Skip to main content

Advertisement

Log in

Treatment of Dementia With Lewy Bodies

  • DEMENTIA (E MCDADE, SECTION EDITOR)
  • Published:
Current Treatment Options in Neurology Aims and scope Submit manuscript

Opinion statement

Dementia with Lewy bodies (DLB) is a multisystem disorder with diverse disease expression. A treatment regime restricted to the cognitive aspects of the disease does no favor to patients. Instead, patients should be educated to recognize the symptoms of this multisystem involvement. There are no treatments that slow the progression of disease, but symptomatic treatments can be effective. When thinking about treatment, we find it useful to divide the symptoms and signs into five categories: (a) cognitive features, (b) neuropsychiatric features, (c) motor dysfunction, (d) autonomic dysfunction, and (e) sleep dysfunction. Clinicians, funding bodies and industry are increasingly recognizing the importance of this common and debilitating disease.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. McKeith I et al. Dementia with Lewy bodies: diagnosis and management: third report of the DLB Consortium. Neurology. 2005;65:1863–72.

    Article  PubMed  CAS  Google Scholar 

  2. McKeith IG et al. Prospective validation of consensus criteria for the diagnosis of dementia with Lewy bodies. Neurology. 2000;54(5):1050–8.

    Article  PubMed  CAS  Google Scholar 

  3. Rahkonen T et al. Dementia with Lewy bodies according to the consensus criteria in a general population aged 75 years or older. J Neurol Neurosurg Psychiatry. 2003;74:720–4.

    Article  PubMed  CAS  Google Scholar 

  4. Zaccai J, McCracken C, Brayne C. A systematic review of prevalence and incidence studies of dementia with Lewy bodies. Age Ageing. 2005;34(6):561–6.

    Article  PubMed  Google Scholar 

  5. Boeve B. Diagnosis and management of non Alzheimer dementias. In: Noseworthy JW, editor. Neurological therapeutics: principles and practice. 2005.

  6. McKeith IG et al. Consensus guidelines for the clinical and pathologic diagnosis of dementia with Lewy bodies (DLB): report of the consortium on DLB international workshop. Neurology. 1996;47(5):1113–24.

    Article  PubMed  CAS  Google Scholar 

  7. Vann Jones SA, O’Brien JT. The prevalence and incidence of dementia with Lewy bodies: a systematic review of population and clinical studies. Psychol Med. 2013:1–11. A thorough study of DLB epidemiology, although these are likely understimates.

  8. Nelson PT et al. Low sensitivity in clinical diagnoses of dementia with Lewy bodies. J Neurol. 2010;257(3):359–66. Study showing that many cases of DLB are not diagnosed during life.

    Article  PubMed  Google Scholar 

  9. Boot B. Letter to the Editor the incidence and prevalence of dementia with Lewy bodies is underestimated. Psychol Med. 2013:1–2.

  10. Ferman TJ et al. Inclusion of RBD improves the diagnostic classification of dementia with Lewy bodies. Neurology. 2011;77(9):875–82. Demonstrates that careful diagnosis can lead to accurate identification of patients.

    Article  PubMed  CAS  Google Scholar 

  11. Boeve BF, et al. Clinicopathologic correlations in 172 cases of rapid eye movement sleep behavior disorder with or without a coexisting neurologic disorder. Sleep Med. 2013. Shows that RBD is highly specific to the synucleinopathies.

  12. Ferman TJ et al. DLB fluctuations: specific features that reliably differentiate DLB from AD and normal aging. Neurology. 2004;62(2):181–7.

    Article  PubMed  CAS  Google Scholar 

  13. Lim SM et al. The 18F-FDG PET cingulate island sign and comparison to 123I-beta-CIT SPECT for diagnosis of dementia with Lewy bodies. J Nucl Med. 2009;50(10):1638–45.

    Article  PubMed  CAS  Google Scholar 

  14. Walker Z et al. Striatal dopamine transporter in dementia with Lewy bodies and Parkinson disease: a comparison. Neurology. 2004;62(9):1568–72.

    Article  PubMed  CAS  Google Scholar 

  15. Walker Z et al. Dementia with Lewy bodies: a comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry. 2007;78:1176–81.

