A Phase II Study to Evaluate the Safety and Efficacy of Prasinezumab in Early Parkinson's Disease (PASADENA): Rationale, Design, and Baseline Data

Gennaro Pagano, Frank G Boess, Kirsten I Taylor, Benedicte Ricci, Brit Mollenhauer, Werner Poewe, Anne Boulay, Judith Anzures-Cabrera, Annamarie Vogt, Maddalena Marchesi, Anke Post, Tania Nikolcheva, Gene G Kinney, Wagner M Zago, Daniel K Ness, Hanno Svoboda, Markus Britschgi, Susanne Ostrowitzki, Tanya Simuni, Kenneth Marek, Martin Koller, Jeff Sevigny, Rachelle Doody, Paulo Fontoura, Daniel Umbricht, Azad Bonni, PASADENA Investigators, Prasinezumab Study Group

Abstract

Background: Currently available treatments for Parkinson's disease (PD) do not slow clinical progression nor target alpha-synuclein, a key protein associated with the disease. Objective: The study objective was to evaluate the efficacy and safety of prasinezumab, a humanized monoclonal antibody that binds aggregated alpha-synuclein, in individuals with early PD. Methods: The PASADENA study is a multicenter, randomized, double-blind, placebo-controlled treatment study. Individuals with early PD, recruited across the US and Europe, received monthly intravenous doses of prasinezumab (1,500 or 4,500 mg) or placebo for a 52-week period (Part 1), followed by a 52-week extension (Part 2) in which all participants received active treatment. Key inclusion criteria were: aged 40-80 years; Hoehn & Yahr (H&Y) Stage I or II; time from diagnosis ≤2 years; having bradykinesia plus one other cardinal sign of PD (e.g., resting tremor, rigidity); DAT-SPECT imaging consistent with PD; and either treatment naïve or on a stable monoamine oxidase B (MAO-B) inhibitor dose. Study design assumptions for sample size and study duration were built using a patient cohort from the Parkinson's Progression Marker Initiative (PPMI). In this report, baseline characteristics are compared between the treatment-naïve and MAO-B inhibitor-treated PASADENA cohorts and between the PASADENA and PPMI populations. Results: Of the 443 patients screened, 316 were enrolled into the PASADENA study between June 2017 and November 2018, with an average age of 59.9 years and 67.4% being male. Mean time from diagnosis at baseline was 10.11 months, with 75.3% in H&Y Stage II. Baseline motor and non-motor symptoms (assessed using Movement Disorder Society-Unified Parkinson's Disease Rating Scale [MDS-UPDRS]) were similar in severity between the MAO-B inhibitor-treated and treatment-naïve PASADENA cohorts (MDS-UPDRS sum of Parts I + II + III [standard deviation (SD)]; 30.21 [11.96], 32.10 [13.20], respectively). The overall PASADENA population (63.6% treatment naïve and 36.4% on MAO-B inhibitor) showed a similar severity in MDS-UPDRS scores (e.g., MDS-UPDRS sum of Parts I + II + III [SD]; 31.41 [12.78], 32.63 [13.04], respectively) to the PPMI cohort (all treatment naïve). Conclusions: The PASADENA study population is suitable to investigate the potential of prasinezumab to slow disease progression in individuals with early PD. Trial Registration: NCT03100149.

Keywords: MDS-UPDRS = Movement Disorder Society—Unified Parkinson's Disease Rating Scale; Parkinson's disease; Phase II clinical trial; alpha-synuclein (α-syn); disease modification treatments; disease progression; monoclonal antibodies; prasinezumab.

