Skip to main content Accessibility help
×
Hostname: page-component-7c8c6479df-7qhmt Total loading time: 0 Render date: 2024-03-19T05:20:51.465Z Has data issue: false hasContentIssue false

Section IV - Clinical Trials in Parkinson's Diease: Lessons, Controversies and Challenges

Published online by Cambridge University Press:  05 March 2016

Néstor Gálvez-Jiménez
Affiliation:
Cleveland Clinic, Florida
Hubert H. Fernandez
Affiliation:
Cleveland Clinic, Ohio
Alberto J. Espay
Affiliation:
University of Cincinnati
Susan H. Fox
Affiliation:
Toronto Western Hospital
Get access
Type
Chapter
Information
Parkinson's Disease
Current and Future Therapeutics and Clinical Trials
, pp. 231 - 360
Publisher: Cambridge University Press
Print publication year: 2016

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Acquadro, C, Berzon, R, Dubois, D, et al. Incorporating the patient's perspective into drug development and communication: an ad hoc task force report of the Patient-Reported Outcomes (PRO) Harmonization Group meeting at the Food and Drug Administration, February 16, 2001. Value Health 2003; 6: 52231.Google Scholar
Scientific Advisory Committee of the Medical Outcomes Trust. Assessing health status and quality-of-life instruments: attributes and review criteria. Qual Life Res 2002; 11: 193205.Google Scholar
Mokkink, LB, Terwee, CB, Patrick, DL, et al. The COSMIN checklist for assessing the methodological quality of studies on measurement properties of health status measurement instruments: an international Delphi study. Qual Life Res 2010; 19: 53949.Google Scholar
Terwee, CB, Dekker, FW, Wiersinga, WM, Prummel, MF, Bossuyt, PMM. On assessing responsiveness of health-related quality of life instruments: guidelines for instrument evaluation. Qual Life Res 2003; 12: 34962.Google Scholar
Wyrwich, KW, Metz, SM, Kroenke, K, et al. Triangulating patient and clinician perspectives on clinically important differences in health-related quality of life among patients with heart disease. Health Serv Res 2007; 42: 225774.Google Scholar
Hoehn, MM, Yahr, MD. Parkinsonism: onset, progression and mortality. Neurology 1967; 17: 42742.Google Scholar
Goetz, CG, Poewe, W, Rascol, O, et al. Movement Disorder Society Task Force report on the Hoehn and Yahr staging scale: status and recommendations. Mov Disord 2004; 19: 10208.Google Scholar
Martínez-Martín, P, Forjaz, MJ, Cubo, E, Frades, B, de Pedro Cuesta, J. Global versus factor-related impression of severity in Parkinson's disease: a new clinimetric index (CISI-PD). Mov Disord 2006; 21: 20814.Google Scholar
Martínez-Martín, P, Rodríguez-Blázquez, C, Forjaz, MJ, de Pedro, J. The Clinical Impression of Severity Index for Parkinson's Disease: international validation study. Mov Disord 2009; 24: 21117.Google Scholar
Fahn, S, Elton, R, UPDRS Program Members. Unified Parkinson's Disease Rating Scale. In: Fahn, S, Marsden, C, Goldstein, M, Calne, D, eds. Recent Developments in Parkinson’s Disease. Florham Park, NJ: Macmillan Healthcare Information; 1987; 15363.Google Scholar
Movement Disorder Society Task Force on Rating Scales for Parkinson's Disease. The Unified Parkinson's Disease Rating Scale (UPDRS): status and recommendations. Mov Disord 2003; 18: 73850.Google Scholar
Steffen, T, Seney, M. Test-retest reliability and minimal detectable change on balance and ambulation tests, the 36-item Short-Form Health Survey, and the unified Parkinson disease rating scale in people with parkinsonism. Phys Ther 2008; 88: 73346.Google Scholar
Oertel, WH, Wolters, E, Sampaio, C, et al. Pergolide versus levodopa monotherapy in early Parkinson's disease patients: the PELMOPET study. Mov Disord 2006; 21: 34353.Google Scholar
Hauser, RA, Auinger, P, Parkinson Study Group. Determination of minimal clinically important change in early and advanced Parkinson's disease. Mov Disord 2011; 26: 8138.CrossRefGoogle ScholarPubMed
Goetz, CG, Fahn, S, Martinez-Martin, P, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): process, format, and clinimetric testing plan. Mov Disord 2007; 22: 417.Google Scholar
Goetz, CG, Tilley, BC, Shaftman, SR, 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: 212970.Google Scholar
Goetz, CG, Stebbins, GT, LaPelle, N, Huang, J, Tilley, BC. MDS-UPDRS non-English translation program. Mov Disord 2012; 27 (Suppl. 1): S96.Google Scholar
Poewe, W, Hauser, R, Lang, AE. Rasagiline 1 mg/day provides benefits in the progression of nonmotor symptoms in patients with early Parkinson's disease: Assessment with the revised MDS-UPDRS. Mov Disord 2009; 24 (Suppl. 1): S272.Google Scholar
Lang, AE, Eberly, S, Goetz, CG, et al. Movement Disorder Society Unified Parkinson Disease Rating Scale experiences in daily living: longitudinal changes and correlation with other assessments. Mov Disord 2013; 28: 19806.Google Scholar
Goetz, CG, Stebbins, GT, Simkus, V. Severity ranges on the MDS-UPDRS motor examination: comparison to CGI-severity scores. Mov Disord 2009; 24 (Suppl. 1): S434.Google Scholar
Stebbins, GT, Goetz, CG, Simkus, V. Minimal clinically important change and the MDS-UPDRS motor examination. Mov Disord 2009; 24 (Suppl. 1): S436.Google Scholar
Merello, M, Gerschcovich, ER, Ballesteros, D, Cerquetti, D. Correlation between the Movement Disorders Society Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and the Unified Parkinson's Disease Rating Scale (UPDRS) during l-dopa acute challenge. Parkinsonism Relat Disord 2011; 17: 7057.Google Scholar
Goetz, CG, Stebbins, GT, Tilley, BC. Calibration of Unified Parkinson's Disease Rating Scale scores to Movement Disorder Society – Unified Parkinson's Disease rating scale scores. Mov Disord 2012; 27: 123942.Google Scholar
Verbaan, D, van Rooden, SM, Benit, CP, et al. SPES/SCOPA and MDS-UPDRS: formulas for converting scores of two motor scales in Parkinson's disease. Parkinsonism Relat Disord 2011; 17: 6324.Google Scholar
Marinus, J, Visser, M, Stiggelbout, AM, et al. A short scale for the assessment of motor impairments and disabilities in Parkinson's disease: the SPES/SCOPA. J Neurol Neurosurg Psychiatry 2004; 75: 38895.Google Scholar
Martínez-Martín, P, Benito-León, J, Burguera, JA, et al. The SCOPA-Motor Scale for assessment of Parkinson's disease is a consistent and valid measure. J Clin Epidemiol 2005; 58: 6749.Google Scholar
Schwab, R, England, A. Third Symposium for Parkinson’s Disease. Edinburgh, UK: Livingstone; 1969; 1527.Google Scholar
Martinez-Martin, P, Prieto, L, Forjaz, MJ. Longitudinal metric properties of disability rating scales for Parkinson's disease. Value Health 2006; 9: 38693.Google Scholar
Schrag, A, Spottke, A, Quinn, NP, Dodel, R. Comparative responsiveness of Parkinson's disease scales to change over time. Mov Disord 2009; 24: 81318.Google Scholar
Guy, W. Abnormal involuntary movement scale. In: ECDEU Assessment Manual for Psychopharmacology. Washington, DC: US Government Printing Office; 1976; 5347.Google Scholar
Colosimo, C, Martínez-Martín, P, Fabbrini, G, et al. Task force report on scales to assess dyskinesia in Parkinson's disease: critique and recommendations. Mov Disord 2010; 25: 113142.Google Scholar
Goetz, CG, Stebbins, GT, Chung, KA, et al. Which dyskinesia scale best detects treatment response? Mov Disord 2013; 28: 3416.Google Scholar
Goetz, CG, Stebbins, GT, Shale, HM, et al. Utility of an objective dyskinesia rating scale for Parkinson's disease: inter- and intrarater reliability assessment. Mov Disord 1994; 9: 3904.Google Scholar
Goetz, CG, Stebbins, GT, Chung, KA, et al. Which dyskinesia scale best detects treatment response? Mov Disord 2013; 28: 3416.CrossRefGoogle ScholarPubMed
Goetz, CG, Nutt, JG, Stebbins, GT. The Unified Dyskinesia Rating Scale: presentation and clinimetric profile. Mov Disord 2008; 23: 2398403.Google Scholar
Colosimo, C. Dyskinesia rating scales in Parkinson's disease. In: Sampaio, C, Goetz, CG, Schrag, A, eds. Rating Scales in Parkinson’s Disease. New York: Oxford University Press; 2012; 8498.Google Scholar
Chaudhuri, KR, Martinez-Martin, P, Brown, RG, et al. The metric properties of a novel non-motor symptoms scale for Parkinson's disease: results from an international pilot study. Mov Disord 2007; 22: 190111.Google Scholar
Martinez-Martin, P, Rodriguez-Blazquez, C, Abe, K, et al. International study on the psychometric attributes of the non-motor symptoms scale in Parkinson disease. Neurology 2009; 73: 158491.Google Scholar
Reddy, P, Martinez-Martin, P, Rizos, A, et al. Intrajejunal levodopa versus conventional therapy in Parkinson disease: motor and nonmotor effects. Clin Neuropharmacol 2012; 35: 2057.Google Scholar
Chaudhuri, KR, Martinez-Martin, P, Antonini, A, et al. Rotigotine and specific non-motor symptoms of Parkinson's disease: post hoc analysis of RECOVER. Parkinsonism Relat Disord 2013; 19: 6605.Google Scholar
Marinus, J, Visser, M, Verwey, NA, et al. Assessment of cognition in Parkinson's disease. Neurology 2003; 61: 12228.Google Scholar
Carod-Artal, FJ, Martínez-Martin, P, Kummer, W, Ribeiro, LdaS. Psychometric attributes of the SCOPA-COG Brazilian version. Mov Disord 2008; 23: 817.Google Scholar
Forjaz, MJ, Frades-Payo, B, Rodriguez-Blazquez, C, Ayala, A, Martinez-Martin, P. Should the SCOPA-COG be modified? A Rasch analysis perspective. Eur J Neurol 2010; 17: 2027.CrossRefGoogle ScholarPubMed
Reuter, I, Mehnert, S, Sammer, G, Oechsner, M, Engelhardt, M. Efficacy of a multimodal cognitive rehabilitation including psychomotor and endurance training in Parkinson's disease. J Aging Res 2012; 2012: 235765.Google Scholar
Visser, M, Verbaan, D, van Rooden, SM, et al. Assessment of psychiatric complications in Parkinson's disease: the SCOPA-PC. Mov Disord 2007; 22: 22218.Google Scholar
Friedberg, G, Zoldan, J, Weizman, A, Melamed, E. Parkinson Psychosis Rating Scale: a practical instrument for grading psychosis in Parkinson's disease. Clin Neuropharmacol 1998; 21: 2804.Google Scholar
Rodriguez-Blazquez, C, Rojo-Abuin, JM, Alvarez-Sanchez, M, et al. The MDS-UPDRS Part II (motor experiences of daily living) resulted useful for assessment of disability in Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 88993.Google Scholar
Stacy, M, Bowron, A, Guttman, M, et al. Identification of motor and nonmotor wearing-off in Parkinson's disease: Comparison of a patient questionnaire versus a clinician assessment. Mov Disord 2005; 20: 72633.Google Scholar
Stacy, M, Hauser, R. Development of a Patient Questionnaire to facilitate recognition of motor and non-motor wearing-off in Parkinson's disease. J Neural Transm 2007; 114: 21117.Google Scholar
Stacy, MA, Murphy, JM, Greeley, DR, et al. The sensitivity and specificity of the 9-item Wearing-off Questionnaire. Parkinsonism Relat Disord 2008; 14: 20512.Google Scholar
Martinez-Martin, P, Tolosa, E, Hernandez, B, Badia, X, ValidQUICK Study Group. Validation of the “QUICK” questionnaire – a tool for diagnosis of “wearing-off” in patients with Parkinson's disease. Mov Disord 2008; 23: 8306.Google Scholar
Martinez-Martin, P, Hernandez, B. The Q10 questionnaire for detection of wearing-off phenomena in Parkinson's disease. Parkinsonism Relat Disord 2012; 18: 3825.Google Scholar
Antonini, A, Martinez-Martin, P, Chaudhuri, RK, et al. Wearing-off scales in Parkinson's disease: Critique and recommendations. Mov Disord 2011; 26: 216975.Google Scholar
Giladi, N, Shabtai, H, Simon, ES, et al. Construction of freezing of gait questionnaire for patients with Parkinsonism. Parkinsonism Relat Disord 2000; 6: 16570.Google Scholar
Nieuwboer, A, Rochester, L, Herman, T, et al. Reliability of the new freezing of gait questionnaire: agreement between patients with Parkinson's disease and their carers. Gait Posture. 2009; 30: 45963.Google Scholar
Katzenschlager, R, Schrag, A, Evans, A, et al. Quantifying the impact of dyskinesias in PD: the PDYS-26: a patient-based outcome measure. Neurology 2007; 69: 55563.Google Scholar
Chaudhuri, KR, Martinez-Martin, P, Schapira, AHV, et al. International multicenter pilot study of the first comprehensive self-completed nonmotor symptoms questionnaire for Parkinson's disease: the NMSQuest study. Mov Disord 2006; 21: 91623.Google Scholar
Martinez-Martin, P, Schapira, AHV, Stocchi, F, et al. Prevalence of nonmotor symptoms in Parkinson's disease in an international setting; study using nonmotor symptoms questionnaire in 545 patients. Mov Disord 2007; 22: 16239.Google Scholar
Martinez-Martin, P, Chaudhuri, KR, Rojo-Abuin, JM, et al. Assessing the non-motor symptoms of Parkinson's disease: MDS-UPDRS and NMS Scale. Eur J Neurol 2013; 22: 3743.Google Scholar
Chou, KL, Taylor, JL, Patil, PG. The MDS-UPDRS tracks motor and non-motor improvement due to subthalamic nucleus deep brain stimulation in Parkinson disease. Parkinsonism Relat Disord 2013; 19: 9669.Google Scholar
Weintraub, D, Mamikonyan, E, Papay, K, et al. Questionnaire for Impulsive-Compulsive Disorders in Parkinson's Disease – Rating Scale. Mov Disord 2012; 27: 2427.Google Scholar
Martinez-Martin, P, Frades-Payo, B, Agüera-Ortiz, L, Ayuga-Martinez, A. A short scale for evaluation of neuropsychiatric disorders in Parkinson's disease: first psychometric approach. J Neurol 2012; 259: 2299308.Google Scholar
Weintraub, D, Stewart, S, Shea, JA, et al. Validation of the Questionnaire for Impulsive-Compulsive Behaviors in Parkinson's Disease (QUIP). Mov Disord 2009; 24: 14617.Google Scholar
Chaudhuri, KR, Pal, S, DiMarco, A, et al. The Parkinson's disease sleep scale: a new instrument for assessing sleep and nocturnal disability in Parkinson's disease. J Neurol Neurosurg Psychiatry 2002; 73: 62935.Google Scholar
Marinus, J, Visser, M, van Hilten, JJ, Lammers, GJ, Stiggelbout, AM. Assessment of sleep and sleepiness in Parkinson disease. Sleep. 2003; 26: 104954.Google Scholar
Trenkwalder, C, Kohnen, R, Högl, B, et al. Parkinson's disease sleep scale – validation of the revised version PDSS-2. Mov Disord 2011; 26: 64452.Google Scholar
Högl, B, Arnulf, I, Comella, C, et al. Scales to assess sleep impairment in Parkinson's disease: critique and recommendations. Mov Disord 2010; 25: 270416.Google Scholar
Zibetti, M, Rizzone, M, Merola, A, et al. Sleep improvement with levodopa/carbidopa intestinal gel infusion in Parkinson disease. Acta Neurol Scand 2013; 127: e2832.Google Scholar
Takanashi, M, Shimo, Y, Hatano, T, Oyama, G, Hattori, N. Efficacy and safety of a once-daily extended-release formulation of pramipexole switched from an immediate-release formulation in patients with advanced Parkinson's disease: results from an open-label study. Drug Res. 2013; 63: 63943.Google Scholar
Martinez-Martin, P, Visser, M, Rodriguez-Blazquez, C, et al. SCOPA-sleep and PDSS: two scales for assessment of sleep disorder in Parkinson's disease. Mov Disord 2008; 23: 16818.Google Scholar
Chahine, LM, Daley, J, Horn, S, et al. Association between dopaminergic medications and nocturnal sleep in early-stage Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 85963.Google Scholar
Perez Lloret, S, Pirán Arce, G, Rossi, M, et al. Validation of a new scale for the evaluation of sialorrhea in patients with Parkinson's disease. Mov Disord 2007; 22: 10711.Google Scholar
Manor, Y, Giladi, N, Cohen, A, Fliss, DM, Cohen, JT. Validation of a swallowing disturbance questionnaire for detecting dysphagia in patients with Parkinson's disease. Mov Disord 2007; 22: 191721.Google Scholar
Evatt, ML, Chaudhuri, KR, Chou, KL, et al. Dysautonomia rating scales in Parkinson's disease: sialorrhea, dysphagia, and constipation – critique and recommendations by movement disorders task force on rating scales for Parkinson's disease. Mov Disord 2009; 24: 63546.Google Scholar
Visser, M, Marinus, J, Stiggelbout, AM, van Hilten, JJ. Assessment of autonomic dysfunction in Parkinson's disease: the SCOPA-AUT. Mov Disord 2004; 19: 130612.Google Scholar
Rodriguez-Blazquez, C, Forjaz, MJ, Frades-Payo, B, de Pedro-Cuesta, J, Martinez-Martin, P. Independent validation of the scales for outcomes in Parkinson's disease-autonomic (SCOPA-AUT). Eur J Neurol 2010; 17: 194201.Google Scholar
Forjaz, MJ, Ayala, A, Rodriguez-Blazquez, C, Frades-Payo, B, Martinez-Martin, P. Assessing autonomic symptoms of Parkinson's disease with the SCOPA-AUT: a new perspective from Rasch analysis. Eur J Neurol 2010; 17: 2739.Google Scholar
Brown, RG, Dittner, A, Findley, L, Wessely, SC. The Parkinson fatigue scale. Parkinsonism Relat Disord 2005; 11: 4955.Google Scholar
Friedman, JH, Alves, G, Hagell, P, et al. Fatigue rating scales critique and recommendations by the Movement Disorders Society task force on rating scales for Parkinson's disease. Mov Disord 2010; 25: 80522.Google Scholar
Rascol, O, Fitzer-Attas, CJ, Hauser, R, 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: 41523.Google Scholar
Martinez-Martin, P, Jeukens-Visser, M, Lyons, KE, et al. Health-related quality-of-life scales in Parkinson's disease: critique and recommendations. Mov Disord 2011; 26: 237180.Google Scholar
Bergner, M, Bobbitt, RA, Carter, WB, Gilson, BS. The Sickness Impact Profile: development and final revision of a health status measure. Med Care 1981; 19: 787805.Google Scholar
Hunt, SM, McEwen, J, McKenna, SP. Measuring health status: a new tool for clinicians and epidemiologists. J R Coll Gen Pract 1985; 35: 1858.Google Scholar
EuroQol Group. EuroQol – a new facility for the measurement of health-related quality of life. Health Policy 1990; 16: 199208.Google Scholar
Ware, JE Jr, Sherbourne, CD. The MOS 36-item Short-Form Health Survey (SF-36). I. Conceptual framework and item selection. Med Care 1992; 30: 47383.Google Scholar
Peto, V, Jenkinson, C, Fitzpatrick, R, Greenhall, R. The development and validation of a short measure of functioning and well being for individuals with Parkinson's disease. Qual Life Res. 1995; 4: 2418.Google Scholar
Jenkinson, C, Fitzpatrick, R, Peto, V, Greenhall, R, Hyman, N. The Parkinson's Disease Questionnaire (PDQ-39): development and validation of a Parkinson's disease summary index score. Age Ageing 1997; 26: 3537.Google Scholar
Martinez-Martin, P, Deuschl, G. Effect of medical and surgical interventions on health-related quality of life in Parkinson's disease. Mov Disord 2007; 22: 75765.Google Scholar
Martinez-Martin, P, Kurtis, MM. Health-related quality of life as an outcome variable in Parkinson's disease. Ther Adv Neurol Disord 2012; 5: 10517.Google Scholar
Peto, V, Jenkinson, C, Fitzpatrick, R. Determining minimally important differences for the PDQ-39 Parkinson's Disease Questionnaire. Age Ageing 2001; 30: 299302.Google Scholar
Fitzpatrick, R, Norquist, JM, Jenkinson, C. Distribution-based criteria for change in health-related quality of life in Parkinson's disease. J Clin Epidemiol 2004; 57: 404.Google Scholar
Jenkinson, C, Fitzpatrick, R, Peto, V, Greenhall, R, Hyman, N. The PDQ-8: development and validation of a short-form Parkinson's Disease Questionnaire. Psychol Health 1997; 12: 80514.Google Scholar
De Boer, AG, Wijker, W, Speelman, JD, de Haes, JC. Quality of life in patients with Parkinson's disease: development of a questionnaire. J Neurol Neurosurg Psychiatry 1996; 61: 704.Google Scholar
Calne, S, Schulzer, M, Mak, E, et al. Validating a quality of life rating scale for idiopathic parkinsonism: Parkinson's Impact Scale (PIMS). Parkinsonism Relat Disord 1996; 2: 5561.Google Scholar
Marinus, J, Visser, M, Martínez-Martín, P, van Hilten, JJ, Stiggelbout, AM. A short psychosocial questionnaire for patients with Parkinson's disease: the SCOPA-PS. J Clin Epidemiol 2003; 56: 617.Google Scholar
Martínez-Martin, P, Carod-Artal, FJ, da Silveira Ribeiro, L, et al. Longitudinal psychometric attributes, responsiveness, and importance of change: an approach using the SCOPA-Psychosocial questionnaire. Mov Disord 2008; 23: 151623.Google Scholar