    Article  PubMed  Google Scholar 

  16. Treglia G et al. MIBG scintigraphy in differential diagnosis of Parkinsonism: a meta-analysis. Clin Auton Res. 2012;22(1):43–55.

    Article  PubMed  Google Scholar 

  17. Treglia G, et al. Iodine-123 metaiodobenzylguanidine scintigraphy and iodine-123 ioflupane single photon emission computed tomography in Lewy body diseases: complementary or alternative techniques? J Neuroimaging. 2012.

  18. Jellinger KA. Interaction between pathogenic proteins in neurodegenerative disorders. J Cell Mol Med. 2012;16(6):1166–83.

    Article  PubMed  CAS  Google Scholar 

  19. Jellinger KA. Alpha-synuclein pathology in Parkinson’s and Alzheimer’s disease brain: incidence and topographic distribution—a pilot study. Acta Neuropathol. 2003;106(3):191–201.

    Article  PubMed  Google Scholar 

  20. Uchikado H et al. Alzheimer disease with amygdala Lewy bodies: a distinct form of alpha-synucleinopathy. J Neuropathol Exp Neurol. 2006;65(7):685–97.

    Article  PubMed  CAS  Google Scholar 

  21. Schneider JA et al. The neuropathology of older persons with and without dementia from community versus clinic cohorts. J Alzheimers Dis. 2009;18(3):691–701.

    PubMed  Google Scholar 

  22. Ballard C et al. Psychiatric morbidity in dementia with Lewy bodies: a prospective clinical and neuropathological comparative study with Alzheimer’s disease. Am J Psychiatr. 1999;156(7):1039–45.

    PubMed  CAS  Google Scholar 

  23. Ballard C et al. Attention and fluctuating attention in patients with dementia with Lewy bodies and Alzheimer disease. Arch Neurol. 2001;58:977–82.

    Article  PubMed  CAS  Google Scholar 

  24. McKeith I et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomised, double-blind, placebo-controlled international study. Lancet. 2000;356:2031–6.

    Article  PubMed  CAS  Google Scholar 

  25. Aarsland D, Mosimann UP, McKeith IG. Role of cholinesterase inhibitors in Parkinson’s disease and dementia with Lewy bodies. J Geriatr Psychiatry Neurol. 2004;17(3):164–71.

    Article  PubMed  Google Scholar 

  26. Samuel W et al. Better cognitive and psychopathologic response to donepezil in patients prospectively diagnosed as dementia with Lewy bodies: a preliminary study. Int J Geriatr Psychiatry. 2000;15(9):794–802.

    Article  PubMed  CAS  Google Scholar 

  27. Mori E, Ikeda M, Kosaka K. Donepezil for dementia with Lewy bodies: a randomized, placebo-controlled trial. Ann Neurol. 2012;72(1):41–52. Evidence for efficacy of donepezil.

    Article  PubMed  CAS  Google Scholar 

  28. Wild R, Pettit T, Burns A. Cholinesterase inhibitors for dementia with Lewy bodies. Cochrane Database Syst Rev. 2003;3, CD003672.

    PubMed  Google Scholar 

  29. Levy R et al. Lewy bodies and response to tacrine in Alzheimer’s disease. Lancet. 1994;343(8890):176.

    Article  PubMed  CAS  Google Scholar 

  30. Klein JC et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson disease dementia in vivo. Neurology. 2010;74(11):885–92.

    Article  PubMed  CAS  Google Scholar 

  31. Rolinski M et al. Cholinesterase inhibitors for dementia with Lewy bodies. Parkinson’s disease dementia and cognitive impairment in Parkinson’s disease. Cochrane Database Syst Rev. 2012;3, CD006504.

    PubMed  Google Scholar 

  32. Aarsland D et al. Donepezil for cognitive impairment in Parkinson’s disease: a randomised controlled study. J Neurol Neurosurg Psychiatry. 2002;72(6):708–12.