Conflict of interest statement

GP, FB, KT, BR, JA-C, AV, MM, TN, HS, MB, SO, RD, DU, and ABon are employees of F. Hoffmann-La Roche Ltd. ABou is an employee of Idorsia Pharmaceuticals Ltd. AP is an employee of Feetme SAS. JS is an employee of Prevail Therapeutics. GK, WZ, DN, and MK are employees of Prothena Biosciences Inc. The authors declare that F. Hoffmann-La Roche Ltd was the sponsor of the study. F. Hoffmann-La Roche Ltd and Prothena Corporation plc were the funders of the study. F. Hoffmann-La Roche Ltd and Prothena Corporation plc were involved in the study design, collection, analysis, interpretation of data, the writing of this article and the decision to submit it for publication. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Pagano, Boess, Taylor, Ricci, Mollenhauer, Poewe, Boulay, Anzures-Cabrera, Vogt, Marchesi, Post, Nikolcheva, Kinney, Zago, Ness, Svoboda, Britschgi, Ostrowitzki, Simuni, Marek, Koller, Sevigny, Doody, Fontoura, Umbricht, Bonni and PASADENA Investigators and Prasinezumab Study Group.

Figures

Figure 1
Figure 1
PASADENA study design schematic. *Low dose = 1,500 mg, †High dose = 4,500 mg for ≥65 kg; 3,500 mg for <65 kg. DaT-SPECT, dopamine transporter imaging with single-photon emission computerized tomography; IV, intravenous; Q4W, every month.
Figure 2
Figure 2
Schedule of activities in PASADENA Part 1. *Parkinson's Disease Questionnaire−39 (PDQ39) is at baseline, Week 20, and Week 48; †SCOPA-AUT (Scales for Outcomes in Parkinson's Disease—Autonomic Dysfunction) is at baseline, Weeks 16, 28, 40 and 52. ‡Digital includes PASADENA Digital Motor Score, Patient Global Impression of Severity, Daily diary, Patient Assessment of Constipation Symptoms (PAC-SYM), EuroQol-5D (EQ-5D) and Hospital Anxiety and Depression Scale (HADS). §Magnetic resonance imaging (MRI) includes safety, diffusion tensor imaging (DTI), resting state and arterial spin labeling (ASL). CGI-C, Clinical Global Impression of Change; CGI-I, Clinical Global Impression of Improvement; DaT-SPECT, dopamine transporter imaging with single-photon emission computerized tomography; H&Y, Hoehn & Yahr; MDS-UPDRS, Movement Disorder Society—Unified Parkinson's Disease Rating Scale; MMSE, Mini Mental State Examination; MoCA, Montreal Cognitive Assessment; PDSS-2, Parkinson's Disease Sleep Scale Revised Version 2; PGI-C, Patient Global Impression of Change; RBDSQ, Rapid Eye Movement Sleep Behavior Disorder Screening Questionnaire; SE-ADL, Schwab and England Activities of Daily Living.
Figure 3
Figure 3
Schedule of activities in PASADENA Part 2. *Digital includes PASADENA Digital Motor Score, Patient Global Impression of Severity, Daily diary, Patient Assessment of Constipation Symptoms (PAC-SYM), EuroQol-5D (EQ-5D), and Hospital Anxiety and Depression Scale (HADS). †Magnetic resonance imaging (MRI) includes safety, diffusion tensor imaging (DTI), resting state and arterial spin labeling (ASL). CGI-I, Clinical Global Impression of Improvement; DaT-SPECT, dopamine transporter imaging with single-photon emission computerized tomography; MDS-UPDRS, Movement Disorder Society—Unified Parkinson's Disease Rating Scale; MoCA, Montreal Cognitive Assessment; PDQ39, Parkinson's Disease Questionnaire−39; PDSS-2, Parkinson's Disease Sleep Scale Revised Version 2; PGI-C, Patient Global Impression of Change; SCOPA-AUT, Scales for Outcomes in Parkinson's Disease—Autonomic Dysfunction; SE-ADL, Schwab and England Activities of Daily Living.
Figure 4
Figure 4
Table of digital measures included in the Roche Parkinson's Disease Mobile Application v2. SDMT, Symbol Digit Modalities Test.

