maraki MI, Giagkou N, et al. Late life psychotic features in prodromal Parkinson’s disease. Parkinsonism Relat Disord. 2021;86:67-73. [DOI] [PubMed] [Google Scholar]
- 159.Ravina B, Marder K, Fernandez HH, et al. Diagnostic criteria for psychosis in Parkinson’s disease: Report of an NINDS, NIMH work group. Mov Disord. 2007;22:1061-1068. [DOI] [PubMed] [Google Scholar]
- 160.Cummings J, Pinto LC, Cruz M, et al. Criteria for psychosis in major and mild neurocognitive disorders: International psychogeriatric association (IPA) consensus clinical and research definition. Am J Geriatr Psychiatry. 2020;28:1256-1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Lyketsos CG, Breitner JC, Rabins PV. An evidence-based proposal for the classification of neuropsychiatric disturbance in Alzheimer’s disease. Int J Geriatr Psychiatry. 2001;16:1037-1042. [DOI] [PubMed] [Google Scholar]
- 162.Höglinger GU, Respondek G, Stamelou M, et al. Clinical diagnosis of progressive supranuclear palsy: The movement disorder society criteria. Mov Disord. 2017;32:853-864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.McKeith I, Taylor JP, Thomas A, Donaghy P, Kane J. Revisiting DLB diagnosis: A consideration of prodromal DLB and of the diagnostic overlap with Alzheimer disease. J Geriatr Psychiatry Neurol. 2016;29:249-253. [DOI] [PubMed] [Google Scholar]
- 164.Ismail Z, Agüera-Ortiz L, Brodaty H, et al. The mild behavioral impairment checklist (MBI-C): A rating scale for neuropsychiatric symptoms in pre-dementia populations. J Alzheimers Dis. 2017;56(3):929-938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Hu S, Patten S, Charlton A, et al. Validating the mild behavioral impairment checklist in a cognitive clinic: Comparisons with the neuropsychiatric inventory questionnaire. J Geriatr Psychiatry Neurol. 2022;36:107-120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Reus VI, Fochtmann LJ, Eyler AE, et al. The American psychiatric association practice guideline on the use of antipsychotics to treat agitation or psychosis in patients with dementia. Am J Psychiatry. 2016;173:543-546. [DOI] [PubMed] [Google Scholar]
- 167.Kales HC, Gitlin LN, Lyketsos CG; , Detroit expert panel on assessment and management of neuropsychiatric symptoms of dementia. Management of neuropsychiatric symptoms of dementia in clinical settings: recommendations from a multidisciplinary expert panel. J Am Geriatr Soc. 2014;62:762-769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: An international Delphi consensus. Int Psychogeriatr. 2019;31:83-90. [DOI] [PubMed] [Google Scholar]
- 169.Diederich NJ, Pieri V, Goetz CG. Coping strategies for visual hallucinations in Parkinson’s disease. Mov Disord. 2003;18:831-832. [DOI] [PubMed] [Google Scholar]
- 170.Agüera-Ortiz L, Babulal GM, Bruneau MA, et al. Psychosis as a treatment target in dementia: A roadmap for designing interventions. J Alzheimers Dis. 2022;88:1203-1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Ballard C, Kales HC, Lyketsos C, et al. Psychosis in Alzheimer’s disease. Curr Neurol Neurosci Rep. 2020;20:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Pahwa R, Isaacson SH, Small GW, Torres-Yaghi Y, Pagan F, Sabbagh M. Screening, diagnosis, and management of parkinson’s disease psychosis: Recommendations from an expert panel. Neurol Ther. 2022;11:1571-1582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Yunusa I, Alsumali A, Garba AE, Regestein QR, Eguale T. Assessment of reported comparative effectiveness and safety of atypical antipsychotics in the treatment of behavioral and psychological symptoms of dementia: A network meta-analysis. JAMA Netw Open. 2019;2:e190828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Maust DT, Kim HM, Seyfried LS, et al. Antipsychotics, other psychotropics, and the risk of death in patients with dementia: Number needed to harm. JAMA Psychiatry. 