    Article  PubMed  CAS  Google Scholar 

  33. Emre M et al. Rivastigmine for dementia associated with Parkinson’s disease. N Engl J Med. 2004;351:2509–18.

    Article  PubMed  CAS  Google Scholar 

  34. Ravina B et al. Donepezil for dementia in Parkinson’s disease: a randomised, double blind, placebo controlled, crossover study. J Neurol Neurosurg Psychiatry. 2005;76(7):934–9.

    Article  PubMed  CAS  Google Scholar 

  35. Emre M et al. Memantine for patients with Parkinson’s disease dementia or dementia with Lewy bodies: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2010;9(10):969–77. Evidence for efficacy of memantine.

    Article  PubMed  CAS  Google Scholar 

  36. Aarsland D et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicentre trial. Lancet Neurol. 2009;8:613–8.

    Article  PubMed  CAS  Google Scholar 

  37. Cummings JL. The Neuropsychiatric Inventory: assessing psychopathology in dementia patients. Neurology. 1997;48(5 Suppl 6):S10–6.

    Article  PubMed  CAS  Google Scholar 

  38. Thomas A et al. Pathological gambling in Parkinson disease is reduced by amantadine. Ann Neurol. 2010;68(3):400–4.

    Article  PubMed  Google Scholar 

  39. Boot BP et al. Risk factors for dementia with Lewy bodies: a case-control study. Neurology. 2013;81(9):833–40. Risk factors compared to control subjects; also identifies factors that help to differentiate DLB from AD.

    Article  PubMed  CAS  Google Scholar 

  40. Nelson JC, Devanand DP. A systematic review and meta-analysis of placebo-controlled antidepressant studies in people with depression and dementia. J Am Geriatr Soc. 2011;59(4):577–85.

    Article  PubMed  Google Scholar 

  41. Banerjee S et al. Sertraline or mirtazapine for depression in dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-controlled trial. Lancet. 2011;378(9789):403–11.

    Article  PubMed  CAS  Google Scholar 

  42. Weintraub D et al. Antidepressant studies in Parkinson’s disease: a review and meta-analysis. Mov Disord. 2005;20(9):1161–9.

    Article  PubMed  Google Scholar 

  43. Chung TH et al. Systematic review of antidepressant therapies in Parkinson’s disease. Parkinsonism Relat Disord. 2003;10(2):59–65.

    Article  PubMed  CAS  Google Scholar 

  44. Seppi K et al. The movement disorder society evidence-based medicine review update: treatments for the non-motor symptoms of Parkinson’s disease. Mov Disord. 2011;26 Suppl 3:S42–80. This excellent evidence-based review covers many issues applicable to DLB patients.

    Article  PubMed  Google Scholar 

  45. Slaughter JR et al. Prevalence, clinical manifestations, etiology, and treatment of depression in Parkinson’s disease. J Neuropsychiatry Clin Neurosci. 2001;13(2):187–96.

    Article  PubMed  CAS  Google Scholar 

  46. Lemke MR. Depressive symptoms in Parkinson’s disease. Eur J Neurol. 2008;15 Suppl 1:21–5.

    Article  PubMed  Google Scholar 

  47. Takahashi S et al. Depression associated with dementia with Lewy bodies (DLB) and the effect of somatotherapy. Psychogeriatrics. 2009;9(2):56–61.

    Article  PubMed  Google Scholar 

  48. Truong DD, Bhidayasiri R, Wolters E. Management of non-motor symptoms in advanced Parkinson disease. J Neurol Sci. 2008;266(1–2):216–28.

    Article  PubMed  Google Scholar 

  49. Klatka LA, Louis ED, Schiffer RB. Psychiatric features in diffuse Lewy body disease: a clinicopathologic study using Alzheimer’s disease and Parkinson’s disease comparison groups. Neurology. 1996;47(5):1148–52.

    Article  PubMed  CAS  Google Scholar 

  50. Ferman TJ et al. The phenomenology of psychotic features in dementia with Lewy bodies (DLB) and Alheimer’s diesease (AD). Neurology. 2005;64:A257.

    Google Scholar 

  51. Wood LD et al. Clinical review of treatment options for select nonmotor symptoms of Parkinson’s disease. Am J Geriatr Pharmacother. 2010;8(4):294–315.

