6,341
Views
29
CrossRef citations to date
0
Altmetric
Review

Clinical drug development for dementia with Lewy bodies: past and present

ORCID Icon, , , &
Pages 951-965 | Received 30 Aug 2019, Accepted 14 Oct 2019, Published online: 28 Oct 2019

References

  • Mueller C, Ballard C, Corbett A, et al. The prognosis of dementia with Lewy bodies. Lancet Neurol. 2017;16:390–398.
  • Hogan DB, Fiest KM, Roberts JI, et al. The prevalence and incidence of dementia with Lewy bodies: a systematic review. Can J Neurol Sci. 2016;43(1):S83–95.
  • Mason AR, Ziemann A, Finkbeiner S. Targeting the low-hanging fruit of neurodegeneration. Neurology. 2014;83:1470–1473.
  • Aarsland D, Ballard C, Rongve A, et al. Clinical trials of dementia with Lewy bodies and Parkinson’s disease dementia. Curr Neurol Neurosci Rep. 2012;12(5):492–501.
  • Palermo G, Ceravolo R, Bonuccelli U. Advances in the pharmacotherapeutic management of dementia with Lewy bodies. Expert Opin Pharmacother. 2018;19(15):1643–1653.
  • Walker Z, Possin KL, Boeve BF, et al. Lewy body dementias. Lancet. 2015;386:1683–1697.
  • Lippa CF, Duda JE, Grossman M, et al. DLB and PDD boundary issues: diagnosis, treatment, molecular pathology, and biomarkers. Neurology. 2007;68(11):812–819.
  • McKeith I, Mintzer J, Aarsland D, et al. Dementia with Lewy bodies. Lancet Neurol. 2004;3:19–28.
  • Emre M, Aarsland D, Brown R, et al. Clinical diagnostic criteria for dementia associated with Parkinson’s disease. Mov Disord. 2007;22(12):1689–1707.
  • Merdes AR, Hansen LA, Jeste DV, et al. Influence of Alzheimer pathology on clinical diagnostic accuracy in dementia with Lewy bodies. Neurology. 2003;60:1586–1590.
  • Berg D, Postuma RB, Bloem B, et al. Time to redefine PD? Introductory statement of the MDS task force on the definition of Parkinson’s disease. Mov Disord. 2014;29(4):454–462.
  • Boeve BF, Dickson DW, Duda JE, et al. Arguing against the proposed definition changes of PD. Mov Disord. 2016;31(11):1619–1622.
  • McKeith IG, Boeve BF, Dickson DW, et al. Diagnosis and management of dementia with Lewy bodies: fourth report of the DLB consortium. Neurology. 2017;89:88–100.
  • McKeith IG, Dickson DW, Lowe J, et al. Dementia with Lewy bodies: diagnosis and management: third report of the DLB consortium. Neurology. 2005;65:1863–1872.
  • McKeith I, Taylor JP, Thomas A, et al. Revisiting DLB diagnosis: a consideration of prodromal DLB and of the diagnostic overlap with Alzheimer’s disease. J Geriatr Psychiatry Neurol. 2016;29:249–253.
  • Ferman TJ, Smith GE, Boeve BF, et al. Neuropsychological differentiation of dementia with Lewy bodies from normal aging and Alzheimer’s disease. Clin Neuropsychol. 2006;20(4):623–636.
  • Ballard C, Aarsland D, Francis P, et al. Neuropsychiatric symptoms in patients with dementias associated with cortical Lewy bodies: pathophysiology, clinical features, and pharmacological management. Drug Aging. 2013;30:603–611.
  • Chan PC, Lee HH, Hong CT, et al. REM sleep behavior disorder (RBD) in dementia with Lewy bodies (DLB). Behav Neurol. 2018;2018:1–10.
  • Ferman TJ, Boeve BF, Smith GE, et al. Inclusion of RBD improves the diagnostic classification of dementia with Lewy bodies. Neurology. 2011;77:875–882.
  • Chwiszczuk L, Breitve M, Hynninen M, et al. Higher frequency and complexity of sleep disturbances in dementia with Lewy bodies as compared to Alzheimer’s disease. Neurodegener Dis. 2016;16:152–160.
  • Burn DJ, Rowan EN, Minett T, et al. Extrapyramidal features in Parkinson’s disease with and without dementia and dementia with Lewy bodies: a cross-sectional comparative study. Mov Disord. 2003;18:884–889.