2015;72:438-445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Sultzer DL, Davis SM, Tariot PN, et al. Clinical symptom responses to atypical antipsychotic medications in Alzheimer’s disease: Phase 1 outcomes from the CATIE-AD effectiveness trial. Am J Psychiatry. 2008;165:844-854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Sink KM, Holden KF, Yaffe K. Pharmacological treatment of neuropsychiatric symptoms of dementia: A review of the evidence. JAMA. 2005;293:596-608. [DOI] [PubMed] [Google Scholar]
- 177.Mintzer JE, Tune LE, Breder CD, et al. Aripiprazole for the treatment of psychoses in institutionalized patients with Alzheimer dementia: A multicenter, randomized, double-blind, placebo-controlled assessment of three fixed doses. Am J Geriatr Psychiatry. 2007;15:918-931. [DOI] [PubMed] [Google Scholar]
- 178.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;17:213-222. [DOI] [PubMed] [Google Scholar]
- 179.Tariot PN, Cummings JL, Soto-Martin ME, et al. Trial of pimavanserin in dementia-related psychosis. N Engl J Med. 2021;385:309-319. [DOI] [PubMed] [Google Scholar]
- 180.Porsteinsson AP, Drye LT, Pollock BG, et al. Effect of citalopram on agitation in Alzheimer disease: The CitAD randomized clinical trial. JAMA. 2014;311:682-691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Leonpacher AK, Peters ME, Drye LT, et al. Effects of citalopram on neuropsychiatric symptoms in Alzheimer’s dementia: Evidence from the CitAD study. Am J Psychiatry. 2016;173:473-480. [DOI] [PubMed] [Google Scholar]
- 182.U.S. Food and Drug Administration . FDA/Drug/Safety/Communication:Revised/recommendations/for/Celexa/(citalopram/hydrobromide)/related/to/a/potential/risk/of/abnormal/heart/rhythms/with/high/doses/FDA. December 15, 2017. Accessed October 22, 2022.
- 183.Devanand DP, Crocco E, Forester BP, et al. Low dose lithium treatment of behavioral complications in Alzheimer’s disease: Lit-AD randomized clinical trial. Am J Geriatr Psychiatry. 2022;30:32-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Tan ECK, Johnell K, Bell JS, et al. Do acetylcholinesterase inhibitors prevent or delay psychotropic prescribing in people with dementia? Analyses of the Swedish dementia registry. Am J Geriatr Psychiatry. 2020;28:108-117. [DOI] [PubMed] [Google Scholar]
- 185.Zahodne LB, Fernandez HH. Parkinson’s psychosis. Curr Treat Options Neurol. 2010;12:200-211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Zhang H, Wang L, Fan Y, et al. Atypical antipsychotics for Parkinson’s disease psychosis: A systematic review and meta-analysis. Neuropsychiatr Dis Treat. 2019;15:2137-2149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.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;383:533-540. [DOI] [PubMed] [Google Scholar]
- 188.Isaacson SH, Ballard CG, Kreitzman DL, et al. Efficacy results of pimavanserin from a multi-center, open-label extension study in Parkinson’s disease psychosis patients. Parkinsonism Relat Disord. 2021;87:25-31. [DOI] [PubMed] [Google Scholar]
- 189.U.S. Food and Drug Administration . FDA/analysis/finds/no/new/or/unexpected/safety/risks/associated/with/Nuplazid/(pimavanserin)/a/medication/to/treat/the/hallucinations/and/delusions/of/Parkinson’s/disease/psychosis/FDA. September 20, 2018. Accessed October 27, 2022.