    Article  PubMed  CAS  Google Scholar 

  52. Kurlan R et al. Quetiapine for agitation or psychosis in patients with dementia and parkinsonism. Neurology. 2007;68(17):1356–63.

    Article  PubMed  CAS  Google Scholar 

  53. Schneider LS, Dagerman K, Insel PS. Efficacy and adverse effects of atypical antipsychotics for dementia: meta-analysis of randomized, placebo-controlled trials. Am J Geriatr Psychiatr. 2006;14(3):191–210.

    Article  Google Scholar 

  54. Schneider L et al. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med. 2006;355:1525–38.

    Article  PubMed  CAS  Google Scholar 

  55. Ballard C, Howard R. Neuroleptic drugs in dementia: benefits and harm. Nat Rev Neurosci. 2006;7(6):492–500.

    Article  PubMed  CAS  Google Scholar 

  56. Rochon PA et al. Antipsychotic therapy and short-term serious events in older adults with dementia. Arch Intern Med. 2008;168(10):1090–6.

    Article  PubMed  Google Scholar 

  57. Musicco M et al. Association between prescription of conventional or atypical antipsychotic drugs and mortality in older persons with Alzheimer’s disease. Dement Geriatr Cogn Disord. 2011;31(3):218–24.

    Article  PubMed  CAS  Google Scholar 

  58. Schneider LS, Dagerman KS, Insel P. Risk of death with atypical antipsychotic drug treatment for dementia: meta-analysis of randomized placebo-controlled trials. JAMA. 2005;294(15):1934–43.

    Article  PubMed  CAS  Google Scholar 

  59. Lopez O, et al. The long-term effects of typical and atypical antipsychotics in patients with Probable Alzheimer’s disease. Am J Psychiatry. (in press).

  60. Aarsland D et al. Neuroleptic sensitivity in Parkinson’s disease and parkinsonian dementias. J Clin Psychiatry. 2005;66(5):633–7.

    Article  PubMed  Google Scholar 

  61. Ballard C et al. Neuroleptic sensitivity in dementia with Lewy bodies and Alzheimer’s disease. Lancet. 1998;351(9108):1032–3.

    Article  PubMed  CAS  Google Scholar 

  62. McKeith I et al. Neuroleptic sensitivity in patients with senile dementia of Lewy body type. BMJ. 1992;305(6855):673–8.

    Article  PubMed  CAS  Google Scholar 

  63. Rochon PA et al. Atypical antipsychotics and parkinsonism. Arch Intern Med. 2005;165(16):1882–8.

    Article  PubMed  CAS  Google Scholar 

  64. Hassan I. Consultation-liaison psychiatry and prevention of severe neuroleptic sensitivity reactions in dementia with Lewy bodies. Australas Psychiatry. 2011;19(6):536–7.

    Article  PubMed  Google Scholar 

  65. Johnell K, Religa D, Eriksdotter M. Differences in drug therapy between dementia disorders in the Swedish Dementia Registry: a nationwide study of over 7,000 patients. Dement Geriatr Cogn Disord. 2013;35:239–48.

    Article  PubMed  CAS  Google Scholar 

  66. Ballard C et al. The dementia antipsychotic withdrawal trial (DART-AD): long-term follow-up of a randomised placebo-controlled trial. Lancet Neurol. 2009;8(2):151–7.

    Article  PubMed  CAS  Google Scholar 

  67. Frieling H et al. Treating dopamimetic psychosis in Parkinson’s disease: structured review and meta-analysis. Eur Neuropsychopharmacol. 2007;17(3):165–71.

    Article  PubMed  CAS  Google Scholar 

  68. Friedman JH, Factor SA. Atypical antipsychotics in the treatment of drug-induced psychosis in Parkinson’s disease. Mov Disord. 2000;15(2):201–11.

    Article  PubMed  CAS  Google Scholar 

  69. Schneider F et al. Psychiatric symptoms in Parkinson’s disease. Eur Arch Psychiatry Clin Neurosci. 2008;258 Suppl 5:55–9.

    Article  PubMed  Google Scholar 

  70. Fernandez HH et al. Long-term outcome of quetiapine use for psychosis among Parkinsonian patients. Mov Disord. 2003;18(5):510–4.