References

    1. Verschuur CVM, Suwijn SR, Boel JA, Post B, Bloem BR, van Hilten JJ, et al. . Randomized delayed-start trial of levodopa in Parkinson's disease. New Engl J Med. (2019) 380:315–24. 10.1056/NEJMoa1809983
    1. Lang AE, Espay AJ. Disease modification in Parkinson's disease: current approaches, challenges, and future considerations. Mov Disord. (2018) 33:660–77. 10.1002/mds.27360
    1. Cheng HC, Ulane CM, Burke RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol. (2010) 67:715–25. 10.1002/ana.21995
    1. Jankovic J, Aguilar LG. Current approaches to the treatment of Parkinson's disease. Neuropsychiatr Dis Treat. (2008) 4:743–57. 10.2147/NDT.S2006
    1. Bonnet AM. Involvement of non-dopaminergic pathways in Parkinson's disease. CNS Drugs. (2000) 13:351–64. 10.2165/00023210-200013050-00005
    1. Giguere N, Burke Nanni S, Trudeau LE. On cell loss and selective vulnerability of neuronal populations in Parkinson's disease. Front Neurol. (2018) 9:455. 10.3389/fneur.2018.00455
    1. Lees AJ. The on-off phenomenon. J Neurol Neurosurg Psychiatry. (1989) 52(Suppl.) 29–37. 10.1136/jnnp.52.Suppl.29
    1. Jenner P. Treatment of the later stages of Parkinson's disease - pharmacological approaches now and in the future. Transl Neurodegener. (2015) 4:3. 10.1186/2047-9158-4-3
    1. Martinez-Martin P, Rodriguez-Blazquez C, Forjaz MJ. Quality of life and burden in caregivers for patients with Parkinson's disease: concepts, assessment and related factors. Expert Rev Pharmacoeconomics Outcomes Res. (2012) 12:221–30. 10.1586/erp.11.106
    1. Seppi K, Ray Chaudhuri K, Coelho M, Fox SH, Katzenschlager R, Perez Lloret S, et al. . Update on treatments for nonmotor symptoms of Parkinson's disease-an evidence-based medicine review. Mov Disord. (2019) 34:180–98. 10.1002/mds.27602
    1. Jankovic J, Goodman I, Safirstein B, Marmon TK, Schenk DB, Koller M, et al. . Safety and tolerability of multiple ascending doses of PRX002/RG7935, an anti-alpha-synuclein monoclonal antibody, in patients with Parkinson disease: a randomized clinical trial. JAMA Neurol. (2018) 75:1206–14. 10.1001/jamaneurol.2018.1487
    1. Masliah E, Rockenstein E, Mante M, Crews L, Spencer B, Adame A, et al. . Passive immunization reduces behavioral and neuropathological deficits in an alpha-synuclein transgenic model of Lewy body disease. PLoS ONE. (2011) 6:e19338. 10.1371/journal.pone.0019338
    1. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, et al. . Parkinson disease. Nat Rev Dis Prim. (2017) 3:17013. 10.1038/nrdp.2017.13
    1. Beach TG, Adler CH, Lue L, Sue LI, Bachalakuri J, Henry-Watson J, et al. . Unified staging system for Lewy body disorders: correlation with nigrostriatal degeneration, cognitive impairment and motor dysfunction. Acta Neuropathol. (2009) 117:613–34. 10.1007/s00401-009-0538-8
    1. Del Tredici K, Braak H. Review: Sporadic Parkinson's disease: development and distribution of alpha-synuclein pathology. Neuropathol Appl Neurobiol. (2016) 42:33–50. 10.1111/nan.12298
    1. Shahmoradian SH, Lewis AJ, Genoud C, Hench J, Moors TE, Navarro PP, et al. . Lewy pathology in Parkinson's disease consists of crowded organelles and lipid membranes. Nat Neurosci. (2019) 22:1099–109. 10.1038/s41593-019-0423-2
    1. Moors TE, Maat CA, Niedieker D, Mona D, Petersen D, Timmermans-Huisman E, et al. . The subcellular arrangement of alpha-synuclein proteoforms in the Parkinson's disease brain as revealed by multicolor STED microscopy. Acta Neuropathol. (2021) 142:423–48. 10.1007/s00401-021-02329-9
