542
Views
3
CrossRef citations to date
0
Altmetric
Editorial

Psychosis in Parkinson’s disease: how should we treat it?

, &
Pages 1243-1246 | Published online: 10 Jan 2014

Psychosis (delusions and/or hallucinations) is a common and problematic non-motor feature of Parkinson’s disease (PD). Hallucinations are sensory perceptions that occur without external stimulation of the relevant sensory organ. In PD, a typical visual hallucination is a complex visual image experienced when the patient is alert and has their eyes open. The image usually appears somewhat blurred. There is a predominance of human or animal forms, some with emotional significance. The hallucinations are sometimes ‘friendly’ but may also consist of frightening images. Hallucinations are generally more common in the evening Citation[1]. Most patients ‘know’ that they are hallucinating (benign hallucinosis), but approximately 5% experience delusions and hallucinations with lack of insight Citation[2]. Auditory and tactile hallucinations may also occur, but are less frequent and usually co-exist with visual hallucinations Citation[1]. A sense of presence is another common hallucinatory symptom.

Delusions are false beliefs based on incorrect inferences. They are commonly anteceded by hallucinations, but can occur in their absence. In PD, common themes of the delusions are spousal infidelity or persecution.

Estimates of prevalence vary. In one US study of 102 consecutive clinic attenders, a prevalence of visual hallucinations of 26% was found Citation[3]. In a systematic review of clinic populations, the prevalence of hallucinations alone was between 21 and 46% Citation[4]. A community survey in Norway yielded 9% prevalence for hallucinations and 6% for combined hallucinations and delusions Citation[2]. In patients attending a specialist clinic, the figure for the combined symptoms was between 10 and 15% Citation[5,6]. The combination of hallucinations and delusions together is regarded as a more serious problem as it usually results in subjective distress and behavioral disturbance.

Psychosis is an indicator of poor prognosis in PD Citation[7] and is often the factor that leads to institutional care Citation[8]. In addition, carer burden and mortality are high Citation[7–10]. There is no consistent relationship with dose, drug type or on–off phenomena Citation[11]. There is some evidence that the incidence of psychosis is higher in patients treated with dopamine agonists when compared with those receiving levodopa monotherapy Citation[12–15].

Treating psychosis is one of the most difficult challenges for the parkinsonologist. Even in early monotherapy in new patients, drug- induced psychosis can be a major dose-limiting side effect. There is an increase in frequency with disease duration. Other risk factors are age, cognitive impairment and sleep disturbance Citation[4,16]. There are some general principles for managing PD psychosis. The most important first step is to exclude delirium in all cases of first-presentation psychosis. Drug effects, concurrent physical illness, electrolyte imbalance and systemic infection should all be excluded. For fully effective management, a multidisciplinary approach is desirable. Ideally, this should be comprised of the patient and carer, PD physician, PD nurse, psychiatrist and social worker. Education and ‘insight-building’ work with the patient and carer will allow them to better understand and cope with the condition. Several simple measures may improve the situation; for example, improved lighting and avoidance of patterned furniture/carpets. Some patients find that staring directly at the hallucinations causes them to disappear. Occasional visual hallucinations with preserved insight often do not require anything other than reassurance.

If these simple approaches are unsuccessful, it is necessary to consider reduction of dose or elimination of one or more antiparkinsonian drugs. This is usually carried out in the following order: anticholinergics, amantadine, selegiline, dopamine agonists, catechol-O-methyltransferase (COMT) inhibitors and l-dopa. There is often a tricky balancing act between psychosis and motor function (the motion/emotion dilemma) Citation[6]. It may be necessary to accept some deterioration in motor function.

Drug treatment is necessary if all the above measures fail. Attempts to control psychosis using the older ‘typical’ antipsychotic or neuroleptic drugs led to worsening of motor symptoms. The introduction of second-generation or ‘atypical’ antipsychotic drugs and cholinesterase inhibitors opened up a new range of therapeutic options. The atypical antipsychotics are loosely defined as a group of neuroleptic drugs that fail to cause catalepsy in rats (rigid maintenance of a body position over an extended period of time). Their characteristics in humans include a low incidence of extrapyramidal side effects and a lack of induction of significant prolactin elevation. They have a higher serotonin (5HT)-2 versus D2 receptor affinity than classic neuroleptic drugs. Of the atypical antipsychotics, risperidone, olanzapine, clozapine and quetiapine have all been used to treat psychosis in PD with varying success Citation[17,18]. Risperidone and olanzapine are associated with worsening of motor function Citation[19,20]. Quetiapine is the treatment favored by many psychiatrists and neurologists in clinical practice. However, the evidence for its efficacy is mixed. In two recent randomized trials where it was compared with clozapine, it was found to be at least as effective Citation[21,22]. In addition, in two recent open-label trials, it showed benefit in the control of psychosis Citation[23,24]. However, two recent placebo-controlled trials had negative results Citation[25,26]. Further randomized, controlled trials are required to settle the debate. Clozapine is the most effective agent and does not worsen parkinsonism. However, the need for regular white blood cell monitoring owing to the risk of fatal agranulocytosis tends to restrict its use to patients who have not responded to other agents. In addition, patients usually need to be started on it in an inpatient setting, which is obviously costly. Aripiprazole and ziprasidone have also been used. However, the evidence for these is weak, as there have been no randomized, controlled trials. One case series using aripirazole was not particularly encouraging, causing a significant worsening of PD symptoms Citation[27].