References

Ravina, B, Eidelberg, D, Ahlskog, JE, et al. The role of radiotracer imaging in Parkinson disease. Neurology 2005; 64: 20815.Google Scholar
Bernheimer, H, Birkmayer, W, Hornykiewicz, O, Jellinger, K, Seitelberger, F. Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci 1973; 20: 41555.Google Scholar
Stoessl, AJ, Martin, WW, McKeown, MJ, Sossi, V. Advances in imaging in Parkinson's disease. Lancet Neurol 2011; 10: 9871001.Google Scholar
Brooks, DJ, Pavese, N. Imaging biomarkers in Parkinson's disease. Prog Neurobiol 2011; 95: 61428.Google Scholar
Brooks, DJ. The role of structural and functional imaging in parkinsonian states with a description of PET technology. Semin Neurol 2008; 28: 43545.Google Scholar
Vingerhoets, FJ, Schulzer, M, Calne, DB, Snow, BJ. Which clinical sign of Parkinson's disease best reflects the nigrostriatal lesion? Ann Neurol, 1997; 41: 5864.Google Scholar
de la Fuente-Fernandez, R, Schulzer, M, Mak, E, Sossi, V. Trials of neuroprotective therapies for Parkinson's disease: problems and limitations. Parkinsonism Relat Disord 2010; 16: 3659.Google Scholar
Scherfler, C, Schwarz, J, Antonini, A, et al. Role of DAT-SPECT in the diagnostic work up of parkinsonism. Mov Disord 2007; 22: 122938.Google Scholar
Laruelle, M. Imaging synaptic neurotransmission with in vivo binding competition techniques: a critical review. J Cereb Blood Flow Metab 2000; 20: 42351.Google Scholar
Pavese, N, Evans, AH, Tai, YF, et al. Clinical correlates of levodopa-induced dopamine release in Parkinson disease: a PET study. Neurology 2006; 67: 161217.Google Scholar
Baglieri, A, Marino, MA, Morabito, R, et al. Differences between conventional and nonconventional MRI techniques in Parkinson's disease. Funct Neurol 2013; 28: 7382.Google Scholar
Cho, ZH, Oh, SH, Kim, JM, et al. Direct visualization of Parkinson's disease by in vivo human brain imaging using 7.0T magnetic resonance imaging. Mov Disord 2011; 26: 71318.Google Scholar
Mahlknecht, P, Hotter, A, Hussl, A, et al. Significance of MRI in diagnosis and differential diagnosis of Parkinson's disease. Neurodegener Dis 2010; 7: 30018.Google Scholar
Peran, P, Cherubini, A, Assogna, F, et al. Magnetic resonance imaging markers of Parkinson's disease nigrostriatal signature. Brain 2010; 133: 342333.Google Scholar
Tang, CC, Eidelberg, D. Abnormal metabolic brain networks in Parkinson's disease from blackboard to bedside. Prog Brain Res 2010; 184: 16176.Google Scholar
Niethammer, M, Feigin, A, Eidelberg, D. Functional neuroimaging in Parkinson's disease. Cold Spring Harb Perspect Med 2012; 2: a009274.Google Scholar
Stoessl, AJ. Neuroimaging in Parkinson's disease. Neurotherapeutics 2011; 8: 7281.Google Scholar
Nandhagopal, R, McKeown, MJ, Stoessl, AJ. Functional imaging in Parkinson disease. Neurology 2008; 70: 147888.Google Scholar
Berg, D. Hyperechogenicity of the substantia nigra: pitfalls in assessment and specificity for Parkinson's disease. J Neural Transm 2011; 118: 45361.Google Scholar
Ouchi, Y, Yagi, S, Yokokura, M, Sakamoto, M. Neuroinflammation in the living brain of Parkinson's disease. Parkinsonism Relat Disord 2009; 15 (Suppl. 3): S2004.Google Scholar
Yarnall, A, Rochester, L, Burn, DJ. The interplay of cholinergic function, attention, and falls in Parkinson's disease. Mov Disord 2011; 26: 2496503.Google Scholar
Vernon, AC, Ballard, C, Modo, M. Neuroimaging for Lewy body disease: is the in vivo molecular imaging of α-synuclein neuropathology required and feasible? Brain Res Rev 2010; 65: 2855.Google Scholar
Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog Neurobiol 2011; 95: 62935.Google Scholar
Catana, C, Drzezga, A, Heiss, WD, Rosen, BR. PET/MRI for neurologic applications. J Nucl Med 2012; 53: 191625.Google Scholar
Stern, MB, Lang, A, Poewe, W. Toward a redefinition of Parkinson's disease. Mov Disord 2012; 27: 5460.Google Scholar
Brooks, DJ, Frey, KA, Marek, KL, et al. Assessment of neuroimaging techniques as biomarkers of the progression of Parkinson's disease. Exp Neurol 2003; 184 (Suppl. 1): S6879.Google Scholar
Agarwal, PA, Stoessl, AJ. Biomarkers for trials of neuroprotection in Parkinson's disease. Mov Disord 2013; 28: 7185.Google Scholar
Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287: 165361.Google Scholar
Whone, AL, Watts, RL, Stoessl, AJ, et al. Slower progression of Parkinson's disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54: 93101.Google Scholar
Fahn, S, Parkinson Study Group. Does levodopa slow or hasten the rate of progression of Parkinson's disease? J Neurol 2005; 252 (Suppl. 4): IV3742.Google Scholar
Jack, CR Jr, Holtzman, DM. Biomarker modeling of Alzheimer's disease. Neuron 2013; 80: 134758.Google Scholar
Lim, SY, Fox, SH, Lang, AE. Overview of the extranigral aspects of Parkinson disease. Arch Neurol 2009; 66: 16772.Google Scholar
Doherty, KM, Silveira-Moriyama, L, Parkkinen, L, et al. Parkin disease: a clinicopathologic entity? JAMA Neurol 2013; 70: 5719.Google Scholar
Alves, G, Wentzel-Larsen, T, Aarsland, D, Larsen, JP. Progression of motor impairment and disability in Parkinson disease: a population-based study. Neurology 2005; 65: 143641.Google Scholar
Benedetti, F, Carlino, E, Pollo, A. How placebos change the patient's brain. Neuropsychopharmacology 2011; 36: 33954.Google Scholar
Lidstone, SC, Schulzer, M, Dinelle, K, et al. Effects of expectation on placebo-induced dopamine release in Parkinson disease. Arch Gen Psychiatry 2010; 67: 85765.Google Scholar
Weiner, MW, Veitch, DP, Aisen, PS, et al. The Alzheimer's Disease Neuroimaging Initiative: a review of papers published since its inception. Alzheimers Dement 2012; 8 (Suppl.): S168.Google Scholar