- 190.Hwang YJ, Alexander GC, An H, Moore TJ, Mehta HB. Risk of hospitalization and death associated with pimavanserin use in older adults with Parkinson disease. Neurology. 2021;97:e1266-e1275. [DOI] [PubMed] [Google Scholar]
- 191.Weintraub D, Chen P, Ignacio RV, Mamikonyan E, Kales HC. Patterns and trends in antipsychotic prescribing for Parkinson disease psychosis. Arch Neurol. 2011;68:899-904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Desmarais P, Massoud F, Filion J, Nguyen QD, Bajsarowicz P. Quetiapine for psychosis in Parkinson disease and neurodegenerative Parkinsonian disorders: A systematic review. J Geriatr Psychiatry Neurol. 2016;29(4):227-236. [DOI] [PubMed] [Google Scholar]
- 193.Horn S, Richardson H, Xie SX, Weintraub D, Dahodwala N. Pimavanserin versus quetiapine for the treatment of psychosis in Parkinson’s disease and dementia with Lewy bodies. Parkinsonism Relat Disord. 2019;69:119-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Younce JR, Davis AA, Black KJ. A systematic review and case series of ziprasidone for psychosis in Parkinson’s disease. J Parkinsons Dis. 2019;9:63-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Pintor L, Valldeoriola F, Baillés E, Marti MJ, Muniz A, Tolosa E. Ziprasidone versus clozapine in the treatment of psychotic symptoms in Parkinson disease: A randomized open clinical trial. Clin Neuropharmacol. 2012;35:61–66. [DOI] [PubMed] [Google Scholar]
- 196.Kashihara K, Maeda T, Yoshida K. Safety and tolerability of aripiprazole in patients with psychosis associated with Parkinson’s disease-results of a multicenter open trial. Neuropsychopharmacol Rep. 2022;42:135-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Rolinski M, Fox C, Maidment I, McShane R. Cholinesterase inhibitors for dementia with Lewy bodies, Parkinson’s disease dementia and cognitive impairment in Parkinson’s disease. Cochrane Database Syst Rev. 2012;2012(3):CD006504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Seritan AL, Ureste PJ, Duong T, Ostrem JL. Psychopharmacology for patients with Parkinson’s disease and deep brain stimulation: Lessons learned in an academic center. Curr Psychopharmacol. 2019;8:41-54. [Google Scholar]
- 199.van Mierlo TJM, Foncke EMJ, Post B, et al. Rivastigmine for minor visual hallucinations in Parkinson’s disease: A randomized controlled trial with 24 months follow-up. Brain Behav. 2021;11:e2257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Godschalx-Dekker JA, Siegers HP. Reduction of parkinsonism and psychosis with mirtazapine: A case report. Pharmacopsychiatry. 2014;47:81-83. [DOI] [PubMed] [Google Scholar]
- 201.Bergman J, Lerner PP, Sokolik S, Lerner V, Kreinin A, Miodownik C. Successful use of escitalopram for the treatment of visual hallucinations in patients with Parkinson disease. Clin Neuropharmacol. 2017;40:246-250. [DOI] [PubMed] [Google Scholar]
- 202.Tagai K, Nagata T, Shinagawa S, Tsuno N, Ozone M, Nakayama K. Mirtazapine improves visual hallucinations in Parkinson’s disease: A case report. Psychogeriatrics. 2013;13:103-107. [DOI] [PubMed] [Google Scholar]
- 203.Sid-Otmane L, Huot P, Panisset M. Effect of antidepressants on psychotic symptoms in Parkinson disease: A review of case reports and case series. Clin Neuropharmacol. 2020;43:61-65. [DOI] [PubMed] [Google Scholar]
- 204.Chatham AN, Shafi H, Hermida AP. The use of ECT in the elderly-looking beyond depression. Curr Psychiatry Rep. 2022;24:451-461. [DOI] [PubMed] [Google Scholar]