Recently, there has been concern regarding the safety of the atypical antipsychotics in certain patient populations. A recent meta-analysis examining 15 trials of atypicals in patients with dementia showed that death occurred more often among patients randomized to drug Citation[28]. The odds ratio was 1.54 (95% confidence interval: 1.06–2.23; p = 0.01). The US FDA report a similar increased risk between 1.6 and 1.7. Deaths were most frequently due to cardiac-related events or infections such as pneumonia. Consequently, a black box warning has been added to the label of all atypical antipsychotics in the USA Citation101.

Cholinesterase inhibitors are another recently introduced treatment option for PD psychosis, particularly where there is associated dementia. The brains of PD patients show marked degeneration of cholinergic neurons Citation[29]. This has also been shown in vivo using functional neuroimaging Citation[30]. Relative dopaminergic over-activity in the mesocortical system combined with subcortical cholinergic neuronal loss could provide a more permissive environment for the development of psychosis. A review published in 2003 tentatively stated that cholinesterase inhibitors were a potentially useful treatment in PD psychosis Citation[31]. This conclusion drew on the evidence from one double-blind, placebo controlled trial of rivastigmine in dementia with Lewy bodies patients Citation[32] and from four small open-label studies in PD dementia Citation[33–36].

Since 2003, four double-blind, placebo-controlled randomized, controlled trials have been reported examining the use of cholinesterase inhibitors in Parkinson’s disease with dementia (PDD) where neuropsychiatric symptoms were reported as secondary end points. A large randomized, controlled trial using rivastigmine Citation[37] reported a statistically significant reduction in Neuropsychiatric Inventory (NPI) Citation[38] scores compared with placebo. Nausea and tremor were common adverse events. It is important to note, however, that the initial NPI scores were rather low (the mean score in the rivastigmine group was 12.7 out of a possible maximum of 120). The mean change in this score at week 24 was of only 2 points and so it is difficult to be certain of the clinical significance of this. In addition, NPI scores represent a composite of various neuropsychiatric symptoms. It is therefore difficult to assign clinical meaning to a single change in this score. Three randomized, controlled trials with donepezil showed no significant improvement in NPI scores versus placebo Citation[39–41]. An open-label study using donepezil in PDD reports significant improvements in hallucinations and delusions Citation[42]. Aarsland and colleagues report a further open-label trial using galantamine in patients with PD dementia Citation[43]. This trial again showed improvements in hallucinations.

With regard to tolerability, there is evidence from the above trials that cholinesterase inhibitors worsen motor function, especially tremor, although this is not always picked up by United Parkinson’s Disease Rating Scale scores Citation[44]. In addition, nausea appears to be a significant problem, affecting 29% of patients receiving rivastigmine in the largest trial.

Further randomized, controlled trials are required to specifically assess the efficacy of cholinesterase inhibitors in PD psychosis.

In a recent paper discussing the etiology of visual hallucinations, Collerton and colleagues postulate a primary role for cholinergic dysfunction and a secondary role for dopamine dysfunction Citation[45]. Therefore, cholinergic hypoactivity alone, or dopamine hyperactivity only if cholinergic hypoactivity is present, are the neurochemical mechanisms for hallucinations. This suggestion is supported by evidence that combined cholinergic and neuroleptic treatment is effective in reducing hallucinations in Alzheimer’s disease Citation[46]. One future direction would be to carry out a similar study in PD patients. As the disease progresses, deterioration in cholinergic function combined with dopaminergic hyperactivity is one postulated set of factors that leads to the development of hallucinations.