References

Biomarkers Definitions Working Group. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther 2001; 69: 8995.Google Scholar
Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287: 165361.Google Scholar
Whone, AL, Watts, RL, Stoessl, AJ, et al. Slower progression of Parkinson's disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54: 93101.CrossRefGoogle ScholarPubMed
Fahn, S, Oakes, D, Shoulson, I, et al. Levodopa and the progression of Parkinson's disease. N Engl J Med 2004; 351: 2498508.Google Scholar
Freed, CR, Greene, PE, Breeze, RE, et al. Transplantation of embryonic dopamine neurons for severe Parkinson's disease. N Engl J Med 2001; 344: 71019.Google Scholar
Olanow, CW, Goetz, CG, Kordower, JH, et al. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson's disease. Ann Neurol 2003; 54: 40314.Google Scholar
Feigin, A, Kaplitt, MG, Tang, C, et al. Modulation of metabolic brain networks after subthalamic gene therapy for Parkinson's disease. Proc Natl Acad Sci U S A 2007; 104: 1955964.Google Scholar
Parkinson Progression Marker Initiative. The Parkinson Progression Marker Initiative (PPMI). Prog Neurobiol 2011; 95: 62935.Google Scholar
Kang, UJ, Alcalay, R, Goldman, JG, et al. The BioFIND study (Fox Investigation for New Discovery of Biomarkers in Parkinson's disease): design and methodology. Neurology 2014; 10 (Suppl.): P4.043.Google Scholar
Parkinson Study Group. DATATOP: a decade of neuroprotective inquiry. Deprenyl And Tocopherol Antioxidative Therapy Of Parkinsonism. Ann Neurol 1998; 44: S1606.Google Scholar
Gerlach, M, Maetzler, W, Broich, K, et al. Biomarker candidates of neurodegeneration in Parkinson's disease for the evaluation of disease-modifying therapeutics. J Neural Transm 2012; 119: 3952.CrossRefGoogle ScholarPubMed
Michell, AW, Lewis, SJ, Foltynie, T, Barker, RA. Biomarkers and Parkinson's disease. Brain 2004; 127: 1693705.Google Scholar
LeWitt, P. Recent advances in CSF biomarkers for Parkinson's disease. Parkinsonism Relat Disord 2012; 18 (Suppl. 1): S4951.Google Scholar
LeWitt, P, Schultz, L, Auinger, P, et al. CSF xanthine, homovanillic acid, and their ratio as biomarkers of Parkinson's disease. Brain Res 2011; 1408: 8897.Google Scholar
Zhou, G, Miura, Y, Shoji, H, Yamada, S, Matsuishi, T. Platelet monoamine oxidase B and plasma β-phenylethylamine in Parkinson's disease. J Neurol Neurosurg Psychiatry 2001; 70: 22931.Google Scholar
Barbanti, P, Fabbrini, G, Ricci, A, et al. Increased expression of dopamine receptors on lymphocytes in Parkinson's disease. Mov Disord 1999; 14: 76471.Google Scholar
Gui, YX, Wan, Y, Xiao, Q, et al. Verification of expressions of Kir2 as potential peripheral biomarkers in lymphocytes from patients with Parkinson's disease. Neurosci Lett 2011; 505: 1048.Google Scholar
Iwanaga, K, Wakabayashi, K, Yoshimoto, M, et al. Lewy body-type degeneration in cardiac plexus in Parkinson's and incidental Lewy body diseases. Neurology 1999; 52: 126971.Google Scholar
Minguez-Castellanos, A, Chamorro, CE, Escamilla-Sevilla, F, et al. Do α-synuclein aggregates in autonomic plexuses predate Lewy body disorders? A cohort study. Neurology 2007; 68: 201218.Google Scholar
Beach, TG, Adler, CH, Dugger, BN, et al. Submandibular gland biopsy for the diagnosis of Parkinson disease. J Neuropathol Exp Neurol 2013; 72: 1306.CrossRefGoogle ScholarPubMed
Wang, N, Gibbons, CH, Lafo, J, et al. α-Synuclein in cutaneous autonomic nerves. Neurology 2013; 81: 160410.Google Scholar
El-Agnaf, OM, Salem, SA, Paleologou, KE, et al. Detection of oligomeric forms of α-synuclein protein in human plasma as a potential biomarker for Parkinson's disease. Faseb J 2006; 20: 41925.CrossRefGoogle ScholarPubMed
Tinsley, RB, Kotschet, K, Modesto, D, et al. Sensitive and specific detection of α-synuclein in human plasma. J Neurosci Res 2010; 88: 2693700.Google Scholar
Foulds, PG, Mitchell, JD, Parker, A, et al. Phosphorylated α-synuclein can be detected in blood plasma and is potentially a useful biomarker for Parkinson's disease. FASEB J 2011; 25: 412737.CrossRefGoogle ScholarPubMed
Tokuda, T, Salem, SA, Allsop, D, et al. Decreased α-synuclein in cerebrospinal fluid of aged individuals and subjects with Parkinson's disease. Biochem Biophys Res Commun 2006; 349: 1626.Google Scholar
Lee, PH, Lee, G, Park, HJ, et al. The plasma α-synuclein levels in patients with Parkinson's disease and multiple system atrophy. J Neural Transm 2006; 113: 14359.Google Scholar
Shi, M, Zabetian, CP, Hancock, AM, et al. Significance and confounders of peripheral DJ-1 and α-synuclein in Parkinson's disease. Neuroscience Lett 2010; 480: 7882.Google Scholar
Li, QX, Mok, SS, Laughton, KM, et al. Plasma α-synuclein is decreased in subjects with Parkinson's disease. Exp Neurol 2007; 204: 5838.Google Scholar
Ohrfelt, A, Grognet, P, Andreasen, N, et al. Cerebrospinal fluid α-synuclein in neurodegenerative disorders-a marker of synapse loss? Neurosci Lett 2009; 450: 3325.CrossRefGoogle ScholarPubMed
Hong, Z, Shi, M, Chung, K, et al. DJ-1 and α-synuclein in human cerebrospinal fluid as biomarkers of Parkinson's disease. Brain 2010; 133: 71326.Google Scholar
Waragai, M, Sekiyama, K, Sekigawa, A, et al. α-Synuclein and DJ-1 as potential biological fluid biomarkers for Parkinson's Disease. Int J Mol Sci 2010; 11: 425766.Google Scholar
Mollenhauer, B, Locascio, JJ, Schulz-Schaeffer, W, et al. α-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: a cohort study. Lancet Neurol 2011; 10: 23040.Google Scholar
Wennstrom, M, Surova, Y, Hall, S, et al. Low CSF levels of both α-synuclein and the α-synuclein cleaving enzyme neurosin in patients with synucleinopathy. PLoS One 2013; 8: e53250.Google Scholar
Hall, S, Ohrfelt, A, Constantinescu, R, et al. Accuracy of a panel of 5 cerebrospinal fluid biomarkers in the differential diagnosis of patients with dementia and/or parkinsonian disorders. Arch Neurol 2012; 69: 144552.Google Scholar
Kang, JH, Irwin, DJ, Chen-Plotkin, AS, et al. Association of cerebrospinal fluid beta-amyloid 1–42, T-tau, P-tau181, and α-synuclein levels with clinical features of drug-naive patients with early Parkinson disease. JAMA Neurol 2013; 70: 127787.Google Scholar
Mollenhauer, B, Trautmann, E, Taylor, P, et al. Total CSF α-synuclein is lower in de novo Parkinson patients than in healthy subjects. Neurosci Lett 2013; 532: 448.Google Scholar
Shi, M, Bradner, J, Hancock, AM, et al. Cerebrospinal fluid biomarkers for Parkinson disease diagnosis and progression. Ann Neurol 2011; 69: 57080.CrossRefGoogle ScholarPubMed
Stewart, T, Liu, C, Ginghina, C, et al. Cerebrospinal fluid α-synuclein predicts cognitive decline in Parkinson disease progression in the DATATOP cohort. Am J Pathol 2014; 184: 96675.Google Scholar
Masliah, E, Rockenstein, E, Adame, A, et al. Effects of α-synuclein immunization in a mouse model of Parkinson's disease. Neuron 2005; 46: 85768.CrossRefGoogle Scholar
Masliah, E, Rockenstein, E, Mante, M, et al. Passive immunization reduces behavioral and neuropathological deficits in an α-synuclein transgenic model of Lewy body disease. PLoS One 2011; 6: e19338.Google Scholar
Nasstrom, T, Goncalves, S, Sahlin, C, et al. Antibodies against α-synuclein reduce oligomerization in living cells. PLoS One 2011; 6: e27230.Google Scholar
Blennow, K, Zetterberg, H, Rinne, JO, et al. Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol 2012; 69: 100210.Google ScholarPubMed
Salloway, S, Sperling, R, Fox, NC, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med 2014; 370: 32233.Google Scholar
Farlow, M, Arnold, SE, van Dyck, CH, et al. Safety and biomarker effects of solanezumab in patients with Alzheimer's disease. Alzheimers Dement 2012; 8: 26171.Google Scholar
Doody, RS, Thomas, RG, Farlow, M, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med 2014; 370: 31121.CrossRefGoogle ScholarPubMed
Maita, C, Tsuji, S, Yabe, I, et al. Secretion of DJ-1 into the serum of patients with Parkinson's disease. Neurosci Lett 2008; 431: 869.Google Scholar
Waragai, M, Nakai, M, Wei, J, et al. Plasma levels of DJ-1 as a possible marker for progression of sporadic Parkinson's disease. Neurosci Lett 2007; 425: 1822.Google Scholar
Waragai, M, Wei, J, Fujita, M, et al. Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson's disease. Biochem Biophys Res Commun 2006; 345: 96772.Google Scholar
Lin, X, Cook, TJ, Zabetian, CP, et al. DJ-1 isoforms in whole blood as potential biomarkers of Parkinson disease. Sci Rep 2012; 2: 954.Google Scholar
Saito, Y, Hamakubo, T, Yoshida, Y, et al. Preparation and application of monoclonal antibodies against oxidized DJ-1. Significant elevation of oxidized DJ-1 in erythrocytes of early-stage Parkinson disease patients. Neurosci Lett 2009; 465: 15.Google Scholar
Rosén, C, Hansson, O, Blennow, K, Zetterberg, H. Fluid biomarkers in Alzheimer's disease – current concepts. Mol Neurodegener 2013; 8: 2031.Google Scholar
Maarouf, CL, Beach, TG, Adler, CH, et al. Quantitative appraisal of ventricular cerebrospinal fluid biomarkers in neuropathologically diagnosed Parkinson's disease cases lacking Alzheimer's disease pathology. Biomark Insights 2013; 8: 1928.Google Scholar
Siderowf, A, Xie, SX, Hurtig, H, et al. CSF amyloid β1–42 predicts cognitive decline in Parkinson disease. Neurology 2010; 75: 105561.Google Scholar
Wang, Y, Hancock, AM, Bradner, J, et al. Complement 3 and factor h in human cerebrospinal fluid in Parkinson's disease, Alzheimer's disease, and multiple-system atrophy. Am J Pathol 2011; 178: 150916.Google Scholar
Leverenz, JB, Watson, GS, Shofer, J, et al. Cerebrospinal fluid biomarkers and cognitive performance in non-demented patients with Parkinson's disease. Parkinsonism Relat Disord 2011; 17: 614.Google Scholar
Henchcliffe, C, Beal, MF. Mitochondrial biology and oxidative stress in Parkinson disease pathogenesis. Nat Clin Pract Neurol 2008; 4: 6009.Google Scholar
Hauser, DN, Hastings, TG. Mitochondrial dysfunction and oxidative stress in Parkinson's disease and monogenic parkinsonism. Neurobiol Dis 2013; 51: 3542.Google Scholar