    Article  PubMed  Google Scholar 

  71. Factor SA et al. Clozapine for the treatment of drug-induced psychosis in Parkinson’s disease: results of the 12-week open-label extension in the PSYCLOPS trial. Mov Disord. 2001;16(1):135–9.

    Article  PubMed  CAS  Google Scholar 

  72. Klein C et al. Clozapine in Parkinson’s disease psychosis: 5-year follow-up review. Clin Neuropharmacol. 2003;26(1):8–11.

    Article  PubMed  CAS  Google Scholar 

  73. Pollak P et al. Clozapine in drug induced psychosis in Parkinson’s disease: a randomised, placebo controlled study with open follow up. J Neurol Neurosurg Psychiatry. 2004;75(5):689–95.

    Article  PubMed  CAS  Google Scholar 

  74. Morgante L et al. Quetiapine and clozapine in parkinsonian patients with dopaminergic psychosis. Clin Neuropharmacol. 2004;27(4):153–6.

    Article  PubMed  CAS  Google Scholar 

  75. Merims D et al. Rater-blinded, prospective comparison: quetiapine versus clozapine for Parkinson’s disease psychosis. Clin Neuropharmacol. 2006;29(6):331–7.

    Article  PubMed  CAS  Google Scholar 

  76. Husebo BS, Ballard C, Aarsland D. Pain treatment of agitation in patients with dementia: a systematic review. Int J Geriatr Psychiatry. 26(10):1012–8.

  77. Cohen-Mansfield J, Libin A, Marx MS. Nonpharmacological treatment of agitation: a controlled trial of systematic individualized intervention. J Gerontol A Biol Sci Med Sci. 2007;62(8):908–16.

    Article  PubMed  Google Scholar 

  78. Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological interventions for neuropsychiatric symptoms of dementia. Am J Psychiatry. 169(9):946–53. An excellent review of treatment for all-cause dementia.

  79. Rhodes-Kropf J et al. Managing the patient with dementia in long-term care. Clin Geriatr Med. 2011;27(2):135–52.

    Article  PubMed  Google Scholar 

  80. Ballard C et al. Brief psychosocial therapy for the treatment of agitation in Alzheimer disease (the CALM-AD trial). Am J Geriatr Psychiatr. 2009;17(9):726–33.

    Article  Google Scholar 

  81. Konovalov S, Muralee S, Tampi RR. Anticonvulsants for the treatment of behavioral and psychological symptoms of dementia: a literature review. Int Psychogeriatr. 2008;20(2):293–308.

    Article  PubMed  Google Scholar 

  82. Hicks SW et al. Valproate for the treatment of medication-induced impulse-control disorders in three patients with Parkinson’s disease. Parkinsonism Relat Disord. 2011;17(5):379–81.

    Article  PubMed  CAS  Google Scholar 

  83. Hasnain M et al. Pharmacological management of psychosis in elderly patients with parkinsonism. Am J Med. 2009;122(7):614–22.

    Article  PubMed  CAS  Google Scholar 

  84. Molloy S et al. The role of levodopa in the management of dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2005;76(9):1200–3.

    Article  PubMed  CAS  Google Scholar 

  85. Lucetti C et al. Levodopa response in dementia with lewy bodies: a 1-year follow-up study. Parkinsonism Relat Disord. 2010;16(8):522–6.

    Article  PubMed  Google Scholar 

  86. Lawrence J, Parmenter T, McDonald T. Failure to manage constipation in Parkinson’s disease. A review of medical services a patients perspective. Mov Disord. 2013;28 Suppl 1:187.

    Google Scholar 

  87. Jost WH. Gastrointestinal motility problems in patients with Parkinson’s disease. Effects of antiparkinsonian treatment and guidelines for management. Drugs Aging. 1997;10(4):249–58.

    Article  PubMed  CAS  Google Scholar 

  88. Ashraf W et al. Constipation in Parkinson’s disease: objective assessment and response to psyllium. Mov Disord. 1997;12(6):946–51.

    Article  PubMed  CAS  Google Scholar 

  89. Zangaglia R et al. Macrogol for the treatment of constipation in Parkinson’s disease. A randomized placebo-controlled study. Mov Disord. 2007;22(9):1239–44.