References

  • Fenelon G, Mahieux F, Huon R, Ziegler M. Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors. Brain123(Pt 4), 733–745 (2000).
  • Aarsland D, Larsen JP, Cummins JL, Laake K. Prevalence and clinical correlates of psychotic symptoms in Parkinson disease: a community-based study. Arch. Neurol.56(5), 595–601 (1999).
  • Holroyd S, Currie L, Wooten GF. Prospective study of hallucinations and delusions in Parkinson’s disease. J. Neurol. Neurosurg. Psychiatr.70(6), 734–738 (2001).
  • Barnes J, David AS. Visual hallucinations in Parkinson’s disease: a review and phenomenological survey. J. Neurol. Neurosurg. Psychiatr.70(6), 727–733 (2001).
  • Cummings JL. Behavioral complications of drug treatment of Parkinson’s disease. J. Am. Geriatr. Soc.39(7), 708–716 (1991).
  • Cummings JL. Managing psychosis in patients with Parkinson’s disease. N. Engl. J. Med.340(10), 801–803 (1999).
  • Goetz CG, Stebbins GT. Mortality and hallucinations in nursing home patients with advanced Parkinson’s disease. Neurology45(4), 669–671 (1995).
  • Aarsland D, Larsen JP, Tandberg E, Laake K. Predictors of nursing home placement in Parkinson’s disease: a population-based, prospective study. J. Am. Geriatr. Soc.48(8), 938–942 (2000).
  • Goetz CG, Leurgans S, Pappert EJ, Raman R, Stemer AB. Prospective longitudinal assessment of hallucinations in Parkinson’s disease. Neurology57(11), 2078–2082 (2001).
  • Gimenez-Roldan S, Navarro E, Mateo D. Effects of quetiapine at low doses on psychosis motor disability and stress of the caregiver in patients with Parkinson’s disease. Rev. Neurol.36(5), 401–404 (2003).
  • Goetz CG, Pappert EJ, Blasucci LM et al. Intravenous levodopa in hallucinating Parkinson’s disease patients: high-dose challenge does not precipitate hallucinations. Neurology50(2), 515–517 (1998).
  • Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson’s disease: a randomized controlled trial. JAMA284, 1931–1938 (2000).
  • Rascol O BD, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N. Engl. J. Med.342, 1484–1491 (2000).
  • Bracco F, Battaglia A, Chouza C et al. The long-acting dopamine receptor agonist cabergoline in early Parkinson's disease: final results of a 5-year, double-blind, levodopa-controlled study. CNS Drugs18(11), 733–746 (2004) [erratum appears in CNS Drugs19(7), 633 (2005)].
  • Clarke CE, Speller JM. Pergolide for levodopa-induced complications in Parkinson’s disease. Cochrane Database Syst. Rev.2, CD000235 (2000).
  • Barnes J, Boubert L, Harris J, Lee A, David AS. Reality monitoring and visual hallucinations in Parkinson’s disease. Neuropsychologia41(5), 565–574 (2003).
  • Friedman JH, Factor SA. Atypical antipsychotics in the treatment of drug-induced psychosis in Parkinson’s disease. Mov. Disord.15(2), 201–211 (2000).
  • Wolters EC, Berendse HW. Management of psychosis in Parkinson’s disease. Curr. Opin. Neurol.14(4), 499–504 (2001).
  • Goetz CG, Blasucci LM, Leurgans S, Pappert EJ. Olanzapine and clozapine: comparative effects on motor function in hallucinating PD patients. Neurology55(6), 789–794 (2000).
  • Ellis T, Cudkowicz ME, Sexton PM, Growdon JH. Clozapine and risperidone treatment of psychosis in Parkinson’s disease. J. Neuropsychiatry Clin. Neurosci.12(3), 364–369 (2000).
  • Merims DSH, Balas M, Peretz C, Giladi N. Rater-blinded, prospective study comparing the therapeutic benefit of clozapine vs. quetiapine on psychosis in patients with Parkinson’s disease. Presented at the 9th International Congress of Parkinson’s Disease and Movement Disorders (2005).
  • Morgante L, Epifanio A, Spina E et al. Quetiapine and clozapine in parkinsonian patients with dopaminergic psychosis. Clin. Neuropharmacol.27(4), 153–156 (2004) [Erratum appears in Clin. Neuropharmacol.27(5), 256 (2004)].
  • Juncos JL, Roberts VJ, Evatt ML et al. Quetiapine improves psychotic symptoms and cognition in Parkinson’s disease. Mov. Disord.19(1), 29–35 (2004).
  • Mancini F, Tassorelli C, Martignoni E et al. Long-term evaluation of the effect of quetiapine on hallucinations, delusions and motor function in advanced Parkinson disease. Clin. Neuropharmacol.27(1), 33–37 (2004).