Younes-Mhenni, S, Frih-Ayed, M, Kerkeni, A, Bost, M, Chazot, G. Peripheral blood markers of oxidative stress in Parkinson's disease. Eur Neurol 2007; 58: 7883.Google Scholar
Götz, ME, Gerstner, A, Harth, R, et al. Altered redox state of platelet coenzyme Q10 in Parkinson's disease. J Neural Transm 2000; 107: 418.Google ScholarPubMed
Sohmiya, M, Tanaka, M, Tak, NW, et al. Redox status of plasma coenzyme Q10 indicates elevated systemic oxidative stress in Parkinson's disease. J Neurol Sci 2004; 223: 1616.Google Scholar
Isobe, C, Abe, T, Terayama, Y. Levels of reduced and oxidized coenzymeQ-10 and 8-hydroxy-2'-deoxyguanosine in the cerebrospinal fluid of patients with living Parkinson's disease demonstrate that mitochondrial oxidative damage and/or oxidative DNA damage contributes to the neurodegenerative process. Neurosci Lett 2010; 469: 15963.CrossRefGoogle Scholar
Ilic, TV, Jovanovic, M, Jovicic, A, Tomovic, M. Oxidative stress indicators are elevated in de novo Parkinson's disease patients. Funct Neurol 1999; 14: 1417.Google Scholar
Seet, RC, Lee, CY, Lim, EC, et al. Oxidative damage in Parkinson disease: Measurement using accurate biomarkers. Free Radic Biol Med 2010; 48: 5606.Google Scholar
Bogdanov, M, Matson, WR, Wang, L, et al. Metabolomic profiling to develop blood biomarkers for Parkinson's disease. Brain 2008; 131: 38996.Google Scholar
Gmitterova, K, Heinemann, U, Gawinecka, J, et al. 8-OHdG in cerebrospinal fluid as a marker of oxidative stress in various neurodegenerative diseases. Neurodegener Dis 2009; 6: 2639.Google Scholar
Andreux, PA, Houtkooper, RH, Auwerx, J. Pharmacological approaches to restore mitochondrial function. Nat Rev Drug Discov 2013; 12: 46583.Google Scholar
Seet, RC, Lim, EC, Tan, JJ, et al. Effects of high-dose coenzyme Q10 on biomarkers of oxidative damage and clinical outcomes in Parkinson disease. Antioxidants & Redox Signaling 2014; 21: 21117.Google Scholar
Cipriani, S, Desjardins, CA, Burdett, TC, et al. Urate and its transgenic depletion modulate neuronal vulnerability in a cellular model of Parkinson's disease. PLoS One 2012; 7: e37331.Google Scholar
Gong, L, Zhang, QL, Zhang, N, et al. Neuroprotection by urate on 6-OHDA-lesioned rat model of Parkinson's disease: linking to Akt/GSK3β signaling pathway. J Neurochem 2012; 123: 87685.Google Scholar
Chen, X, Burdett, TC, Desjardins, CA, et al. Disrupted and transgenic urate oxidase alter urate and dopaminergic neurodegeneration. Proc Natl Acad Sci U S A 2013; 110: 3005.Google Scholar
Cipriani, S, Chen, X, Schwarzschild, MA. Urate: a novel biomarker of Parkinson's disease risk, diagnosis and prognosis. Biomark Med 4: 70112.Google Scholar
Ascherio, A, LeWitt, PA, Xu, K, et al. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol 2009; 66: 14608.Google Scholar
Schwarzschild, MA, Schwid, SR, Marek, K, et al. Serum urate as a predictor of clinical and radiographic progression in Parkinson disease. Arch Neurol 2008; 65: 71623.Google Scholar
The Parkinson Study Group. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol 2014; 71: 14150.Google Scholar
Johansen, KK, Wang, L, Aasly, JO, et al. Metabolomic profiling in LRRK2-related Parkinson's disease. PLoS One 2009; 4: e7551.Google Scholar
Rentzos, M, Nikolaou, C, Andreadou, E, et al. Circulating interleukin-15 and RANTES chemokine in Parkinson's disease. Acta Neurol Scand 2007; 116: 3749.Google Scholar
Dufek, M, Hamanova, M, Lokaj, J, et al. Serum inflammatory biomarkers in Parkinson's disease. Parkinsonism Relat Disord 2009; 15: 31820.Google Scholar
Mogi, M, Harada, M, Riederer, P, et al. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci Lett 1994; 165: 20810.Google Scholar
Barnum, CJ, Tansey, MG. Neuroinflammation and non-motor symptoms: the dark passenger of Parkinson's disease? Curr Neurol Neurosci Rep 2012; 12: 3508.Google Scholar
Rocha, NP, Teixeira, AL, Scalzo, PL, et al. Plasma levels of soluble tumor necrosis factor receptors are associated with cognitive performance in Parkinson's disease. Mov Disord 2014; 29: 52731.Google Scholar
Lindqvist, D, Hall, S, Surova, Y, et al. Cerebrospinal fluid inflammatory markers in Parkinson's disease – associations with depression, fatigue, and cognitive impairment. Brain Behav Immun 2013; 33: 1839.Google Scholar
Hisanaga, K, Asagi, M, Itoyama, Y, Iwasaki, Y. Increase in peripheral CD4 bright+ CD8 dull+ T cells in Parkinson disease. Arch Neurol 2001; 58: 15803.Google Scholar
Fiszer, U, Mix, E, Fredrikson, S, Kostulas, V, Link, H. Parkinson's disease and immunological abnormalities: increase of HLA-DR expression on monocytes in cerebrospinal fluid and of CD45RO+ T cells in peripheral blood. Acta Neurol Scand 1994; 90: 1606.Google Scholar
Double, KL, Rowe, DB, Carew-Jones, FM, et al. Anti-melanin antibodies are increased in sera in Parkinson's disease. Exp Neurol 2009; 217: 297301.Google Scholar
McRae-Degueurce, A, Klawans, HL, Penn, RD, et al. An antibody in the CSF of Parkinson's disease patients disappears following adrenal medulla transplantation. Neurosci Lett 1988; 94: 1927.Google Scholar
Kaplitt, MG, Feigin, A, Tang, C, et al. Safety and tolerability of gene therapy with an adeno-associated virus (AAV) borne GAD gene for Parkinson's disease: an open label, phase I trial. Lancet 2007; 369: 2097105.Google Scholar
Papachroni, KK, Ninkina, N, Papapanagiotou, A, et al. Autoantibodies to α-synuclein in inherited Parkinson's disease. J Neurochem 2007; 101: 74956.Google Scholar
Yanamandra, K, Gruden, MA, Casaite, V, et al. α-synuclein reactive antibodies as diagnostic biomarkers in blood sera of Parkinson's disease patients. PLoS One 2011; 6: e18513.Google Scholar
Caudle, WM, Bammler, TK, Lin, Y, Pan, S, Zhang, J. Using ‘omics’ to define pathogenesis and biomarkers of Parkinson's disease. Expert Rev Neurother 2010; 10: 92542.Google Scholar
Grunblatt, E, Mandel, S, Jacob-Hirsch, J, et al. Gene expression profiling of parkinsonian substantia nigra pars compacta; alterations in ubiquitin-proteasome, heat shock protein, iron and oxidative stress regulated proteins, cell adhesion/cellular matrix and vesicle trafficking genes. J Neural Transm 2004; 111: 154373.Google Scholar
Moran, LB, Graeber, MB. Towards a pathway definition of Parkinson's disease: a complex disorder with links to cancer, diabetes and inflammation. Neurogenetics 2008; 9: 113.Google Scholar
Sutherland, GT, Matigian, NA, Chalk, AM, et al. A cross-study transcriptional analysis of Parkinson's disease. PLoS One 2009; 4: e4955.Google Scholar
Grunblatt, E, Zehetmayer, S, Jacob, CP, et al. Pilot study: peripheral biomarkers for diagnosing sporadic Parkinson's disease. J Neural Transm 2010; 117: 138793.Google Scholar
Scherzer, CR, Eklund, AC, Morse, LJ, et al. Molecular markers of early Parkinson's disease based on gene expression in blood. Proc Natl Acad Sci U S A 2007; 104: 95560.Google Scholar
Molochnikov, L, Rabey, JM, Dobronevsky, E, et al. A molecular signature in blood identifies early Parkinson's disease. Mol Neurodegener 2012; 7: 26.Google Scholar
Lauterbach, EC. Psychotropic drug effects on gene transcriptomics relevant to Parkinson's disease. Prog Neuropsychopharmacol Biol Psychiatry 2012; 38: 10715.Google Scholar
Basso, M, Giraudo, S, Corpillo, D, et al. Proteome analysis of human substantia nigra in Parkinson's disease. Proteomics 2004; 4: 394352.Google Scholar
Werner, CJ, Heyny-von Haussen, R, Mall, G, Wolf, S. Proteome analysis of human substantia nigra in Parkinson's disease. Proteome Science 2008; 6: 822.Google Scholar
Abdi, F, Quinn, JF, Jankovic, J, et al. Detection of biomarkers with a multiplex quantitative proteomic platform in cerebrospinal fluid of patients with neurodegenerative disorders. J Alzheimers Dis 2006; 9: 293348.Google Scholar
Jin, J, Hulette, C, Wang, Y, et al. Proteomic identification of a stress protein, mortalin/mthsp70/GRP75: relevance to Parkinson disease. Mol Cell Proteomics 2006; 5: 1193204.Google Scholar
Pan, S, Rush, J, Peskind, ER, et al. Application of targeted quantitative proteomics analysis in human cerebrospinal fluid using a liquid chromatography matrix-assisted laser desorption/ionization time-of-flight tandem mass spectrometer (LC MALDI TOF/TOF) platform. J Proteome Res 2008; 7: 72030.Google Scholar
Li, YH, Wang, J, Zheng, XL, et al. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry combined with magnetic beads for detecting serum protein biomarkers in parkinson's disease. Eur Neurol 2011; 65: 10511.Google Scholar
Zhao, X, Xiao, WZ, Pu, XP, et al. Proteome analysis of the sera from Chinese Parkinson's disease patients. Neurosci Lett 2010; 479: 1759.Google Scholar
Mila, S, Albo, AG, Corpillo, D, et al. Lymphocyte proteomics of Parkinson's disease patients reveals cytoskeletal protein dysregulation and oxidative stress. Biomark Med 2009; 3: 11728.Google Scholar
Wang, ES, Yao, HB, Chen, YH, et al. Proteomic analysis of the cerebrospinal fluid of Parkinson's disease patients pre- and post-deep brain stimulation. Cell Physiol Biochem 2013; 31: 62537.Google Scholar
Gershon, RC, Cella, D, Fox, NA, et al. Assessment of neurological and behavioural function: the NIH Toolbox. Lancet Neurolo 2010; 9: 1389.Google Scholar
Kozauer, N, Katz, R. Regulatory innovation and drug development for early-stage Alzheimer's disease. New Engl J Med 2013; 368: 116971.Google Scholar
Vellas, B, Carrillo, MC, Sampaio, C, et al. Designing drug trials for Alzheimer's disease: what we have learned from the release of the phase III antibody trials: a report from the EU/US/CTAD Task Force. Alzheimers Dement 2013; 9: 43844.Google Scholar
Mattsson, N, Andreasson, U, Persson, S, et al. CSF biomarker variability in the Alzheimer's Association quality control program. Alzheimers Dement 2013; 9: 25161.Google Scholar
Carrillo, MC, Blennow, K, Soares, H, et al. Global standardization measurement of cerebral spinal fluid for Alzheimer's disease: an update from the Alzheimer's Association Global Biomarkers Consortium. Alzheimers Dement 2013; 9: 13740.Google Scholar