- 205.Takamiya A, Seki M, Kudo S, et al. Electroconvulsive therapy for Parkinson’s disease: A systematic review and meta-analysis. Mov Disord. 2021;36:50-58. [DOI] [PubMed] [Google Scholar]
- 206.Kyle K, Bronstein JM. Treatment of psychosis in Parkinson’s disease and dementia with Lewy Bodies: A review. Parkinsonism Relat Disord. 2020;75:55-62. [DOI] [PubMed] [Google Scholar]
- 207.Stinton C, McKeith I, Taylor JP, et al. Pharmacological management of Lewy Body dementia: A systematic review and meta-analysis. Am J Psychiatry. 2015;172:731-742. [DOI] [PubMed] [Google Scholar]
- 208.Badwal K, Kiliaki SA, Dugani SB, Pagali SR. Psychosis management in Lewy Body Dementia: A comprehensive clinical approach. J Geriatr Psychiatry Neurol. 2022;35:255-261. [DOI] [PubMed] [Google Scholar]
- 209.Mori E, Ikeda M, Kosaka K, Donepezil-DLB Study Investigators . Donepezil for dementia with Lewy bodies: A randomized, placebo-controlled trial. Ann Neurol. 2012;72:41-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Ikeda M, Mori E, Kosaka K, et al. Long-term safety and efficacy of donepezil in patients with dementia with Lewy bodies: Results from a 52-week, open-label, multicenter extension study. Dement Geriatr Cogn Disord. 2013;36:229-241. [DOI] [PubMed] [Google Scholar]
- 211.Mori E, Ikeda M, Nagai R, Matsuo K, Nakagawa M, Kosaka K. Long-term donepezil use for dementia with Lewy bodies: Results from an open-label extension of Phase III trial. Alzheimers Res Ther. 2015;7:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Ukai K, Fujishiro H, Iritani S, Ozaki N. Long-term efficacy of donepezil for relapse of visual hallucinations in patients with dementia with Lewy bodies. Psychogeriatrics. 2015;15:133-137. [DOI] [PubMed] [Google Scholar]
- 213.Fukatsu T, Kanemoto K. Electroconvulsive therapy improves psychotic symptoms in patients with dementia with Lewy bodies. Int J Geriatr Psychiatry 2022;37(11). doi: 10.1002/gps.5832. [DOI] [PubMed] [Google Scholar]
- 214.Murley AG, Rowe JB. Neurotransmitter deficits from frontotemporal lobar degeneration. Brain. 2018;141:1263-1285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Magrath Guimet N, Zapata-Restrepo LM, Miller BL. Advances in treatment of frontotemporal dementia. J Neuropsychiatry Clin Neurosci. 2022;34:316-327. [DOI] [PubMed] [Google Scholar]
- 216.Ljubenkov PA, Boxer AL. FTLD treatment: Current practice and future possibilities. Adv Exp Med Biol. 2021;1281:297-310. [DOI] [PubMed] [Google Scholar]
- 217.Curtis RC, Resch DS. Case of pick’s central lobar atrophy with apparent stabilization of cognitive decline after treatment with risperidone. J Clin Psychopharmacol. 2000;20:384-385. [DOI] [PubMed] [Google Scholar]
- 218.Moretti R, Torre P, Antonello RM, Cazzato G, Griggio S, Bava A. Olanzapine as a treatment of neuropsychiatric disorders of Alzheimer’s disease and other dementias: A 24-month follow-up of 68 patients. Am J Alzheimers Dis Other Demen. 2003;18:205-214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Jha MK, Lambert ES, Beadles BA, et al. A case of frontotemporal dementia presenting with treatment-refractory psychosis and extreme violence: Response to combination of clozapine, medroxyprogesterone, and sertraline. J Clin Psychopharmacol. 2015;35:732-733. [DOI] [PubMed] [Google Scholar]