    Article  PubMed  Google Scholar 

  90. Ondo WG et al. Placebo-controlled trial of lubiprostone for constipation associated with Parkinson disease. Neurology. 2012;78(21):1650–4.

    Article  PubMed  CAS  Google Scholar 

  91. Thaisetthawatkul P et al. Autonomic dysfunction in dementia with Lewy bodies. Neurology. 2004;62(10):1804–9.

    Article  PubMed  CAS  Google Scholar 

  92. Hishikawa N et al. Clinical and neuropathological correlates of Lewy body disease. Acta Neuropathol (Berl). 2003;105:341–50.

    Google Scholar 

  93. Schoffer KL et al. Nonpharmacological treatment, fludrocortisone, and domperidone for orthostatic hypotension in Parkinson’s disease. Mov Disord. 2007;22(11):1543–9.

    Article  PubMed  Google Scholar 

  94. Sakakibara R et al. Lower urinary tract function in dementia of Lewy body type. J Neurol Neurosurg Psychiatry. 2005;76(5):729–32.

    Article  PubMed  CAS  Google Scholar 

  95. Ziemssen T, Reichmann H. Non-motor dysfunction in Parkinson’s disease. Parkinsonism Relat Disord. 2007;13(6):323–32.

    Article  PubMed  Google Scholar 

  96. Kay GG et al. Antimuscarinic drugs for overactive bladder and their potential effects on cognitive function in older patients. J Am Geriatr Soc. 2005;53(12):2195–201.

    Article  PubMed  Google Scholar 

  97. Pagoria D, O’Connor RC, Guralnick ML. Antimuscarinic drugs: review of the cognitive impact when used to treat overactive bladder in elderly patients. Curr Urol Rep. 2011;12(5):351–7.

    Article  PubMed  Google Scholar 

  98. Todorova A, Vonderheid-Guth B, Dimpfel W. Effects of tolterodine, trospium chloride, and oxybutynin on the central nervous system. J Clin Pharmacol. 2001;41(6):636–44.

    Article  PubMed  CAS  Google Scholar 

  99. Zesiewicz TA, Helal M, Hauser RA. Sildenafil citrate (Viagra) for the treatment of erectile dysfunction in men with Parkinson’s disease. Mov Disord. 2000;15(2):305–8.

    Article  PubMed  CAS  Google Scholar 

  100. Hussain IF et al. Treatment of erectile dysfunction with sildenafil citrate (Viagra) in parkinsonism due to Parkinson’s disease or multiple system atrophy with observations on orthostatic hypotension. J Neurol Neurosurg Psychiatry. 2001;71(3):371–4.

    Article  PubMed  CAS  Google Scholar 

  101. Hassan A et al. Dopamine agonist-triggered pathological behaviors: surveillance in the PD clinic reveals high frequencies. Parkinsonism Relat Disord. 2011;17(4):260–4.

    Article  PubMed  CAS  Google Scholar 

  102. Malhotra R, Avidan AY. Neurodegenerative disease and REM behavior disorder. Curr Treat Options Neurol. 2012;14(5):474–92.

    Article  PubMed  Google Scholar 

  103. Hogl B et al. Modafinil for the treatment of daytime sleepiness in Parkinson’s disease: a double-blind, randomized, crossover, placebo-controlled polygraphic trial. Sleep. 2002;25(8):905–9.

    PubMed  CAS  Google Scholar 

  104. Kuntz K, et al. Safety, tolerability, and efficacy of armodafinil therapy for hypersomnia associated with dementia with Lewy bodies. Neurology. 2012;(78):P04.192.

  105. Howell MJ, Arneson PA, Schenck CH. A novel therapy for REM sleep behavior disorder (RBD). J Clin Sleep Med. 2011;7(6):639–44.

    PubMed  Google Scholar 

  106. Boeve B, Silber M, Ferman T. Melatonin for treatment of REM sleep behavior disorder in neurologic disorders: results in 14 patients. Sleep Med. 2003;4:281–4.