References

Lang, AE. Clinical trials of disease-modifying therapies for neurodegenerative diseases: the challenges and the future. Nat Med 2010; 16: 12236.CrossRefGoogle ScholarPubMed
Stocchi, F, Olanow, CW. Obstacles to the development of a neuroprotective therapy for Parkinson's disease. Mov Disord 2013; 28: 37.Google Scholar
Olanow, CW, Kieburtz, K, Schapira, AHV. Why have we failed to achieve neuroprotection in Parkinson's disease? Ann Neurol 2009; 64 (Suppl. 2): S10110.Google Scholar
Hirsch, EC. How to improve neuroprotection in Parkinson's disease? Parkinsonism Relat Disord 2007; 13 (Suppl. 3): S3325.Google Scholar
D'Agostino, RB. The delayed-start study design. N Engl J Med 2009; 361: 13046.Google Scholar
Cheng, HC, Ulane, CM, Burke, RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol 2010; 67: 71525.Google Scholar
Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized controlled trial. JAMA 2000; 284: 19318.Google Scholar
Fahn, S, Oakes, D, Shoulson, I, et al. Levodopa and the progression of Parkinson's disease. N Engl J Med 2004; 351: 2498508.Google Scholar
Whone, AL, Watts, RL, Stoessl, AJ, et al. Slower progression of Parkinson's disease with ropinirole versus levodopa: the REAL-PET study. Ann Neurol 2003; 54: 93101.Google Scholar
Goetz, CG, Tilley, BC, Shaftman, SR, 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: 212970.Google Scholar
Hauser, RA, Auinger, P, Parkinson Study Group. Determination of minimal clinically important change in early and advanced Parkinson's disease. Mov Disord 2011; 26: 8138.Google Scholar
Richards, M, Marder, K, Cote, L, Mayeux, R. Interrater reliability of the Unified Parkinson's Disease Rating Scale motor examination. Mov Disord 1994; 9: 8991.Google Scholar
Goetz, CG, Leurgans, S, Raman, R, Parkinson Study Group. Placebo-associated improvements in motor function: Comparison of subjective and objective sections of the UPDRS in early Parkinson's disease. Mov Disord 2002; 17: 2838.Google Scholar
Parashos, SA, Swearingen, CJ, Biglan, KM, et al. Determinants of the timing of symptomatic treatment in early Parkinson disease: The National Institutes of Health Exploratory Trials in Parkinson Disease (NET-PD) Experience. Arch Neurol 2009; 66: 1099104.Google Scholar
Ravina, B, Camicioli, R, Como, PG, et al. The impact of depressive symptoms in early Parkinson disease. Neurology 2007; 69: 3427.Google Scholar
Marras, C, McDermott, MP, Marek, K, et al. Predictors of time to requiring dopaminergic treatment in 2 Parkinson's disease cohorts. Mov Disord 2011; 26: 60813.Google Scholar
Agarwal, PA, Stoessl, AJ. Biomarkers for trials of neuroprotection in Parkinson's disease. Mov Disord 2012; 28: 7185.Google Scholar
Pavese, N, Kiferle, L, Piccini, P. Neuroprotection and imaging studies in Parkinson's disease. Parkinsonism Relat Disord 2009; 15 (Suppl. 4): S337.Google Scholar
Ravina, B, Eidelberg, D, Ahlskog, JE, et al. The role of radiotracer imaging in Parkinson disease. Neurology 2005; 64: 20815.Google Scholar
Snow, BJ, Tooyama, I, McGeer, EG, et al. Human positron emission tomographic [18F]fluorodopa studies correlate with dopamine cell counts and levels. Ann Neurol 1993; 34: 32430.Google Scholar
Nandhagopal, R, Kuramoto, L, Schulzer, M, et al. Longitudinal progression of sporadic Parkinson's disease: a multi-tracer positron emission tomography study. Brain. 2009; 132: 29709.Google Scholar
Vingerhoets, FJ, Schulzer, M, Calne, DB, Snow, BJ. Which clinical sign of Parkinson's disease best reflects the nigrostriatal lesion? Ann Neurol 1997; 41: 5864.Google Scholar
Pirker, W. Correlation of dopamine transporter imaging with parkinsonian motor handicap: how close is it? Mov Disord 2003; 18: S4351.Google Scholar
Martin, WRW, Wieler, M, Stoessl, AJ, Schulzer, M. Dihydrotetrabenazine positron emission tomography imaging in early, untreated Parkinson's disease. Ann Neurol 2008; 63: 38894.Google Scholar
Morrish, PK. How valid is dopamine transporter imaging as a surrogate marker in research trials in Parkinson's disease? Mov Disord 2003; 18 (Suppl. 7): S6370.Google Scholar
Parkinson Study Group. A randomized controlled trial comparing pramipexole with levodopa in early Parkinson's disease: design and methods of the CALM-PD Study. Clin Neuropharmacol 2000; 23: 3444.Google Scholar
Winogrodzka, A, Booij, J, Wolters, EC. Disease-related and drug-induced changes in dopamine transporter expression might undermine the reliability of imaging studies of disease progression in Parkinson's disease. Parkinsonism Relat Disord 2005; 11: 47584.Google Scholar
Eckert, T, Eidelberg, D. Neuroimaging and therapeutics in movement disorders. NeuroRx 2005; 2: 36171.Google Scholar
Eckert, T, Tang, C, Eidelberg, D. Assessment of the progression of Parkinson's disease: a metabolic network approach. Lancet Neurol 2007; 6: 92632.Google Scholar
Vaillancourt, DE, Spraker, MB, Prodoehl, J, et al. High-resolution diffusion tensor imaging in the substantia nigra of de novo Parkinson disease. Neurology 2009; 72: 137884.Google Scholar
Berg, D. Transcranial ultrasound as a risk marker for Parkinson's disease. Mov Disord 2009; 24 (Suppl. 2): S67783.Google Scholar
Sperling, RA, Aisen, PS, Beckett, LA, et al. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging-Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 2011; 7: 28092.Google Scholar
Lang, AE, Melamed, E, Poewe, W, Rascol, O. Trial designs used to study neuroprotective therapy in Parkinson's disease. Mov Disord 2012; 28: 8695.Google Scholar
Olanow, CW, Rascol, O, Hauser, R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson's disease. N Engl J Med 2009; 361: 126878.Google Scholar
Clarke, CE. Are delayed-start design trials to show neuroprotection in Parkinson's disease fundamentally flawed? Mov Disord 2008; 23: 7849.Google Scholar
Ahlskog, JE, Uitti, RJ. Rasagiline, Parkinson neuroprotection, and delayed-start trials: still no satisfaction? Neurology 2010; 74: 11438.Google Scholar
Schwid, SR, Cutter, GR. Futility studies: spending a little to save a lot. Neurology 2006; 66: 6267.Google Scholar
Bezard, E, Yue, Z, Kirik, D, Spillantini, MG. Animal models of Parkinson's disease: Limits and relevance to neuroprotection studies. Mov Disord 2012; 28: 6170.Google Scholar
Schober, A. Classic toxin-induced animal models of Parkinson's disease: 6-OHDA and MPTP. Cell Tissue Res 2004; J318: 21524.Google Scholar
Luk, KC, Kehm, V, Carroll, J, et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 2012; 338: 94953.Google Scholar
Olanow, CW, Brundin, P. Parkinson's disease and alpha synuclein: is Parkinson's disease a prion-like disorder? Mov Disord 2013; 28: 3140.Google Scholar
Lim, KL, Ng, CH. Genetic models of Parkinson disease. Biochim Biophys Acta 2009; 1792: 60415.Google Scholar
AlDakheel, A, Kalia, LV, Lang, AE. Pathogenesis-targeted, disease-modifying therapies in Parkinson disease. Neurotherapeutics 2013; 11: 623.Google Scholar
Parkinson Study Group. Effect of deprenyl on the progression of disability in early Parkinson's disease. N Engl J Med 1989; 321: 136471.Google Scholar
Olanow, CW, Hauser, RA, Gauger, L, et al. The effect of deprenyl and levodopa on the progression of Parkinson's disease. Ann Neurol 1995; 38: 7717.Google Scholar
Negrotti, A, Bizzarri, G, Calzetti, S. Long-term persistence of symptomatic effect of selegiline in Parkinson's disease. A two-months placebo-controlled withdrawal study. J Neural Transm 2001; 108: 21519.Google Scholar
Parkinson Study Group. A controlled trial of rasagiline in early Parkinson disease: the TEMPO Study. Arch Neurol 2002; 59: 193743.Google Scholar
Clarke, CE, Patel, S, Ives, N, et al. Should treatment for Parkinson's disease start immediately on diagnosis or delayed until functional disability develops? Mov Disord 2011; 26: 118793.Google Scholar
de la Fuente-Fernández, R, Schulzer, M, Mak, E, Sossi, V. Parkinsonism and related disorders. Parkinsonism Relat Disord 2010; 16: 3659.Google Scholar
Schwarzschild, MA. Rasagiline in Parkinson's disease. N Engl J Med 2010; 362: 658; author reply 6589.Google Scholar
Fahn, S. Is levodopa toxic? Neurology 1996; 47 (Suppl. 3): S18495.Google Scholar
Jenner, PG, Brin, MF. Levodopa neurotoxicity: experimental studies versus clinical relevance. Neurology 1998; 50 (Suppl. 6): S3948.CrossRefGoogle ScholarPubMed
Hauser, RA, Holford, NHG. Quantitative description of loss of clinical benefit following withdrawal of levodopa-carbidopa and bromocriptine in early Parkinson's disease. Mov Disord 2002; 17: 9618.Google Scholar
LEAP Study Protocol. Levodopa in early Parkinson's disease: the LEAP study. Availabe at: http://leapamc.nl/wp-content/uploads/2011-12-22-Onderzoeksprotocol.pdf.Google Scholar
Parkinson Study Group. Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. JAMA 2002; 287: 165361.Google Scholar
Parkinson Study Group, CALM Cohort Investigators. Long-term effect of initiating pramipexole vs levodopa in early Parkinson disease. Arch Neurol 2009; 66: 56370.Google Scholar
Schapira, AHV, Albrecht, S, Barone, P, et al. Rationale for delayed-start study of pramipexole in Parkinson's disease: the PROUD study. Mov Disord 2010; 25: 162732.Google Scholar
Schapira, AHV, McDermott, MP, Barone, P, et al. Pramipexole in patients with early Parkinson's disease (PROUD): a randomised delayed-start trial. Lancet Neurol 2013; 12: 74755.Google Scholar
NINDS NET-PD Investigators. A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 2006; 66: 66471.Google Scholar
NET-PD. Study News. Statement on the Termination of NET-PD LS-1 Study. Available at: http://parkinsontrial.ninds.nih.gov/netpd-LS1-study-termination.htm.Google Scholar
Parkinson Study Group PRECEPT Investigators. Mixed lineage kinase inhibitor CEP-1347 fails to delay disability in early Parkinson disease. Neurology 2007; 69: 148090.Google Scholar
Olanow, CW, Schapira, AHV, LeWitt, PA, et al. TCH346 as a neuroprotective drug in Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol 2006; 5: 101320.Google Scholar
Nutt, JG, Burchiel, KJ, Comella, CL, et al. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology 2003; 60: 6973.Google Scholar
Lang, AE, Gill, S, Patel, NK, et al. Randomized controlled trial of intraputamenal glial cell line-derived neurotrophic factor infusion in Parkinson disease. Ann Neurol 2006; 59: 45966.CrossRefGoogle ScholarPubMed
Marks, WJ, Bartus, RT, Siffert, J, et al. Gene delivery of AAV2-neurturin for Parkinson's disease: a double-blind, randomised, controlled trial. Lancet Neurol 2010; 9: 116472.Google Scholar
Bartus, RT, Baumann, TL, Siffert, J, et al. Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients. Neurology 2013; 80: 1698701.Google Scholar
PR Newswire. Ceregene reports data from Parkinson's disease phase 2b study. Available at: http://www.prnewswire.com/news-releases/ceregene-reports-data-from-parkinsons-disease-phase-2b-study-203803541.html.Google Scholar
Kordower, JH, Olanow, CW, Dodiya, HB, et al. Disease duration and the integrity of the nigrostriatal system in Parkinson's disease. Brain 2013; 136: 241931.Google Scholar
Liu, J, Wang, L, Zhan, SY, Xia, Y. Coenzyme Q10 for Parkinson's disease. Cochrane Database Syst Rev 2011; (12): CD008150.Google Scholar
National Institute of Neurological Disorders and Stroke. Statement on Termination of QE3 Study. Available at: http://www.ninds.nih.gov/disorders/clinical_trials/CoQ10-Trial-Update.htm.Google Scholar
Snow, BJ, Rolfe, FL, Lockhart, MM, et al. A double-blind, placebo-controlled study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying therapy in Parkinson's disease. Mov Disord 2010; 25: 16704.Google Scholar