    1. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. (1997) 388:839–40. 10.1038/42166
    1. Mahul-Mellier AL, Burtscher J, Maharjan N, Weerens L, Croisier M, Kuttler F, et al. . The process of Lewy body formation, rather than simply alpha-synuclein fibrillization, is one of the major drivers of neurodegeneration. Proc Natl Acad Sci USA. (2020) 117:4971–82. 10.1073/pnas.1913904117
    1. Rey NL, Steiner JA, Maroof N, Luk KC, Madaj Z, Trojanowski JQ, et al. . Widespread transneuronal propagation of α-synucleinopathy triggered in olfactory bulb mimics prodromal Parkinson's disease. J Exp Med. (2016) 13:1759–78. 10.1084/jem.20160368
    1. Luk KC, Kehm V, Carroll J, Zhang B, O'Brien P, Trojanowski JQ, et al. . Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science. (2012) 338:949–53. 10.1126/science.1227157
    1. Games D, Seubert P, Rockenstein E, Patrick C, Trejo M, Ubhi K, et al. . Axonopathy in an alpha-synuclein transgenic model of Lewy body disease is associated with extensive accumulation of C-terminal-truncated alpha-synuclein. Am J Pathol. (2013) 182:940–53. 10.1016/j.ajpath.2012.11.018
    1. Hansen C, Angot E, Bergstrom AL, Steiner JA, Pieri L, Paul G, et al. . α-synuclein propagates from mouse brain to grafted dopaminergic neurons and seeds aggregation in cultured human cells. J Clin Investig. (2011) 121:715–25. 10.1172/JCI43366
    1. Desplats P, Lee HJ, Bae EJ, Patrick C, Rockenstein E, Crews L, et al. . Inclusion formation and neuronal cell death through neuron-to-neuron transmission of alpha-synuclein. Proc Natl Acad Sci USA. (2009) 106:13010–5. 10.1073/pnas.0903691106
    1. Luk KC, Kehm VM, Zhang B, O'Brien P, Trojanowski JQ, Lee VM. Intracerebral inoculation of pathological alpha-synuclein initiates a rapidly progressive neurodegenerative alpha-synucleinopathy in mice. J Exp Med. (2012) 209:975–86. 10.1084/jem.20112457
    1. Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, et al. . Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron. (2011) 72:57–71. 10.1016/j.neuron.2011.08.033
    1. Kim S, Kwon SH, Kam TI, Panicker N, Karuppagounder SS, Lee S, et al. . Transneuronal propagation of pathologic alpha-synuclein from the gut to the brain models Parkinson's disease. Neuron. (2019) 103:627–41 e7. 10.1016/j.neuron.2019.05.035
    1. Rey NL, Bousset L, George S, Madaj Z, Meyerdirk L, Schulz E, et al. . alpha-Synuclein conformational strains spread, seed and target neuronal cells differentially after injection into the olfactory bulb. Acta Neuropathol Commun. (2019) 7:221. 10.1186/s40478-019-0859-3
    1. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, et al. . Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science. (1997) 276:2045–7. 10.1126/science.276.5321.2045
    1. Singleton AB, Farrer M, Johnson J, Singleton A, Hague S, Kachergus J, et al. . Alpha-synuclein locus triplication causes Parkinson's disease. Science. (2003) 302:841. 10.1126/science.1090278
    1. Ibanez P, Bonnet AM, Debarges B, Lohmann E, Tison F, Pollak P, et al. . Causal relation between alpha-synuclein gene duplication and familial Parkinson's disease. Lancet. (2004) 364:1169–71. 10.1016/S0140-6736(04)17104-3
    1. Kay DM, Factor SA, Samii A, Higgins DS, Griffith A, Roberts JW, et al. . Genetic association between alpha-synuclein and idiopathic Parkinson's disease. Am J Med Genet Part B Neuropsychiatric Genet. (2008) 147B:1222–30. 10.1002/ajmg.b.30758
    1. Bekris LM, Mata IF, Zabetian CP. The genetics of Parkinson disease. J Geriatr Psychiatry Neurol. (2010) 23:228–42. 10.1177/0891988710383572