  • Ballard C, Grace J, McKeith I, et al. Neuroleptic sensitivity in dementia with Lewy bodies and Alzheimer’s disease. Lancet. 1998;351:1032–1033.
  • Trollor JN, Chen X, Sachdev PS. Neuroleptic malignant syndrome associated with atypical antipsychotic drugs. CNS Drugs. 2009;23(6):477–492.
  • Postuma RB, Gagnon JF, Pelletier A, et al. Prodromal autonomic symptoms and signs in Parkinson’s disease and dementia with Lewy bodies. Mov Disord. 2013;28:597–604.
  • Hansen D, Ling H, Lashley T, et al. Review: clinical, neuropathological and genetic features of Lewy body dementias. Neuropathol Appl Neurobiol. 2019 Apr 12. [Epub ahead of print]. DOI:10.1111/nan.12554.
  • Graff-Radford J, Murray ME, Lowe VJ, et al. Dementia with Lewy bodies: basis of cingulate island sign. Neurology. 2014;83:801–809.
  • Pillai JA, Wu G, Tousi B, et al. Amygdala sign, a FDG-PET signature of dementia with Lewy bodies. Parkinsonism Relat Disord. 2019:pii: S1353-8020(19)30098–7. [Epub ahead of print]. DOI:10.1016/j.parkreldis.2019.03.005.
  • Walker L, Stefanis L, Attems J. Clinical and neuropathological differences between Parkinson’s disease, Parkinson’s disease dementia and dementia with Lewy bodies – current issues and future directions. J Neurochem. 2019 Mar 20. [Epub ahead of print]. DOI:10.1111/jnc.14698.
  • Volpicelli-Daley LA, Luk KC, Patel TP, et al. Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. 2011;72(1):57–71.
  • Ballard C, Ziabreva I, Perry R, et al. Differences in neuropathologic characteristics across the Lewy body dementia spectrum. Neurology. 2006;67:1931–1934.
  • Van Steenoven I, Aarsland D, Weintraub D, et al. Cerebrospinal fluid Alzheimer’s disease biomarkers across the spectrum of Lewy body diseases: results from a large multicenter cohort. J Alzheimers Dis. 2016;54:287–295.
  • Halliday GM, Song YI, Harding AJ. Striatal β-amyloid in dementia with Lewy bodies but not Parkinson’s disease. J Neural Transm. 2011;118:713–719.
  • Tsuboi Y, Wszolek ZK, Graff-Radford NR, et al. Tau pathology in the olfactory bulb correlates with Braak stage, Lewy body pathology and apolipoprotein epsilon 4. Neuropathol Appl Neurobiol. 2003;29(5):503–510.
  • Overk CR, Masliah E. Pathogenesis of synaptic degeneration in Alzheimer’s disease and Lewy body disease. Biochem Pharmacol. 2014;88:508–516.
  • Klein JC, Eggers C, Kalbe E, et al. Neurotransmitter changes in dementia with Lewy bodies and Parkinson’s disease dementia in vivo. Neurology. 2010;74:885–892.
  • Creese B, Ballard C, Aarsland D, et al. Determining the association of the 5HTTLPR polymorphism with delusions and hallucinations in Lewy body dementias. Am J Geriatr Psychiatry. 2014;22(6):580–586.
  • Guerreiro R, Ross OA, Kun-Rodrigues C, et al. Investigating the genetic architecture of dementia with Lewy bodies: a two-stage genome-wide association study. Lancet Neurol. 2018;17:64–74.
  • Cummings J, Morstorf T, Lee G. Alzheimer’s disease drug development pipeline: 2016. Alzheimers Demen. 2016;2:222–232.
  • Cummings J, Lee G, Morstorf T, et al. Alzheimer’s disease drug development pipeline: 2017. Alzheimers Demen. 2017;3:367–384.
  • Cummings J, Lee G, Morstorf T, et al. Alzheimer’s disease drug development pipeline: 2018. Alzheimers Demen. 2018;4:195–214.
  • Cummings J, Lee G, Morstorf T, et al. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Demen. 2019;5:272–293.
  • 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.
  • Tousi B. Diagnosis and management of cognitive and behavioral changes in dementia with Lewy bodies. Curr Treat Options Neurol. 2017;19:42.