- 220.Bachoud-Lévi AC, Ferreira J, Massart R, et al. International guidelines for the treatment of huntington’s disease. Front Neurol. 2019;10:710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Oulis P, Mourikis I, Konstantakopoulos G, Papageorgiou SG, Kouzoupis AV. Aripiprazole in the treatment of olanzapine-resistant psychotic and motor symptoms of Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2010;22(3):352. [DOI] [PubMed] [Google Scholar]
- 222.Yavuz KF, Ulusoy S, Alnıak İ. Aripiprazole treatment for choreoathetoid and psychotic symptoms of Huntington’s disease. J Neuropsychiatry Clin Neurosci. 2013;25:E31. [DOI] [PubMed] [Google Scholar]
- 223.van Duijn E. Medical treatment of behavioral manifestations of Huntington disease. Handb Clin Neurol. 2017;144:129-139. [DOI] [PubMed] [Google Scholar]
- 224.Danivas V, Moily NS, Thimmaiah R, et al. Off label use of lithium in the treatment of Huntington’s disease: A case series. Indian J Psychiatry. 2013;55:81-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Sharma M, Aggarwal S, Ragavi N, Kumar M. Management of neuropsychiatric symptoms in Huntington’s disease (HD) with clozapine: A case report. Asian J Psychiatr. 2020;54:102345. [DOI] [PubMed] [Google Scholar]
- 226.Bampton TJ, Hack D, Galletly CA. Clozapine treatment for Huntington’s disease psychosis. Aust N Z J Psychiatry. 2022;56:200. [DOI] [PubMed] [Google Scholar]
- 227.Edlinger M, Seppi K, Fleischhacker W, Hofer A. Treatment of psychotic and behavioral symptoms with clozapine, aripiprazole, and reboxetine in a patient with Huntington’s disease. Int Clin Psychopharmacol. 2013;28:214-216. [DOI] [PubMed] [Google Scholar]
- 228.Hagerman RJ, Hall DA, Coffey S, et al. Treatment of fragile X-associated tremor ataxia syndrome (FXTAS) and related neurological problems. Clin Interv Aging. 2008;3:251-262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Gomide L, Kummer A, Cardoso F, Teixeira AL. Use of clozapine in Brazilian patients with Parkinson’s disease. Arq Neuropsiquiatr. 2008;66(3B):611-614. [DOI] [PubMed] [Google Scholar]
- 230.Kurlan R, Cummings J, Raman R, Thal L, Alzheimer's Disease Cooperative Study Group . Quetiapine for agitation or psychosis in patients with dementia and parkinsonism. Neurology. 2007;68:1356-1363. [DOI] [PubMed] [Google Scholar]
- 231.Geda YE, Roberts RO, Mielke MM, et al. Baseline neuropsychiatric symptoms and the risk of incident mild cognitive impairment: A population-based study. Am J Psychiatry. 2014;171:572-581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Belbeze J, Gallarda T. Very late onset psychotic symptoms: psychosis or dementia? A phenomenological approach. A systematic review. Geriatr Psychol Neuropsychiatr Vieil. 2020;18:77-86. [DOI] [PubMed] [Google Scholar]
- 233.Peters ME, Rosenberg PB, Steinberg M, et al. Neuropsychiatric symptoms as risk factors for progression from CIND to dementia: The cache county study. Am J Geriatr Psychiatry. 2013;21:1116-1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Richard E, Schmand B, Eikelenboom P, et al. Symptoms of apathy are associated with progression from mild cognitive impairment to Alzheimer’s disease in non-depressed subjects. Dement Geriatr Cogn Disord. 2012;33:204-209. [DOI] [PubMed] [Google Scholar]
- 235.Richard E, Reitz C, Honig LH, et al. Late-life depression, mild cognitive impairment, and dementia. JAMA Neurol. 2013;70:374-382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Leoutsakos JMS, Forrester SN, Lyketsos CG, Smith GS. Latent classes of neuropsychiatric symptoms in NACC controls and conversion to mild cognitive impairment or dementia. J Alzheimers Dis. 2015;48:483-493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Ronat L, Hoang VT, Hanganu A. Establishing an individualized model of conversion from normal cognition to Alzheimer’s disease after 4 years, based on cognitive, brain morphology and neuropsychiatric characteristics. Int J Geriatr Psychiatry. 2022;37. doi: 10.1002/gps.5718. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