    1. Devine MJ, Gwinn K, Singleton A, Hardy J. Parkinson's disease and alpha-synuclein expression. Mov Dis. (2011) 26:2160–8. 10.1002/mds.23948
    1. Li JY, Englund E, Holton JL, Soulet D, Hagell P, Lees AJ, et al. . Lewy bodies in grafted neurons in subjects with Parkinson's disease suggest host-to-graft disease propagation. Nat Med. (2008) 14:501–3. 10.1038/nm1746
    1. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW. Lewy body-like pathology in long-term embryonic nigral transplants in Parkinson's disease. Nat Med. (2008) 14:504–6. 10.1038/nm1747
    1. Wilson H, Dervenoulas G, Pagano G, Koros C, Yousaf T, Picillo M, et al. . Serotonergic pathology and disease burden in the premotor and motor phase of A53T alpha-synuclein parkinsonism: a cross-sectional study. Lancet Neurol. (2019) 18:748–59. 10.1016/S1474-4422(19)30140-1
    1. Knudsen K, Fedorova TD, Hansen AK, Sommerauer M, Otto M, Svendsen KB, et al. . In-vivo staging of pathology in REM sleep behaviour disorder: a multimodality imaging case-control study. Lancet Neurol. (2018) 17:618–28. 10.1016/S1474-4422(18)30162-5
    1. Dehay B, Bourdenx M, Gorry P, Przedborski S, Vila M, Hunot S, et al. . Targeting alpha-synuclein for treatment of Parkinson's disease: mechanistic and therapeutic considerations. Lancet Neurol. (2015) 14:855–66. 10.1016/S1474-4422(15)00006-X
    1. Masliah E, Rockenstein E, Adame A, Alford M, Crews L, Hashimoto M, et al. . Effects of alpha-synuclein immunization in a mouse model of Parkinson's disease. Neuron. (2005) 46:857–68. 10.1016/j.neuron.2005.05.010
    1. Games D, Valera E, Spencer B, Rockenstein E, Mante M, Adame A, et al. . Reducing C-terminal-truncated alpha-synuclein by immunotherapy attenuates neurodegeneration and propagation in Parkinson's disease-like models. J Neurosci. (2014) 34:9441–54. 10.1523/JNEUROSCI.5314-13.2014
    1. Spencer B, Valera E, Rockenstein E, Overk C, Mante M, Adame A, et al. . Anti-alpha-synuclein immunotherapy reduces alpha-synuclein propagation in the axon and degeneration in a combined viral vector and transgenic model of synucleinopathy. Acta Neuropathol Commun. (2017) 5:7. 10.1186/s40478-016-0410-8
    1. Tran HT, Chung CH, Iba M, Zhang B, Trojanowski JQ, Luk KC, et al. . Alpha-synuclein immunotherapy blocks uptake and templated propagation of misfolded alpha-synuclein and neurodegeneration. Cell Rep. (2014) 7:2054–65. 10.1016/j.celrep.2014.05.033
    1. Kallab M, Herrera-Vaquero M, Johannesson M, Eriksson F, Sigvardson J, Poewe W, et al. . Region-specific effects of immunotherapy with antibodies targeting alpha-synuclein in a transgenic model of synucleinopathy. Front Neurosci. (2018) 12:452. 10.3389/fnins.2018.00452
    1. Schenk DB, Koller M, Ness DK, Griffith SG, Grundman M, Zago W, et al. . First-in-human assessment of PRX002, an anti-alpha-synuclein monoclonal antibody, in healthy volunteers. Mov Disord. (2017) 32:211–8. 10.1002/mds.26878
    1. PPMI . A Landmark Study of Parkinson's Disease. (2018). Available online at: (accessed May 2021).
    1. Simuni T, Siderowf A, Lasch S, Coffey CS, Caspell-Garcia C, Jennings D, et al. . Longitudinal change of clinical and biological measures in early Parkinson's disease: Parkinson's progression markers initiative cohort. Mov Disord. (2018) 33:771–82. 10.1002/mds.27361
    1. . NCT03100149: A Study to Evaluate the Efficacy of Prasinezumab (RO7046015/PRX002) in Participants with Early Parkinson's Disease (PASADENA). (2017). Available online at: (accessed May 2021).