  • Hershey LA, Coleman-Jackson R. Pharmacological management of dementia with Lewy bodies. Drugs Aging. 2019;36:309–319.
  • Boot BP. Comprehensive treatment of dementia with Lewy bodies. Alzheimers Res Ther. 2015;7(1):45.
  • Matsunaga S, Kishi T, Yasue I, et al. Cholinesterase inhibitors for Lewy body disorders: a meta-analysis. Int J Neuropsychopharmacol. 2015;19:1–15.
  • Bhasin M, Rowan E, Edwars K, et al. Cholinesterase inhibitors in dementia with Lewy bodies: a comparative analysis. Int J Geriatr Psychiatry. 2007;22(9):890–895.
  • Aarsland D, Mosimann UP, McKeith IG. Role of cholinesterase inhibitors in Parkinson’s disease and dementia with Lewy bodies. J Geriatr Psychiatry Neurol. 2004;17:164–171.
  • Tiraboschi P, Hansen LA, Alford M, et al. Cholinergic dysfunction in diseases with Lewy bodies. Neurology. 2000;54(2):407–411.
  • Noufi P, Khoury R, Jeyakumar S, et al. Use of cholinesterase inhibitors in non-Alzheimer’s dementias. Drugs Aging. 2019;36(8):719–731.
  • Eisai [Internet]. News release: Aricept approved in Japan as treatment for dementia with Lewy bodies; 2014 [cited 2019 Jul 21]. Available from: https://www.eisai.com/news/news201452.html
  • Eisai [Internet]. News release: Aricept approved in the Philippines for new indication as treatment for dementia with Lewy bodies; 2016 [cited 2019 Jul 21]. Available from: https://www.eisai.com/news/news201624.html
  • 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(1):41–52.
  • 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(3–4):229–241.
  • Ikeda M, Mori E, Matsuo K, et al. Donepezil for dementia with Lewy bodies: a randomized, placebo-controlled, confirmatory phase III trial. Alzheimers Res Ther. 2015;7:4.
  • Mori E, Ikeda M, Nagai R, et al. Long-term donepezil use for dementia with Lewy bodies: result from an open-label extension of phase III trial. Alzheimers Res Ther. 2015;7(1):5.
  • McKeith I, DelSer T, Spano P, et al. Efficacy of rivastigmine in dementia with Lewy bodies: a randomized, double-blind, placebo-controlled international study. Lancet. 2000;356(9247):2031–2036.
  • Grace J, Daniel S, Stevens T, et al. Long-term use of rivastigmine in patients with dementia with Lewy bodies: an open-label trial. Int Psychogeriatr. 2001;13(2):199–205.
  • Edwards K, Royall D, Hershey L, et al. Efficacy and safety of galantamine in patients with dementia with Lewy bodies: a 24-week open-label study. Dement Geriatr Cogn Disord. 2007;23(6):401–405.
  • Emre M, Tsolaki M, Bonuccelli U, 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):969–977.
  • Aarsland D, Ballard C, Walker Z, et al. Memantine in patients with Parkinson’s disease dementia or dementia with Lewy bodies: a double-blind, placebo-controlled, multicenter trial. Lancet Neurol. 2009;8(7):613–618.
  • Wesnes KA, Aarsland D, Ballard C, et al. Memantine improves attention and episodic memory in Parkinson’s disease dementia and dementia with Lewy bodies. Int J Geriatr Psychiatry. 2015;30(1):46–54.
  • Brennan L, Pantelyat A, Duda JE, et al. Memantine and cognition in Parkinson’s disease dementia/dementia with Lewy bodies: a meta-analysis. Mov Disord Clin Pract. 2015;3(2):161–167.
  • Velayudhan L, Ffytche D, Ballard C, et al. New therapeutic strategies for Lewy body dementias. Curr Neurol Neurosci Rep. 2017;17(9):68.
  • Phan S, Osae S, Morgan J, et al. Neuropsychiatric symptoms in dementia: considerations for pharmacotherapy in the USA. Drugs R D. 2019;19:93–115.
  • Brodaty H, Arasaratnam C. Meta-analysis of nonpharmacological interventions for neuropsychiatric symptoms of dementia. Am J Psychiatry. 2012;169:946–953.