- 238.da Silva J, Gonçalves-Pereira M, Xavier M, Mukaetova-Ladinska EB. Affective disorders and risk of developing dementia: Systematic review. Br J Psychiatry. 2013;202:177-186. [DOI] [PubMed] [Google Scholar]
- 239.Stafford J, Chung WT, Sommerlad A, Kirkbride JB, Howard R. Psychiatric disorders and risk of subsequent dementia: Systematic review and meta-analysis of longitudinal studies. Int J Geriatr Psychiatry. 2022;37:5711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.BMJ Best Practice . How to calculate risk; 2023. https://bestpractice.bmj.com/info/us/toolkit/learn-ebm/how-to-calculate-risk/. Accessed January 3, 2023. [Google Scholar]
- 241.Ghahremani M, Wang M, Chen HY, Zetterberg H, Smith E, Ismail Z. Alzheimer’s disease neuroimaging initiative*plasma phosphorylated tau at threonine 181 and neuropsychiatric symptoms in preclinical and prodromal Alzheimer disease. Neurology. 2023;100:e683-e693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Dickson DW, Uchikado H, Fujishiro H, Tsuboi Y. Evidence in favor of braak staging of Parkinson’s disease. Mov Disord. 2010;25(Suppl 1):S78-82. [DOI] [PubMed] [Google Scholar]
- 243.Uchihara T. An order in Lewy body disorders: Retrograde degeneration in hyperbranching axons as a fundamental structural template accounting for focal/multifocal Lewy body disease. Neuropathology. 2017;37:129-149. [DOI] [PubMed] [Google Scholar]
- 244.Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24:197-211. [DOI] [PubMed] [Google Scholar]
- 245.Harding AJ, Broe GA, Halliday GM. Visual hallucinations in Lewy body disease relate to Lewy bodies in the temporal lobe. Brain. 2002;125:391-403. [DOI] [PubMed] [Google Scholar]
- 246.Harding AJ, Stimson E, Henderson JM, Halliday GM. Clinical correlates of selective pathology in the amygdala of patients with Parkinson’s disease. Brain. 2002;125:2431-2445. [DOI] [PubMed] [Google Scholar]
- 247.Grinberg LT, Rueb U, Alho ATdL, Heinsen H. Brainstem pathology and non-motor symptoms in PD. J Neurol Sci. 2010;289:81-88. [DOI] [PubMed] [Google Scholar]
- 248.Diederich NJ, Goldman JG, Stebbins GT, Goetz CG. Failing as doorman and disc jockey at the same time: Amygdalar dysfunction in Parkinson's disease. Mov Disord. 2016;31:11-22. [DOI] [PubMed] [Google Scholar]
- 249.Coon EA, Cutsforth-Gregory JK, Benarroch EE. Neuropathology of autonomic dysfunction in synucleinopathies. Mov Disord. 2018; 33: 349-358. [DOI] [PubMed] [Google Scholar]
- 250.Dayan E, Browner N. Alterations in striato-thalamo-pallidal intrinsic functional connectivity as a prodrome of Parkinson’s disease. Neuroimage Clin. 2017;16:313-318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Rolinski M, Griffanti L, Piccini P, et al. Basal ganglia dysfunction in idiopathic REM sleep behaviour disorder parallels that in early Parkinson’s disease. Brain. 2016;139:2224-2234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Mendez MF, Shapira JS, Woods RJ, Licht EA, Saul RE. Psychotic symptoms in frontotemporal dementia: Prevalence and review. Dement Geriatr Cogn Disord. 2008;25:206-211. [DOI] [PubMed] [Google Scholar]