    Article  PubMed  Google Scholar 

  107. Dowling GA et al. Melatonin for sleep disturbances in Parkinson’s disease. Sleep Med. 2005;6(5):459–66.

    Article  PubMed  Google Scholar 

  108. Medeiros CA et al. Effect of exogenous melatonin on sleep and motor dysfunction in Parkinson’s disease. A randomized, double blind, placebo-controlled study. J Neurol. 2007;254(4):459–64.

    Article  PubMed  CAS  Google Scholar 

  109. Mace N, Rabins P. The 36-hour day: a family guide to caring for persons with Alzheimer disease, related dementing illnesses, and memory loss in later life. 5th ed. Baltimore: JHUP; 2011.

    Google Scholar 

  110. Chenoweth L et al. Caring for Aged Dementia Care Resident Study (CADRES) of person-centred care, dementia-care mapping, and usual care in dementia: a cluster-randomised trial. Lancet Neurol. 2009;8(4):317–25.

    Article  PubMed  Google Scholar 

  111. Moyle W et al. The effect of massage on agitated behaviours in older people with dementia: a literature review. J Clin Nurs. 2013;22(5–6):601–10.

    PubMed  Google Scholar 

  112. Holliday-Welsh DM, Gessert CE, Renier CM. Massage in the management of agitation in nursing home residents with cognitive impairment. Geriatr Nurs. 2009;30(2):108–17.

    Article  PubMed  Google Scholar 

  113. Ueda T et al. Effects of music therapy on behavioral and psychological symptoms of dementia: a systematic review and meta-analysis. Ageing Res Rev. 2013;12(2):628–41.

    Article  PubMed  Google Scholar 

  114. Raglio A et al. Music, music therapy and dementia: a review of literature and the recommendations of the Italian Psychogeriatric Association. Maturitas. 2012;72(4):305–10.

    Article  PubMed  CAS  Google Scholar 

  115. Iranzo A et al. Decreased striatal dopamine transporter uptake and substantia nigra hyperechogenicity as risk markers of synucleinopathy in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study. Lancet Neurol. 2010;9(11):1070–7.

    Article  PubMed  CAS  Google Scholar 

  116. McDade EM, Boot BP, Christianson TJ, Pankratz VS, Boeve BF, Ferman TJ, et al. Subtle gait changes in patients with REM sleep behavior disorder. Mov Disord. 2013. doi:10.1002/mds.25653.

  117. Boeve B. REM sleep behavior disorder: updated review of the core features, the REM sleep behavior disorder-neurodegenerative disease association, evolving concepts, controversies, and future directions. Ann N Y Acad Sci. 2010;1184:17–56.

    Article  Google Scholar 

Download references

Compliance with Ethics Guidelines

Conflict of Interest

Brendon P. Boot has served as an investigator for clinical trials sponsored by Pfizer, Janssen Pharmaceutica, and Bristol-Myers Squibb.

Eric M. McDade has served as a consultant for the Dominantly Inherited Alzheimer Network Therapeutic Trial Unit, has received grant support from the National Institutes of Health, The Pittsburgh Foundation, and the University of Pittsburgh Alzheimer Disease Research Center, has received honoraria from the Alzheimer Association, and has received royalties from UptoDate.

Scott M. McGinnis has served as an investigator for clinical trials sponsored by Pfizer, Janssen Pharmaceutica, Bristol-Myers Squibb, Mediviation, TauRx, and Allon Therapeutics.

Bradley F. Boeve has served as an investigator for clinical trials sponsored by Cephalon, Allon Pharmaceuticals and GE Healthcare. He receives royalties from the publication of a book entitled Behavioral Neurology of Dementia (Cambridge Medicine, 2009). He has received honoraria for development of educational presentations from the American Academy of Neurology. He serves on the Scientific Advisory Board of the Tau Consortium. He receives research support from the National Institute on Aging (P50 AG016574, U01 AG006786, RO1 AG032306, RO1 AG041797) and the Mangurian Foundation.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Brendon P. Boot MBBS.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Boot, B.P., McDade, E.M., McGinnis, S.M. et al. Treatment of Dementia With Lewy Bodies. Curr Treat Options Neurol 15, 738–764 (2013). https://doi.org/10.1007/s11940-013-0261-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11940-013-0261-6

Keywords

Navigation