References

Dalaker, TO, Larsen, JP, Bergsland, N, et al. Brain atrophy and white matter hyperintesisites in early Parkisnon's disease. Mov Disord 2009; 24: 223341.Google Scholar
Jakobson, S, Linder, J, Forsgren, L, et al. Pre- and postsynaptic dopamine SPECT in early phase of idiopathic parkinsonism: a population-based study. Eur J Nucl Med Mol Imaging 2010; 37: 215464.Google Scholar
Laere, KV, Clerinx, K, D'Hondt, E, de Groot, T, Vandenberghe, W. Combine striatal binding and cerebral influx analysis of dynamic 11C-raclopride PET improves early differentiation between multiple-system atrophy and Parkinson disease. J Nucl Med 2010; 51: 58895.Google Scholar
Evatt, ML, DeLong, MR, Kumari, M, et al. High prevalence of hypovitaminosis D status in patient with early Parkinson disease. Arch Neurol 2011; 68: 31419.Google Scholar
Shiraishi, M, Kobayashi, T, Watanabe, H, Kamo, , Hasegawa, Y. Serum somatostatin in early stage Parkinson's disease. Acta Neurol Scand 2010; 121: 2259.Google Scholar
Charles, PD, Dolhun, RM, Gill, CE, et al. Deep brain stimulation in early Parkinson's disease: Enrollment experience from a pilot trial. Parkinsonism Relat Disord 2012; 18: 26873.Google Scholar
Gill, CE, Allen, LA, Konrad, PE, et al. Deep brain stimulation of early-stage Parkinson's disease: an illustrative case. Neuromodulation 2011; 14: 51522.Google Scholar
Phillips, L, Litcofsky, KA, Pelster, M, et al. Subthalamic nucleus deep brain stimulation impacts language in early Parkinson's disease. PLoS One 2012; 7: e42829.Google Scholar
Kahn, E, D’Haese, PF, Dawant, B, et al. Deep brain stimulation in early stage Parkinson's disease: operative experience from a prospective randomized clinical trial. J Neurol Neurosurg Psychiatry 2012; 83: 16470.Google Scholar
Shill, HA, Obradov, S, Katsnelson, Y, Pizinger, R. A randomized, double-blind trial of transcranial electrostimulation in early Parkinson's disease. Mov Disord 2011; 26: 147780.Google Scholar
Hauser, RA, Schapira, AHV, Rasciol, O, et al. Randomized double-blind, multicenter evaluation of pramipexole extended release once daily in early Parkinson's disease. Mov Disord 2010; 25: 25429.Google Scholar
Schapira, AHV, Barone, P, Hauser, RA, et al. Patient reported convenience of once-daily versus three-times-daily dosing during long-term studies of pramipexole in early and advance Parkinson's disease. Eur J Neurol 2013; 20: 506.Google Scholar
Poewe, W, Rascol, O, Barone, P, et al. Extended-release pramipexole in early Parkinson disease. Neurology 2011; 75965.Google Scholar
Rascol, O, Barone, P, Hauser, RA, et al. Efficacy, safety and tolerability of overnight switching from immediate-to once daily extended-release pramipexole in early Parkinson's disease. Mov Disord 2010; 25: 232632.Google Scholar
Parkinson Study Group CALM Investigators. Long-term effect of initiating pramipexole vs levodopa in early Parkinson's disease. Arch Neurol 2009; 66: 56370.Google Scholar
Parkinson Study Group PramiBID Investigators. Twice-daily, low-dose parmipexole in early Parkinson's disease: a randomized, placebo-controlled trial. Mov Disord 2011; 26: 3744.Google Scholar
Hauser, RA, Reichmann, H, Lew, M, et al. Long-term, open-label study of once-daily ropinirole prolonged release in early Parkinson's disease. International J Neurosci 2011; 121: 24653.Google Scholar
Watts, RL, Lyons, KE, Pahwa, R, et al. Onset of dyskinesia with adjunct ropinirole prolonged-release or additional levodopa in early Parkinson's disease. Mov Disord 2010; 25: 85866.Google Scholar
Bronzova, J, Sampiao, C, Hauser, RA, et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson's disease. Mov Disord 2010; 25: 73846.Google Scholar
Sampiao, C, Bronzova, J, Hauser, RA, et al. Pardoprunox in early Parkinson's disease: results from 2 large randomized double-blind trials. Mov Disord 2011; 26: 146476.Google Scholar
Shapira, AHV, Stocchi, F, Borgohain, R, et al. Long-term efficacy and safety of safinamide as add-on therapy in early Parkinson's disease. Eur J Neurol 2013; 20: 27180.Google Scholar
Stocchi, F, Borgohain, R, Onofrj, M, et al. A randomized, double-blind, placebo-controlled trial of safinamide as add-on therapy in early Parkinson's disease patients. Mov Disord 2012; 27: 10612.Google Scholar
Simuni, T, Borushko, E, Avram, MJ, et al. Tolerability of isradipine in early Parkinson's disease: a pilot dose escalation study. Mov Disord 2010; 25: 28636.Google Scholar
Viallet, F, Pitel, S, Lancrenon, S, Blin, O. Evaluation of the safety and tolerability of rasagiline in the treatment of the early stages of Parkinson's disease. Curr Med Res Opin 2013; 29: 2331.Google Scholar
Olanow, W, Hauser, RA, Jankovick, J, et al. A randomized, double-blind, placebo-controlled, delayed start study to assess rasagiline as a disease modifying therapy in Parkinson's disease (the ADAGIO study): rationale, design and baseline characteristics. Mov Disord 2008; 23: 2194201.Google Scholar
Olanow, W, Rascol, O, Hauser, RA, et al. A double blind, delayed-start trial of rasagaline in Parkinson's disease. N Engl J Med 2009; 361: 126878.Google Scholar
Hauser, RA, Lew, MF, Hurtig, HI, et al. Long-term outcome of early versus delayed rasagiline treatment in early Parkinson's disease. Mov Disord 2009; 24: 56473.Google Scholar
Schenkman, M, Hall, D, Barón, AE, et al. Exercise for people in early- or mid-stage Parkinson disease: a 16-month randomized controlled trial. Phys Ther 2012; 92: 1395410.Google Scholar

References

Weaver, FM, Follett, K, Stern, M, et al. Bilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson disease: a randomized controlled trial. JAMA 2009; 301: 6373.Google Scholar
Siegfried, J, Lippitz, B. Bilateral chronic electrostimulation of ventroposterolateral pallidum: a new therapeutic approach for alleviating all parkinsonian symptoms. Neurosurgery 1994; 35: 11269; discussion 1129–30.Google Scholar
Pollak, P, Benabid, AL, Gross, C, et al. [Effects of the stimulation of the subthalamic nucleus in Parkinson disease]. Rev Neurol (Paris) 1993; 149: 1756 (in French).Google Scholar
Boucai, L, Cerquetti, D, Merello, M. Functional surgery for Parkinson's disease treatment: a structured analysis of a decade of published literature. Br J Neurosurg 2004; 18: 21322.Google Scholar
Gross, RE, Krack, P, Rodriguez-Oroz, MC, Rezai, AR, Benabid, AL. Electrophysiological mapping for the implantation of deep brain stimulators for Parkinson's disease and tremor. Mov Disord 2006; 21 (Suppl. 14): S259283.Google Scholar
Bour, LJ, Contarino, MF, Foncke, EM, et al. Long-term experience with intraoperative microrecording during DBS neurosurgery in STN and GPi. Acta Neurochir (Wien) 2010; 152: 206977.Google Scholar
Deep-Brain Stimulation for Parkinson's Disease Study Group. Deep-brain stimulation of the subthalamic nucleus or the pars interna of the globus pallidus in Parkinson's disease. N Engl J Med 2001; 345: 95663.Google Scholar
Cleary, DR, Raslan, AM, Rubin, JE, et al. Deep brain stimulation entrains local neuronal firing in human globus pallidus internus. J Neurophysiol 2013; 109: 97887.Google Scholar
Lai, HY, Younce, JR, Albaugh, DL, Kao, YC, Shih, YY. Functional MRI reveals frequency-dependent responses during deep brain stimulation at the subthalamic nucleus or internal globus pallidus. Neuroimage 2013; 84: 1118.Google Scholar
Vitek, JL. Deep brain stimulation for Parkinson's disease. A critical re-evaluation of STN versus GPi-DBS. Stereotact Funct Neurosurg 2002; 78: 11931.Google Scholar
Okun, MS, Foote, KD. Subthalamic nucleus vs globus pallidus interna deep brain stimulation, the rematch: will pallidal deep brain stimulation make a triumphant return? Arch Neurol 2005; 62: 5336.Google Scholar
Mann, JM, Foote, KD, Garvan, CW, et al. Brain penetration effects of microelectrodes and DBS leads in STN or GPi. J Neurol Neurosurg Psychiatry 2009; 80: 7947.Google Scholar
Benabid, AL, Pollak, P, Gross, C, et al. Acute and long-term effects of subthalamic nucleus stimulation in Parkinson's disease. Stereotact Funct Neurosurg 1994; 62: 7684.Google Scholar
Ghika, J, Villemure, JG, Fankhauser, H, et al. Efficiency and safety of bilateral contemporaneous pallidal stimulation (deep brain stimulation) in levodopa-responsive patients with Parkinson's disease with severe motor fluctuations: a 2-year follow-up review. J Neurosurg 1998; 89: 71318.Google Scholar
Keitel, A, Ferrea, S, Sudmeyer, M, Schnitzler, A, Wojtecki, L. Expectation modulates the effect of deep brain stimulation on motor and cognitive function in tremor-dominant Parkinson's disease. PLoS One 2013; 8: e81878.Google Scholar
Anderson, VC, Burchiel, KJ, Hogarth, P, Favre, J, Hammerstad, JP. Pallidal vs subthalamic nucleus deep brain stimulation in Parkinson disease. Arch Neurol 2005; 62: 55460.Google Scholar
Krack, P, Batir, A, Van Blercom, N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson's disease. N Engl J Med 2003; 349: 192534.Google Scholar
Nakamura, K, Christine, CW, Starr, PA, Marks, WJ Jr. Effects of unilateral subthalamic and pallidal deep brain stimulation on fine motor functions in Parkinson's disease. Mov Disord 2007; 22: 61926.Google Scholar
Krack, P, Pollak, P, Limousin, P, et al. Subthalamic nucleus or internal pallidal stimulation in young onset Parkinson's disease. Brain 1998; 121: 4517.Google Scholar
Robertson, LT, St George, RJ, Carlson-Kuhta, P, et al. Site of deep brain stimulation and jaw velocity in Parkinson disease. J Neurosurg 2011; 115: 98594.Google Scholar
St George, RJ, Nutt, JG, Burchiel, KJ, Horak, FB. A meta-regression of the long-term effects of deep brain stimulation on balance and gait in PD. Neurology 2010; 75: 12929.Google Scholar
Rodriguez-Oroz, MC, Obeso, JA, Lang, AE, et al. Bilateral deep brain stimulation in Parkinson's disease: a multicentre study with 4 years follow-up. Brain 2005; 128: 22409.Google Scholar
Follett, KA, Weaver, FM, Stern, M, et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson's disease. N Engl J Med 2010; 362: 207791.Google Scholar
Follett, KA. Comparison of pallidal and subthalamic deep brain stimulation for the treatment of levodopa-induced dyskinesias. Neurosurg Focus 2004; 17: E3.Google Scholar
Volkmann, J. Deep brain stimulation for the treatment of Parkinson's disease. J Clin Neurophysiol 2004; 21: 617.Google Scholar
Kumar, N, Van Gerpen, JA, Bower, JH, Ahlskog, JE. Levodopa-dyskinesia incidence by age of Parkinson's disease onset. Mov Disord 2005; 20: 3424.Google Scholar
Krause, M, Fogel, W, Heck, A, et al. Deep brain stimulation for the treatment of Parkinson's disease: subthalamic nucleus versus globus pallidus internus. J Neurol Neurosurg Psychiatry 2001; 70: 46470.Google Scholar
Minguez-Castellanos, A, Escamilla-Sevilla, F, Katati, MJ, et al. Different patterns of medication change after subthalamic or pallidal stimulation for Parkinson's disease: target related effect or selection bias? J Neurol Neurosurg Psychiatry 2005; 76: 349.Google Scholar
Peppe, A, Pierantozzi, M, Bassi, A, et al. Stimulation of the subthalamic nucleus compared with the globus pallidus internus in patients with Parkinson disease. J Neurosurg 2004; 101: 195200.Google Scholar
Odekerken, VJ, van Laar, T, Staal, MJ, et al. Subthalamic nucleus versus globus pallidus bilateral deep brain stimulation for advanced Parkinson's disease (NSTAPS study): a randomised controlled trial. Lancet Neurol 2013; 12: 3744.Google Scholar
Moro, E, Lozano, AM, Pollak, P, et al. Long-term results of a multicenter study on subthalamic and pallidal stimulation in Parkinson's disease. Mov Disord 2010; 25: 57886.Google Scholar
Weaver, FM, Follett, KA, Stern, M, et al. Randomized trial of deep brain stimulation for Parkinson disease: thirty-six-month outcomes. Neurology 2012; 79: 5565.Google Scholar
Weaver, F, Follett, K, Hur, K, Ippolito, D, Stern, M. Deep brain stimulation in Parkinson disease: a metaanalysis of patient outcomes. J Neurosurg 2005; 103: 95667.Google Scholar
Borgohain, R, Kandadai, RM, Jabeen, A, Kannikannan, MA. Nonmotor outcomes in Parkinson's disease: is deep brain stimulation better than dopamine replacement therapy? Ther Adv Neurol Disord 2012; 5: 2341.Google Scholar
Yaguez, L, Costello, A, Moriarty, J, et al. Cognitive predictors of cognitive change following bilateral subthalamic nucleus deep brain stimulation in Parkinson's disease. J Clin Neurosci 2014; 21: 44550.Google Scholar
Burdick, AP, Foote, KD, Wu, S, et al. Do patient's get angrier following STN, GPi, and thalamic deep brain stimulation. Neuroimage 2011; 54 (Suppl. 1): S22732.Google Scholar
Okun, MS, Fernandez, HH, Wu, SS, et al. Cognition and mood in Parkinson's disease in subthalamic nucleus versus globus pallidus interna deep brain stimulation: the COMPARE trial. Ann Neurol 2009; 65: 58695.Google Scholar
Jahanshahi, M, Ardouin, CM, Brown, RG, et al. The impact of deep brain stimulation on executive function in Parkinson's disease. Brain 2000; 123: 114254.Google Scholar
Limousin, P, Greene, J, Pollak, P, et al. Changes in cerebral activity pattern due to subthalamic nucleus or internal pallidum stimulation in Parkinson's disease. Ann Neurol 1997; 42: 28391.Google Scholar
Volkmann, J, Allert, N, Voges, J, et al. Safety and efficacy of pallidal or subthalamic nucleus stimulation in advanced PD. Neurology 2001; 56: 54851.Google Scholar
Hariz, MI, Rehncrona, S, Quinn, NP, et al. Multicenter study on deep brain stimulation in Parkinson's disease: an independent assessment of reported adverse events at 4 years. Mov Disord 2008; 23: 41621.Google Scholar
Zahodne, LB, Okun, MS, Foote, KD, et al. Greater improvement in quality of life following unilateral deep brain stimulation surgery in the globus pallidus as compared to the subthalamic nucleus. J Neurol 2009; 256: 13219.Google Scholar
Weaver, FM, Stroupe, KT, Cao, L, et al. Parkinson's disease medication use and costs following deep brain stimulation. Mov Disord 2012; 27: 1398403.Google Scholar
Evidente, VG, Premkumar, AP, Adler, CH, et al. Medication dose reductions after pallidal versus subthalamic stimulation in patients with Parkinson's disease. Acta Neurol Scand 2011; 124: 21114.Google Scholar
Schupbach, WM, Chastan, N, Welter, ML, et al. Stimulation of the subthalamic nucleus in Parkinson's disease: a 5 year follow-up. J Neurol Neurosurg Psychiatry 2005; 76: 16404.Google Scholar
Burchiel, KJ, McCartney, S, Lee, A, Raslan, AM. Accuracy of deep brain stimulation electrode placement using intraoperative computed tomography without microelectrode recording. J Neurosurg 2013; 119: 3016.Google Scholar
O'Gorman, RL, Shmueli, K, Ashkan, K, et al. Optimal MRI methods for direct stereotactic targeting of the subthalamic nucleus and globus pallidus. Eur Radiol 2011; 21: 1306.Google Scholar
Khan, FR, Henderson, JM. Deep brain stimulation surgical techniques. Handb Clin Neurol 2013; 116: 2737.Google Scholar
Holloway, KL, Gaede, SE, Starr, PA, et al. Frameless stereotaxy using bone fiducial markers for deep brain stimulation. J Neurosurg 2005; 103: 40413.Google Scholar
Bjartmarz, H, Rehncrona, S. Comparison of accuracy and precision between frame-based and frameless stereotactic navigation for deep brain stimulation electrode implantation. Stereotact Funct Neurosurg 2007; 85: 23542.Google Scholar
Volkmann, J, Moro, E, Pahwa, R. Basic algorithms for the programming of deep brain stimulation in Parkinson's disease. Mov Disord 2006; 21 (Suppl. 14): S2849.Google Scholar
Broen, M, Duits, A, Visser-Vandewalle, V, Temel, Y, Winogrodzka, A. Impulse control and related disorders in Parkinson's disease patients treated with bilateral subthalamic nucleus stimulation: a review. Parkinsonism Relat Disord 2011; 17: 41317.Google Scholar
Binder, DK, Rau, GM, Starr, PA. Risk factors for hemorrhage during microelectrode-guided deep brain stimulator implantation for movement disorders. Neurosurgery 2005; 56: 72232; discussion 722–32.Google Scholar
Ben-Haim, S, Asaad, WF, Gale, JT, Eskandar, EN. Risk factors for hemorrhage during microelectrode-guided deep brain stimulation and the introduction of an improved microelectrode design. Neurosurgery 2009; 64: 75462; discussion 762–3.Google Scholar
Volkmann, J, Albanese, A, Kulisevsky, J, et al. Long-term effects of pallidal or subthalamic deep brain stimulation on quality of life in Parkinson's disease. Mov Disord 2009; 24: 115461.Google Scholar
Sauleau, P, Leray, E, Rouaud, T, et al. Comparison of weight gain and energy intake after subthalamic versus pallidal stimulation in Parkinson's disease. Mov Disord 2009; 24: 214955.Google Scholar
Mills, KA, Scherzer, R, Starr, PA, Ostrem, JL. Weight change after globus pallidus internus or subthalamic nucleus deep brain stimulation in Parkinson's disease and dystonia. Stereotact Funct Neurosurg 2012; 90: 38693.Google Scholar
Rieu, I, Derost, P, Ulla, M, et al. Body weight gain and deep brain stimulation. J Neurol Sci 2011; 310: 26770.Google Scholar