    1. Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, et al. . Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord. (2008) 23:2129–70. 10.1002/mds.22340
    1. Taylor KI, Staunton H, Lipsmeier F, Nobbs D, Lindemann M. Outcome measures based on digital health technology sensor data: data- and patient-centric approaches. NPJ Digit Med. (2020) 3:97. 10.1038/s41746-020-0305-8
    1. Lipsmeier F, Simillion C, Bamdadian A, Smith A, Schobel S, Gossens C, et al. . Reliability, feasibility and validity of a novel digital monitoring platform assessing cognitive and motor symptoms in people with Stage I and II Huntington's disease (HD). Neurotherapeutics. (2019) 16:1350–90. 10.1007/s13311-019-00788-3
    1. Marek K, Chowdhury S, Siderowf A, Coffey CS, Caspell-Garcia CJ, Simuni T, et al. . The Parkinson's progression markers initiative (PPMI) - establishing a PD biomarker cohort. Ann Clin Transl Neurol. (2018) 5:1460–77. 10.1002/acn3.644
    1. Horvath K, Aschermann Z, Kovacs M, Makkos A, Harmat M, Janszky J, et al. . Minimal clinically important differences for the experiences of daily living parts of movement disorder society-sponsored unified Parkinson's disease rating scale. Mov Disord. (2017) 32:789–93. 10.1002/mds.26960
    1. Parkinson Progression Marker Initiative . The Parkinson progression marker initiative (PPMI). Prog Neurobiol. (2011) 95:629–35. 10.1016/j.pneurobio.2011.09.005
    1. Stuart EA, Lee BK, Leacy FP. Prognostic score-based balance measures can be a useful diagnostic for propensity score methods in comparative effectiveness research. J Clin Epidemiol. (2013) 66(Suppl. 8):S84– 90.e1. 10.1016/j.jclinepi.2013.01.013
    1. Rascol O, Fitzer-Attas CJ, Hauser R, Jankovic J, Lang A, Langston JW, et al. . A double-blind, delayed-start trial of rasagiline in Parkinson's disease (the ADAGIO study): prespecified and post-hoc analyses of the need for additional therapies, changes in UPDRS scores, and non-motor outcomes. Lancet Neurol. (2011) 10:415–23. 10.1016/S1474-4422(11)70073-4
    1. Parkinson Study Group . A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol. (2002) 59:1937–43. 10.1001/archneur.59.12.1937
    1. Kalia LV, Kalia SK, Lang AE. Disease-modifying strategies for Parkinson's disease. Mov Disord. (2015) 30:1442–50. 10.1002/mds.26354
    1. Mollenhauer B, Trautmann E, Sixel-Doring F, Wicke T, Ebentheuer J, Schaumburg M, et al. . Nonmotor and diagnostic findings in subjects with de novo Parkinson disease of the DeNoPa cohort. Neurology. (2013) 81:1226–34. 10.1212/WNL.0b013e3182a6cbd5
    1. A controlled trial of lazabemide (Ro 19-6327) in levodopa-treated Parkinson's disease . Parkinson Study Group. Arch Neurol. (1994) 51:342–7. 10.1001/archneur.1994.00540160036006
    1. Teo KC, Ho SL. Monoamine oxidase-B (MAO-B) inhibitors: implications for disease-modification in Parkinson's disease. Transl Neurodegener. (2013) 2:19. 10.1186/2047-9158-2-19
    1. Qamar MA, Sauerbier A, Politis M, Carr H, Loehrer P, Chaudhuri KR. Presynaptic dopaminergic terminal imaging and non-motor symptoms assessment of Parkinson's disease: evidence for dopaminergic basis? NPJ Parkinsons Dis. (2017) 3:5. 10.1038/s41531-016-0006-9
    1. Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA. (2014) 311:1670–83. 10.1001/jama.2014.3654
    1. Holden SK, Finseth T, Sillau SH, Berman BD. Progression of MDS-UPDRS scores over five years in de novo Parkinson disease from the Parkinson's progression markers initiative cohort. Mov Disord Clin Pract. (2018) 5:47–53. 10.1002/mdc3.12553
    1. Pagano G, Niccolini F, Politis M. Imaging in Parkinson's disease. Clin Med. (2016) 16:371–5. 10.7861/clinmedicine.16-4-371
    1. Nandhagopal R, Kuramoto L, Schulzer M, Mak E, Cragg J, McKenzie J, et al. . Longitudinal evolution of compensatory changes in striatal dopamine processing in Parkinson's disease. Brain. (2011) 134(Pt 11):3290–8. 10.1093/brain/awr233
    1. Schwarz J, Storch A, Koch W, Pogarell O, Radau PE, Tatsch K. Loss of dopamine transporter binding in Parkinson's disease follows a single exponential rather than linear decline. J Nucl Med. (2004) 45:1694–7.