  • Ondo W. Double-blind, placebo-controlled, unforced titration parallel trial of quetiapine for dopaminergic-induced hallucinations in Parkinson’s disease. Mov. Disord. (2004).
  • Rabey JM, Prohorov T, Miniovich A, Klein C. The effect of quetiapine in Parkinson’s disease (PD) psychotic patients: a double-blind labelled study of 3 months duration. Presented at the 9th International Congress of Parkinson’s Disease and Movement Disorders (2005).
  • Fernandez HH, Trieschmann ME, Friedman JH. Aripiprazole for drug-induced psychosis in Parkinson disease: preliminary experience. Clin. Neuropharmacol.27(1), 4–5 (2004).
  • Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA294(15), 1934–1943 (2005).
  • Whitehouse PJ, Hedreen JC, White CL 3rd, Price DL. Basal forebrain neurons in the dementia of Parkinson disease. Ann. Neurol.13(3), 243–248 (1983).
  • Bohnen NI, Kaufer DI, Ivanco LS et al. Cortical cholinergic function is more severely affected in parkinsonian dementia than in Alzheimer disease: an in vivo positron emission tomographic study. Arch. Neurol.60(12), 1745–1748 (2003).
  • Poewe W. Psychosis in Parkinson’s disease. Mov. Disord.18(Suppl. 6), 80–87 (2003).
  • McKeith I, Del Ser T, Spano P et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet356(9247), 2031–2036 (2000).
  • Bullock R, Cameron A. Rivastigmine for the treatment of dementia and visual hallucinations associated with Parkinson’s disease: a case series. Curr. Med. Res. Opin.18(5), 258–264 (2002).
  • Fabbrini G, Barbanti P, Aurilia C, Pauletti C, Lenzi GL, Meco G. Donepezil in the treatment of hallucinations and delusions in Parkinson’s disease. Neurol. Sci.23(1), 41–43 (2002).
  • Bergman J, Lerner V. Successful use of donepezil for the treatment of psychotic symptoms in patients with Parkinson’s disease. Clin. Neuropharmacol.25(2), 107–110 (2002).
  • Reading PJ, Luce AK, McKeith IG. Rivastigmine in the treatment of parkinsonian psychosis and cognitive impairment: preliminary findings from an open trial. Mov. Disord.16(6), 1171–1174 (2001).
  • Emre M, Aarsland D, Albanese A et al. Rivastigmine for dementia associated with Parkinson’s disease. N. Engl. J. Med.351(24), 2509–2518 (2004).
  • Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology44, 2308–2314 (1994).
  • Ravina B, Putt M, Siderowf A et al. Donepezil for dementia in Parkinson's disease: a randomised, double blind, placebo controlled, crossover study. J. Neurol. Neurosurg. Psychiatr.76(7), 934–939 (2005).
  • Leroi I, Brandt J, Reich SG et al. Randomized placebo-controlled trial of donepezil in cognitive impairment in Parkinson’s disease. Int. J. Geriatr. Psychiatry19(1), 1–8 (2004).
  • Aarsland D, Laake K, Larsen JP, Janvin C. Donepezil for cognitive impairment in Parkinson’s disease: a randomised controlled study. J. Neurol. Neurosurg. Psychiatr.72(6), 708–712 (2002) [Erratum appears in J. Neurol. Neurosurg. Psychiatry73(3), 354 (2002)].
  • Thomas AJ, Burn DJ, Rowan EN et al. A comparison of the efficacy of donepezil in Parkinson’s disease with dementia and dementia with Lewy bodies. Int. J. Geriatr. Psychiatry20(10), 938–944 (2005).
  • Aarsland D, Hutchinson M, Larsen JP. Cognitive, psychiatric and motor response to galantamine in Parkinson’s disease with dementia. Int. J. Geriatr. Psychiatry18(10), 937–941 (2003).
  • Fahn S ER, Members of the UPDRS Development Committee. Unified Parkinson’s Disease Rating Scale. Recent Developments in Parkinson’s Disease. Fahn S, Marsden CD, Calne DB, Goldstein M (Eds). (11), 153–163 (1987).
  • Collerton D, Perry E, McKeith I. Why people see things that are not there: a novel perception and attention deficit model for recurrent complex visual hallucinations. Behav. Brain Sci.28(6), 737–757; discussion 757–794 (2005).
  • Bergman J, Brettholz I, Shneidman M, Lerner V. Donepezil as add-on treatment of psychotic symptoms in patients with dementia of the Alzheimer’s type. Clin. Neuropharmacol.26(2), 88–92 (2003).

Wesbite

  • US Food and Drug Administration (Public Health Advisory). Deaths with antipsychotics in elderly patients with behavioural disturbances (2005). www.fda.gov/cder/drug/advisory/antipsychotics.htm

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.