References

Horsley, V. The Linacre Lecture on the function of the so-called motor area of the brain: delivered to the master and fellows of St. John's College, Cambridge, May 6th, 1909. Br Med J 1909; 2: 12132.Google Scholar
Spiegel, EA, Wycis, HT, Marks, M, Lee, AJ. Stereotaxic apparatus for operations on the human brain. Science 1947; 106: 34950.Google Scholar
Spiegel, EA, Wycis, HT, Baird, HW 3rd. Long-range effects of electropallidoansotomy in extrapyramidal and convulsive disorders. Neurology 1958; 8: 73440.Google Scholar
Meyers, R. Surgical experiments in the therapy of certain ‘extrapyramidal’ diseases: a current evaluation. Acta Psychiatr Neurol Suppl 1951; 67: 142.Google Scholar
Cooper, IS. Ligation of the anterior choroidal artery for involuntary movements; parkinsonism. Psychiatr Q 1953; 27: 31719.Google Scholar
Cooper, IS. Surgical alleviation of parkinsonism; effects of occlusion of the anterior choroidal artery. J Am Geriatr Soc. 1954; 2: 691718.Google Scholar
Guiot, G, Brion, S. Treatment of abnormal movement by pallidal coagulation. Rev Neurol (Paris) 1953; 89: 57880.Google Scholar
Narabayashi, H, Okuma, T. Procaine oil blocking of the globus pallidus for treatment of rigidity and tremor of parkinsonism: preliminary report. Proc Jpn Acad; 1953; 2 Procaine oil blocking 9: 134.Google Scholar
Hassler, R. The influence of stimulations and coagulations in the human thalamus on the tremor at rest and its physiopathologic mechanism. Proc Second Int Cong Neuropathol 1955; 2: 63742.Google Scholar
Laitinen, LV. Brain targets in surgery for Parkinson's disease. Results of a survey of neurosurgeons. J Neurosurg 1985; 62: 34951.Google Scholar
Svennilson, E, Torvik, A, Lowe, R, Leksell, L. Treatment of parkinsonism by stereotatic thermolesions in the pallidal region. A clinical evaluation of 81 cases. Acta Psychiatr Scand 1960; 35: 35877.Google Scholar
Yahr, MD. Clinical aspects of abnormal movements induced by L-dopa. In: Barbeau, A, McDowell, FH, eds. L-Dopa and Parkinsonism. Davis Company; 1970; 1018.Google Scholar
Barbeau, A. The clinical physiology of side effects in long-term l-DOPA therapy. Adv Neurol 1974; 5: 34765.Google Scholar
Laitinen, LV, Bergenheim, AT, Hariz, MI. Leksell's posteroventral pallidotomy in the treatment of Parkinson's disease. J Neurosurg 1992; 76: 5361.Google Scholar
Benabid, AL, Pollak, P, Louveau, A, Henry, S, de Rougemont, J. Combined (thalamotomy and stimulation) stereotactic surgery of the VIM thalamic nucleus for bilateral Parkinson disease. Appl Neurophysiol 1987; 50: 3446.Google Scholar
Albin, RL, Young, AB, Penney, JB. The functional anatomy of basal ganglia disorders. Trends Neurosci 1989; 12: 36675.Google Scholar
Bergman, H, Wichmann, T, DeLong, MR. Reversal of experimental parkinsonism by lesions of the subthalamic nucleus. Science 1990; 249: 14368.Google Scholar
Pollak, P, Benabid, AL, Gross, C, et al. Effects of the stimulation of the subthalamic nucleus in Parkinson disease. Rev Neurol (Paris) 1993; 149: 1756.Google Scholar
Limousin, P, Krack, P, Pollak, P, et al. Electrical stimulation of the subthalamic nucleus in advanced Parkinson's disease. N Engl J Med 1998; 339: 110511.Google Scholar
Siegfried, J, Lippitz, B. Bilateral chronic electrostimulation of ventroposterolateral pallidum: a new therapeutic approach for alleviating all parkinsonian symptoms. Neurosurgery 1994; 35: 11269; discussion 1129–30.Google Scholar
Vitek, JL, Bakay, RA, Hashimoto, T, et al. Microelectrode-guided pallidotomy: technical approach and its application in medically intractable Parkinson's disease. J Neurosurg 1998; 88: 102743.Google Scholar
Kaplitt, MG, Hutchison, WD, Lozano, AM. Target localization in movement disorders surgery. In: Tarsy, D, Vitek, JL, Lozano, AM, eds. Surgical Treatment of Parkinson's Disease. Humana Press; 2003; 8798.Google Scholar
Schuurman, PR, Bosch, DA, Merkus, MP, Speelman, JD. Long-term follow-up of thalamic stimulation versus thalamotomy for tremor suppression. Mov Disord 2008; 23: 114653.Google Scholar
Loher, TJ, Pohle, T, Krauss, JK. Functional stereotactic surgery for treatment of cervical dystonia: review of the experience from the lesional era. Stereotact Funct Neurosurg 2004; 82: 113.Google Scholar
Krayenbuhl, H, Wyss, OA, Yasargil, MG. Bilateral thalamotomy and pallidotomy as treatment for bilateral parkinsonism. J Neurosurg 1961; 18: 42944.Google Scholar
Moriyama, E, Beck, H, Miyamoto, T. Long-term results of ventrolateral thalamotomy for patients with Parkinson's disease. Neurol Med Chir (Tokyo) 1999; 39: 3506; discussion 356–7.Google Scholar
Jankovic, J, Cardoso, F, Grossman, RG, Hamilton, WJ. Outcome after stereotactic thalamotomy for parkinsonian, essential, and other types of tremor. Neurosurgery 1995; 37: 6806; discussion 686–7.Google Scholar
Wester, K, Hauglie-Hanssen, E. Stereotaxic thalamotomy – experiences from the levodopa era. J Neurol Neurosurg Psychiatry 1990; 53: 42730.Google Scholar
Hallett, M, Litvan, I. Evaluation of surgery for Parkinson's disease: a report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. The Task Force on Surgery for Parkinson's Disease. Neurology 1999; 53: 191021.Google Scholar
Blomstedt, P, Hariz, GM, Hariz, MI. Pallidotomy versus pallidal stimulation. Parkinsonism Relat Disord 2006; 12: 296301.Google Scholar
de Bie, RM, de Haan, RJ, Nijssen, PC, et al. Unilateral pallidotomy in Parkinson's disease: a randomised, single-blind, multicentre trial. Lancet 1999; 354: 16659.Google Scholar
Vitek, JL, Bakay, RA, Freeman, A, et al. Randomized trial of pallidotomy versus medical therapy for Parkinson's disease. Ann Neurol 2003; 53: 55869.Google Scholar
Kondziolka, D, Bonaroti, E, Baser, S, et al. Outcomes after stereotactically guided pallidotomy for advanced Parkinson's disease. J Neurosurg 1999; 90: 197202.Google Scholar
Giller, CA, Dewey, RB, Ginsburg, MI, Mendelsohn, DB, Berk, AM. Stereotactic pallidotomy and thalamotomy using individual variations of anatomic landmarks for localization. Neurosurgery 1998; 42: 5662; discussion 62–5.Google Scholar
Shannon, KM, Penn, RD, Kroin, JS, et al. Stereotactic pallidotomy for the treatment of Parkinson's disease. Efficacy and adverse effects at 6 months in 26 patients. Neurology 1998; 50: 4348.Google Scholar
Kishore, A, Turnbull, IM, Snow, BJ, et al. Efficacy, stability and predictors of outcome of pallidotomy for Parkinson's disease. six-month follow-up with additional 1-year observations. Brain 1997; 120: 72937.Google Scholar
Krauss, JK, Desaloms, JM, Lai, EC, et al. Microelectrode-guided posteroventral pallidotomy for treatment of Parkinson's disease: postoperative magnetic resonance imaging analysis. J Neurosurg 1997; 87: 35867.Google Scholar
Lang, AE, Lozano, AM, Montgomery, E, et al. Posteroventral medial pallidotomy in advanced Parkinson's disease. N Engl J Med 1997; 337: 103642.Google Scholar
Kazumata, K, Antonini, A, Dhawan, V, et al. Preoperative indicators of clinical outcome following stereotaxic pallidotomy. Neurology 1997; 49: 108390.Google Scholar
Uitti, RJ, Wharen, RE Jr, Turk, MF, et al. Unilateral pallidotomy for Parkinson's disease: comparison of outcome in younger versus elderly patients. Neurology 1997; 49: 10727.Google Scholar
Trepanier, LL, Saint-Cyr, JA, Lozano, AM, Lang, AE. Neuropsychological consequences of posteroventral pallidotomy for the treatment of Parkinson's disease. Neurology 1998; 51: 20715.Google Scholar
Blomstedt, P, Hariz, MI. Are complications less common in deep brain stimulation than in ablative procedures for movement disorders? Stereotact Funct Neurosurg 2006; 84: 7281.Google Scholar
Alkhani, A, Lozano, AM. Pallidotomy for Parkinson disease: a review of contemporary literature. J Neurosurg 2001; 94: 439.Google Scholar
Perrine, K, Dogali, M, Fazzini, E, et al. Cognitive functioning after pallidotomy for refractory Parkinson's disease. J Neurol Neurosurg Psychiatry 1998; 65: 1504.Google Scholar
Biousse, V, Newman, NJ, Carroll, C, et al. Visual fields in patients with posterior GPi pallidotomy. Neurology 1998; 50: 25865.Google Scholar
Hariz, MI, De Salles, AA. The side-effects and complications of posteroventral pallidotomy. Acta Neurochir Suppl 1997; 68: 428.Google Scholar
Zahodne, LB, Okun, MS, Foote, KD, et al. Greater improvement in quality of life following unilateral deep brain stimulation surgery in the globus pallidus as compared to the subthalamic nucleus. J Neurol 2009; 256: 13219.Google Scholar
Rodrigues, JP, Walters, SE, Watson, P, Stell, R, Mastaglia, FL. Globus pallidus stimulation improves both motor and nonmotor aspects of quality of life in advanced Parkinson's disease. Mov Disord 2007; 22: 186670.Google Scholar
Germano, IM, Gracies, JM, Weisz, DJ, et al. Unilateral stimulation of the subthalamic nucleus in Parkinson disease: a double-blind 12-month evaluation study. J Neurosurg 2004; 101: 3642.Google Scholar
Mikos, A, Zahodne, L, Okun, MS, Foote, K, Bowers, D. Cognitive declines after unilateral deep brain stimulation surgery in Parkinson's disease: a controlled study using reliable change, part II. Clin Neuropsychol 2010; 24: 23545.Google Scholar
Okun, MS, Fernandez, HH, Wu, SS, et al. Cognition and mood in Parkinson's disease in subthalamic nucleus versus globus pallidus interna deep brain stimulation: the COMPARE trial. Ann Neurol 2009; 65: 58695.Google Scholar
Scott, R, Gregory, R, Hines, N, et al. Neuropsychological, neurological and functional outcome following pallidotomy for Parkinson's disease. A consecutive series of eight simultaneous bilateral and twelve unilateral procedures. Brain 1998; 121: 65975.Google Scholar
Intemann, PM, Masterman, D, Subramanian, I, et al. Staged bilateral pallidotomy for treatment of Parkinson disease. J Neurosurg 2001; 94: 43744.Google Scholar
Counihan, TJ, Shinobu, LA, Eskandar, EN, Cosgrove, GR, Penney, JB Jr. Outcomes following staged bilateral pallidotomy in advanced Parkinson's disease. Neurology 2001; 56: 799802.Google Scholar
Iacono, RP, Lonser, RR, Yamada, S. Contemporaneous bilateral postero-ventral pallidotomy for early onset “juvenile type” Parkinson's disease. Case report. Acta Neurochir (Wien) 1994; 131: 24752.Google Scholar
Iacono, RP, Shima, F, Lonser, RR, et al. The results, indications, and physiology of posteroventral pallidotomy for patients with Parkinson's disease. Neurosurgery 1995; 36: 111825; discussion 1125–7.Google Scholar
Vitek, JL, Bakay, RA, DeLong, MR. Microelectrode-guided pallidotomy for medically intractable Parkinson's disease. Adv Neurol 1997; 74: 18398.Google Scholar
Favre, J, Burchiel, KJ, Taha, JM, Hammerstad, J. Outcome of unilateral and bilateral pallidotomy for Parkinson's disease: patient assessment. Neurosurgery 2000; 46: 34453; discussion 353–5.Google Scholar
Siegel, KL, Metman, LV. Effects of bilateral posteroventral pallidotomy on gait of subjects with Parkinson disease. Arch Neurol 2000; 57: 198204.Google Scholar
de Bie, RM, Schuurman, PR, Esselink, RA, Bosch, DA, Speelman, JD. Bilateral pallidotomy in Parkinson's disease: a retrospective study. Mov Disord 2002; 17: 5338.Google Scholar
Hua, Z, Guodong, G, Qinchuan, L, et al. Analysis of complications of radiofrequency pallidotomy. Neurosurgery 2003; 52: 8999; discussion 99–101.Google Scholar
York, MK, Lai, EC, Jankovic, J, et al. Short and long-term motor and cognitive outcome of staged bilateral pallidotomy: a retrospective analysis. Acta Neurochir (Wien) 2007; 149: 85766; discussion 866.Google Scholar
Movement Disorder Society Task Force. Management of Parkinson's disease: an evidence-based review. Mov Disord 2002; 17 (Suppl. 4): S1166.Google Scholar
Merello, M, Starkstein, S, Nouzeilles, MI, Kuzis, G, Leiguarda, R. Bilateral pallidotomy for treatment of Parkinson's disease induced corticobulbar syndrome and psychic akinesia avoidable by globus pallidus lesion combined with contralateral stimulation. J Neurol Neurosurg Psychiatry 2001; 71: 61114.Google Scholar
Parkin, SG, Gregory, RP, Scott, R, et al. Unilateral and bilateral pallidotomy for idiopathic Parkinson's disease: a case series of 115 patients. Mov Disord 2002; 17: 68292.Google Scholar
de Bie, RM, Schuurman, PR, Esselink, RA, Bosch, DA, Speelman, JD. Bilateral pallidotomy in Parkinson's disease: a retrospective study. Mov Disord 2002; 17: 5338.Google Scholar
de Bie, RM, de Haan, RJ, Schuurman, PR, et al. Morbidity and mortality following pallidotomy in Parkinson's disease: a systematic review. Neurology 2002; 58: 100812.Google Scholar
Deep-Brain Stimulation for Parkinson's Disease Study Group. Deep-brain stimulation of the subthalamic nucleus or the pars interna of the globus pallidus in Parkinson's disease. N Engl J Med 2001; 345: 95663.Google Scholar
Anderson, VC, Burchiel, KJ, Hogarth, P, Favre, J, Hammerstad, JP. Pallidal vs subthalamic nucleus deep brain stimulation in Parkinson disease. Arch Neurol 2005; 62: 55460.Google Scholar
Follett, KA, Weaver, FM, Stern, M, et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson's disease. N Engl J Med 2010; 362: 207791.Google Scholar
Odekerken, VJ, van Laar, T, Staal, MJ, et al. Subthalamic nucleus versus globus pallidus bilateral deep brain stimulation for advanced Parkinson's disease (NSTAPS study): a randomised controlled trial. Lancet Neurol 2013; 12: 3744.Google Scholar
Coban, A, Hanagasi, HA, Karamursel, S, Barlas, O. Comparison of unilateral pallidotomy and subthalamotomy findings in advanced idiopathic Parkinson's disease. Br J Neurosurg 2009; 23: 239.Google Scholar
Alvarez, L, Macias, R, Pavon, N, et al. Therapeutic efficacy of unilateral subthalamotomy in Parkinson's disease: results in 89 patients followed for up to 36 months. J Neurol Neurosurg Psychiatry 2009; 80: 97985.Google Scholar
Slowinski, JL, Putzke, JD, Uitti, RJ, et al. Unilateral deep brain stimulation of the subthalamic nucleus for Parkinson disease. J Neurosurg 2007; 106: 62632.Google Scholar
Linazasoro, G, Van Blercom, N, Lasa, A. Unilateral subthalamic deep brain stimulation in advanced Parkinson's disease. Mov Disord 2003; 18: 71316.Google Scholar
Merello, M, Tenca, E, Perez Lloret, S, et al. Prospective randomized 1-year follow-up comparison of bilateral subthalamotomy versus bilateral subthalamic stimulation and the combination of both in Parkinson's disease patients: a pilot study. Br J Neurosurg 2008; 22: 41522.Google Scholar
Alvarez, L, Macias, R, Lopez, G, et al. Bilateral subthalamotomy in Parkinson's disease: initial and long-term response. Brain 2005; 128: 57083.Google Scholar
Deuschl, G, Schade-Brittinger, C, Krack, P, et al. A randomized trial of deep-brain stimulation for Parkinson's disease. N Engl J Med 2006; 355: 896908.Google Scholar
Schupbach, WM, Maltete, D, Houeto, JL, et al. Neurosurgery at an earlier stage of Parkinson disease: a randomized, controlled trial. Neurology 2007; 68: 26771.Google Scholar
Williams, A, Gill, S, Varma, T, et al. Deep brain stimulation plus best medical therapy versus best medical therapy alone for advanced Parkinson's disease (PD SURG trial): a randomised, open-label trial. Lancet Neurol 2010; 9: 58191.Google Scholar
Krack, P, Batir, A, Van Blercom, N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson's disease. N Engl J Med 2003; 349: 192534.Google Scholar
Weaver, FM, Follett, K, Stern, M, et al. Bilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson disease: a randomized controlled trial. JAMA 2009; 301: 6373.Google Scholar
Witt, K, Daniels, C, Reiff, J, et al. Neuropsychological and psychiatric changes after deep brain stimulation for Parkinson's disease: a randomised, multicentre study. Lancet Neurol 2008; 7: 60514.Google Scholar
Esselink, RA, de Bie, RM, de Haan, RJ, et al. Long-term superiority of subthalamic nucleus stimulation over pallidotomy in Parkinson disease. Neurology 2009; 73: 1513.Google Scholar
Drapier, D, Drapier, S, Sauleau, P, et al. Does subthalamic nucleus stimulation induce apathy in Parkinson's disease? J Neurol 2006; 253: 108391.Google Scholar
Lim, SY, O'sullivan, SS, Kotschet, K, et al. Dopamine dysregulation syndrome, impulse control disorders and punding after deep brain stimulation surgery for Parkinson's disease. J Clin Neurosci 2009; 16: 114852.Google Scholar
Voon, V, Krack, P, Lang, AE, et al. A multicentre study on suicide outcomes following subthalamic stimulation for Parkinson's disease. Brain 2008; 131: 27208.Google Scholar
Bakay, RA, DeLong, MR, Vitek, JL. Posteroventral pallidotomy for Parkinson's disease. J Neurosurg 1992; 77: 4878.Google Scholar
Van Buren, JM, Li, CL, Shapiro, DY, Henderson, WG, Sadowsky, DA. A qualitative and quantitative evaluation of parkinsonians three to six years following thalamotomy. Confin Neurol 1973; 35: 20235.Google Scholar
Fox, SH, Katzenschlager, R, Lim, SY, et al. The Movement Disorder Society Evidence-based Medicine Review Update: treatments for the motor symptoms of Parkinson's disease. Mov Disord 2011; 26 (Suppl. 3): S241.Google Scholar
Hooper, AK, Okun, MS, Foote, KD, et al. Clinical cases where lesion therapy was chosen over deep brain stimulation. Stereotact Funct Neurosurg 2008; 86: 14752.Google Scholar
Elias, WJ, Huss, D, Voss, T, et al. A pilot study of focused ultrasound thalamotomy for essential tremor. N Engl J Med 2013; 369: 6408.Google Scholar