  • Ukai K, Fujishiro H, Iritani S, et al. Long-term efficacy of donepezil for relapse of visual hallucinations in patients with dementia with Lewy bodies. Psychogeriatrics. 2015;15:133–137.
  • Edwards KR, Hershey L, Wray L, et al. Efficacy and safety of galantamine in patients with dementia with Lewy bodies: a 12-week interim analysis. Dement Geriatr Cogn Disord. 2004;17(1):40–48.
  • Takahashi H, Yoshida K, Sugita T, et al. Quetiapine treatment of psychotic symptoms and aggressive behavior in patients with dementia with Lewy bodies: a case series. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(3):549–553.
  • Kurlan R, Cummings J, Raman R, et al. Quetiapine for agitation or psychosis in patients with dementia and parkinsonism. Neurology. 2007;68(17):1356–1363.
  • Pollak P, Tison F, Rascol O, et al. Clozapine in drug induced psychosis in Parkinson’s disease: a randomized, placebo controlled study with open follow up. J Neurol Neurosurg Psychiatry. 2004;75:689–695.
  • Cummings JL, Street J, Masterman D, et al. Efficacy of olanzapine in the treatment of psychosis in dementia with Lewy bodies. Dement Geriatr Cogn Disord. 2002;13:67–73.
  • Teng PR, Yeh CH, Lin CY, et al. Olanzapine-induced neuroleptic malignant syndrome in a patient with probable dementia with Lewy bodies. J Neuropsychiatry Clin Neurosci. 2012;24(4):E1–2.
  • Culo S, Mulsant BH, Rosen J, et al. Treating neuropsychiatric symptoms in dementia with Lewy bodies: a randomized controlled trial. Alzheimer Dis Assoc Disord. 2010;24:360–364.
  • Pollock BG, Mulsant BH, Rosen J, et al. A double-blind comparison of citalopram and risperidone for the treatment of behavioral and psychotic symptoms associated with dementia. Am J Geriatr Psychiatry. 2007;15:942–952.
  • Lucetti C, Logi C, Del Dotto P, et al. Levodopa response in dementia with Lewy bodies: a 1-year follow-up study. Parkinsonism Relat Disord. 2010;16:522–526.
  • Goldman JG, Goetz CG, Brandabur M, et al. Effects of dopaminergic medications on psychosis and motor function in dementia with Lewy bodies. Mov Disord. 2008;23:2248–2250.
  • Molloy S, McKeith IG, O’Brien JT, et al. The role of levodopa in the management of dementia with Lewy bodies. J Neurol Neurosurg Psychiatry. 2005;76(9):1200–1203.
  • Boeve BF, Silber MH, Ferman TJ. Melatonin for treatment of REM sleep behavior disorder in neurologic disorders: results in 14 patients. Sleep Med. 2003;4(4):281–284.
  • Aurora RN, Zak RS, Maganti RK, et al. Best practice guide for the treatment of REM sleep behavior disorder (RBD). J Clin Sleep Med. 2010;6(1):85–95.
  • Lapid MI, Kuntz KM, Mason SS, et al. Efficacy, safety, and tolerability of armodafinil therapy for hypersomnia associated with dementia with Lewy bodies: a pilot study. Dement Geriatr Cogn Disord. 2017;43(5–6):269–280.
  • Prado E, Paholpak P, Ngo M, et al. Agitation and psychosis associated with dementia with Lewy bodies exacerbated by modafinil use. Am J Alzheimers Dis Other Demen. 2012;27(7):468–473.
  • Merck [Internet]. Press release: Merck’s BELSOMRA (suvorexant) C-IV meets primary efficacy endpoint in phase 3 trial for the treatment of insomnia in people with mild-to-moderate Alzheimer’s disease dementia; May 7, 2019 [ cited 2019 Jul 21]. Available from: https://investors.merck.com/news/press-release-details/2019/Mercks-BELSOMRA-suvorexant-C-IV-Meets-Primary-Efficacy-Endpoint-in-Phase-3-Trial-for-the-Treatment-of-Insomnia-in-People-with-Mild-to-Moderate-Alzheimers-Disease-Dementia/default.aspx
  • Janto K, Prichard JR, Pusalavidyasagar S. An update on dual orexin receptor antagonists and their potential role in insomnia therapeutics. J Clin Sleep Med. 2018;14(8):1399–1408.