    1. Maetzler W, Domingos J, Srulijes K, Ferreira JJ, Bloem BR. Quantitative wearable sensors for objective assessment of Parkinson's disease. Mov Disord. (2013) 28:1628–37. 10.1002/mds.25628
    1. Ossig C, Antonini A, Buhmann C, Classen J, Csoti I, Falkenburger B, et al. . Wearable sensor-based objective assessment of motor symptoms in Parkinson's disease. J Neural Transm. (2016) 123:57–64. 10.1007/s00702-015-1439-8
    1. Lipsmeier F, Fernandez Garcia I, Wolf D, Kilchenmann T, Scotland A, Schjodt-Eriksen J, et al. . Successful Passive Monitoring of Early-Stage Parkinson's Disease Patient Mobility in Phase I RG7935/PRX002 Clinical Trial with Smartphone Sensors. Basel: MDS; (2017).
    1. Lipsmeier F, Taylor KI, Kilchenmann T, Wolf D, Scotland A, Schjodt-Eriksen J, et al. . Evaluation of smartphone-based testing to generate exploratory outcome measures in a phase 1 Parkinson's disease clinical trial. Mov Disord. (2018) 33:1287–97. 10.1002/mds.27376
    1. Lipsmeier F, Taylor KI, Postuma R, Kilchenmann T, Wolf D, Zhang Y, et al. . Preliminary validation of a novel, comprehensive digital biomarker smartphone application to assess motor symptoms in recently diagnosed Parkinson patients (P4.7-005). Neurology. (2019) 92.
    1. Jankovic J. Parkinson's disease: clinical features and diagnosis. J Neurol Neurosurg Psychiatry. (2008) 79:368–76. 10.1136/jnnp.2007.131045
    1. Wang N, Gibbons CH, Lafo J, Freeman R. Alpha-synuclein in cutaneous autonomic nerves. Neurology. (2013) 81:1604–10. 10.1212/WNL.0b013e3182a9f449
    1. Donadio V, Incensi A, Leta V, Giannoccaro MP, Scaglione C, Martinelli P, et al. . Skin nerve alpha-synuclein deposits: a biomarker for idiopathic Parkinson disease. Neurology. (2014) 82:1362–9. 10.1212/WNL.0000000000000316
    1. Horvath K, Aschermann Z, Acs P, Deli G, Janszky J, Komoly S, et al. . Minimal clinically important difference on the Motor Examination part of MDS-UPDRS. Parkinsonism Relat Disord. (2015) 21:1421–6. 10.1016/j.parkreldis.2015.10.006
    1. Mischley LK, Lau RC, Weiss NS. Use of a self-rating scale of the nature and severity of symptoms in Parkinson's Disease (PRO-PD): Correlation with quality of life and existing scales of disease severity. NPJ Parkinsons Dis. (2017) 3:20. 10.1038/s41531-017-0021-5
    1. Poewe W, Espay AJ. Long duration response in Parkinson's disease: levodopa revisited. Brain J Neurol. (2020) 143:2332–5. 10.1093/brain/awaa226
    1. Hustad E, Aasly JO. Clinical and imaging markers of prodromal Parkinson's disease. Front Neurol. (2020) 11:395. 10.3389/fneur.2020.00395
    1. Campbell CM, Gilron I, Doshi T, Raja S. Designing and conducting proof-of-concept chronic pain analgesic clinical trials. Pain Rep. (2019) 4:e697. 10.1097/PR9.0000000000000697

Source: PubMed

3
Suscribir