  • Mendoza-Velasquez J, Flores-Vazquez J, Barron-Velazquez E, et al. Autonomic dysfunction in α-synucleinopathies. Front Neurol. 2019;10:363.
  • Khoury R, Grysman N, Gold J, et al. The role of 5HT-6 receptor antagonists in Alzheimer’s disease: an update. Expert Opin Investig Drugs. 2018;27(6):523–533.
  • McKeith I, Aarsland D, Friedhoff L, et al. HEADWAY-DLB: a multinational study evaluating the safety and efficacy of intepirdine (RVT-101) in dementia with Lewy bodies. Alzheimers Demen. 2017;13(7):P936.
  • Axovant Sciences Ltd [Internet]. Press release: Axovant announces negative results for interpirdine in phase 2b HEADWAY and pilot phase 2 gait and balance studies; positive trends in efficacy seen in pilot phase 2 nelotanserin study; January 8, 2018 [ cited 2019 Jul 21]. Available from: http://investors.axovant.com/news-releases/news-release-details/axovant-announces-negative-results-intepirdine-phase-2b-headway
  • Al-Shamma H, Anderson C, Chuang E, et al. Nelotanserin, a novel selective human 5-hydroxytryptamine2A inverse agonists for the treatment of insomnia. J Pharmacol Exp Ther. 2010;332(1):281–290.
  • Ballanger B, Strafella AP, van Eimeren T, et al. Serotonin 2A receptors and visual hallucinations in Parkinson disease. Arch Neurol. 2010;67(4):416–421.
  • Axovant Sciences Ltd [Internet]. Press release: CORRECTION: Axovant announces negative results for interpirdine in phase 2b HEADWAY and pilot phase 2 gait and balance studies; positive trends in efficacy seen in pilot phase 2 nelotanserin study; January 9, 2018 [ cited 2019 Jul 21]. Available from: http://investors.axovant.com/news-releases/news-release-details/correction-axovant-announces-negative-results-intepirdine-phase
  • Axovant Sciences Ltd [Internet]. Press release: Axovant reports results of exploratory phase 2 clinical study of nelotanserin in Lewy body dementia patients experiencing REM sleep behavior disorder; December 10, 2018 [ cited 2019 Jul 21]. Available from: http://investors.axovant.com/news-releases/news-release-details/axovant-reports-results-exploratory-phase-2-clinical-study
  • Sosei Heptares [Internet]. Press release: Sosei provided update on HTL0018318; September 18, 2018 [ cited 2019 Jul 21]. Available from: https://soseiheptares.com/news/109/129/Sosei-Provides-Update-on-HTL0018318
  • Salomoni P, Calabretta B. Tageted therapies and autophagy: new insights from chronic myeloid leukemia. Autophagy. 2009;5:1050–1051.
  • Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013;19:983–997.
  • Lonskaya I, Hebron ML, Desforges NM, et al. Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5:1247–1262.
  • Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improve motor behavior via autophagic degradation of alpha-synuclein in Parkinson’s disease models. Hum Mol Genet. 2013;22:3315–3328.
  • Pagan F, Hebron M, Valadez E, et al. Nilotinib effects in Parkinson’s disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503–517.
  • Goldfine A, Faulkner R, Sadashivam V, et al. Results of a phase 1 dose-ranging trial, and design of a phase 2 trial, of K0706, a novel C-Abl tyrosine kinase inhibitor for Parkinson’s disease. Neurology. 2019;92(15):P2.8–047.
  • Tong L, Prieto GA, Kramar EA, et al. Brain-derived neurotrophic factor-dependent synaptic plasticity is suppressed by interleukin-1beta via p38 mitogen-activated protein kinase. J Neurosci. 2012;32:17714–17724.
  • Scheltens P, Prins N, Lammertsma A, et al. An exploratory clinical study of p38α kinase inhibition in Alzheimer’s disease. Ann Clin Transl Neurol. 2018;5(4):464–473.
  • Alam J, Blackburn K, Patrick D. Neflamapimod: clinical phase 2b-ready oral small molecule inhibitor of p38α to reverse synaptic dysfunction in early Alzheimer’s disease. J Prev Alzheimers Dis. 2017;4(4):273–278.
  • NUPLAZID (pimavanserin) [prescribing information]. San Diego, CA: ACADIA Pharmaceuticals; 2016.
  • Hawkins T, Berman B. Pimavanserin: a novel therapeutic option for Parkinson disease psychosis. Neurol Clin Pract. 2017;7:157–162.
  • Cummings J, Isaacson S, Mills R, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomized, placebo-controlled phase 3 trial. Lancet. 2014;383(9916):533–540.
  • Kitten AK, Hallowell SA, Saklad SR, et al. Pimavanserin: a novel drug approved to treat Parkinson’s disease psychosis. Innov Clin Neurosci. 2018;15(1–2):16–22.
  • Witkin JM, Marek GJ, Johnson BG, et al. Metabotropic glutamate receptors in the control of mood disorders. CNS Neurol Disord Drug Targets. 2007;6(2):87–100.
  • Blacker CJ, Lewis CP, Frye AM, et al. Metabotropic glutamate receptors as emerging research targets in bipolar disorder. Psychiatry Res. 2017;257:327–337.
  • Maksymetz J, Moran SP, Conn PJ. Targeting metabotropic glutamate receptors for novel treatments of schizophrenia. Mol Brain. 2017;10(1):15.
  • Crupi R, Impellizzeri D, Cuzzocrea S. Role of metabotropic glutamate receptors in neurological disorders. Front Mol Neurosci. 2019;8:12.
  • Medina A. Therapeutic utility of phosphodiesterase type I inhibitors in neurological conditions. Front Neurosci. 2011;5:21.
  • Ando M, Kotani S, Watanabe N. Preclinical charaterization of E2027, a novel phosphodiesterase 9 inhibitor. Alzheimers Demen. 2017;13(7):P946.
  • Svensson K, Ardayfio P, Kielbasa W, et al. A single ascending dose (SAD) study of a D1 receptor positive allosteric modulator (LY3154207) in healthy volunteers. Neurology. 2019;92(15):P4.7–004.
  • Arnsten AFT, Girgis RR, Gray DL, et al. Novel dopamine therapeutics for cognitive deficits in schizophrenia. Biol Psychiatry. 2017;81(1):67–77.
  • Wesnes KA. The value of assessing cognitive function in drug development. Dialogues Clin Neurosci. 2000;2(3):183–202.
  • Murata M, Odawara T, Hasegawa K, et al. Adjunct zonisamide to levodopa for DLB parkinsonism. Neurology. 2018;90:e664–e672.
  • Murata M, Hasegawa K, Kanazawa I, et al. Zonisamide improves wearing-off in Parkinson’s disease: a randomized, double-blind study. Mov Disord. 2015;30:1343–1350.
  • Odawara T, Shiozaki K, Togo T, et al. Administration of zonisamide in three cases of dementia with Lewy bodies. Psychiatry Clin Neurosci. 2010;64:327–329.
  • Bergstrom A, Kallunki P, Fog K. Development of passive immunotherapies for synucleinopathies. Mov Disord. 2016;31(2):201–213.
  • Lindstrom V, Ihse E, Fagerqvist T, et al. Immunotherapy targeting α-synuclein with relevance for future treatment of Parkinson’s disease and other Lewy body disorders. Immunotherapy. 2014;6(2):141–153.
  • Mandler M, Valera E, Rockenstein E, et al. Next-generation active immunization approach for synucleinopathies: implications for Parkinson’s disease clinical trials. Acta Neuropathol. 2014;127(6):861–879.
  • Goldberg N, Caesar J, Park A, et al. Neural stem cells rescue cognitive and motor dysfunction in a transgenic model of dementia with Lewy bodies through a BDNF-dependent mechanism. Stem Cell Reports. 2015;5:791–804.
  • Price DL, Koike M, Khan A, et al. The small molecule alpha-synuclein misfolding inhibitor, NPT200-11, produces multiple benefits in an animal model of Parkinson’s disease. Sci Rep. 2018;8(1):161–165.
  • McNeill A, Magalhaes J, Shen C, et al. Ambroxol improves lysosomal biochemistry in glucocerebrocidase mutation-linked Parkinson disease cells. Brain. 2014;137(5):1481–1495.
  • Cumming J. The role of biomarkers in Alzheimer’s disease drug development. Adv Exp Med Biol. 2019;1118:29–61.
  • Walker MP, Ayre GA, Cummings JL, et al. Quantifying fluctuation in dementia with Lewy bodies, Alzheimer’s disease, and vascular dementia. Neurology. 2000;54(8):1616–1625.