References

Obeso, JA, Rodriguez-Oroz, MC, Chana, P, et al. The evolution and origin of motor complications in Parkinson's disease. Neurology 2000; 55 (Suppl. 4): S1320; discussion S21–3.Google Scholar
Deuschl, G, Schade-Brittinger, C, Krack, P. A randomized trial of deep-brain stimulation for Parkinson's disease. N Engl J Med 2006; 355: 896908.Google Scholar
Williams, A, Gill, S, Varma, T, et al. Deep brain stimulation plus best medical therapy versus best medical therapy alone for advanced Parkinson's disease (PD SURG trial): a randomised, open-label trial. Lancet Neurol 2010; 9: 58191.Google Scholar
Follett, KA, Weaver, FM, Stern, M, et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson's disease. N Engl J Med 2010; 362: 207791.Google Scholar
Okun, MS, Gallo, BV, Mandybur, G, et al. Subthalamic deep brain stimulation with a constant-current device in Parkinson's disease: an open-label randomised controlled trial. Lancet Neurol 2012; 11: 1409.Google Scholar
Odekerken, VJ, van Laar, T, Staal, MJ, et al. Subthalamic nucleus versus globus pallidus bilateral deep brain stimulation for advanced Parkinson's disease (NSTAPS study): a randomised controlled trial. Lancet Neurol 2013; 12: 3744.Google Scholar
Weaver, F, Follett, K, Stern, M, et al. Bilateral deep brain stimulation vs best medical therapy for patients with advanced Parkinson disease: a randomized controlled trial. JAMA 2009; 301: 6373.Google Scholar
Okun, M, Fernandez, H, Wu, S, et al. Cognition and mood in Parkinson's disease in subthalamic nucleus versus globus pallidus interna deep brain stimulation: the COMPARE trial. Ann Neurol 2009; 65: 58695.Google Scholar
Krack, P, Batir, A, van Blercom, N, et al. Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson's disease. N Engl J Med 2003; 349: 192534.Google Scholar
Simonin, C, Tir, M, Devos, D, et al. Reduced levodopa-induced complications after 5 years of subthalamic stimulation in Parkinson's disease: a second honeymoon. J Neurol 2009; 256: 173641.Google Scholar
Schüpbach, WM, Maltête, D, Houeto, JL, et al. Neurosurgery at an earlier stage of Parkinson disease: a randomized, controlled trial. Neurology 2007; 68: 26771.Google Scholar
Charles, PD, Padaliya, BB, Newman, WJ, et al. Deep brain stimulation of the subthalamic nucleus reduces antiparkinsonian medication costs. Parkinsonism Relat Disord 2004; 10: 4759.Google Scholar
Weaver, FM, Stroupe, KT, Cao, L, et al. Parkinson's disease medication use and costs following deep brain stimulation. Mov Disord 2012; 27: 1398403.Google Scholar
Schuepbach, WM, Rau, J, Knudsen, K, et al. Neurostimulation for Parkinson's disease with early motor complications. N Engl J Med 2013; 368: 61022.Google Scholar
Dumitriu, A, Popescu, BO. Placebo effects in neurological diseases. J Med Life 2010; 3: 11421.Google Scholar
Paradis, C. Bias in surgical research. Ann Surg 2008; 248: 1808.Google Scholar
Charles, PD, Konrad, PE, Davis, TL, et al. Subthalamic nucleus deep brain stimulation in early stage Parkinson's disease. Parkinsonism Relat Disord 2014; 20: 7317.Google Scholar
Twelves, D, Perkins, KSM, Counsell, C. Systematic review of incidence studies of Parkinson's disease. Mov Disord 2003; 18: 1931.Google Scholar
Bronstein, JM, Tagliati, M, Alterman, RL, et al. Deep brain stimulation for Parkinson disease: an expert consensus and review of key issues. Arch Neurol 2011; 68: 165.Google Scholar
Shih, LC, Tarsy, D. Deep brain stimulation for the treatment of atypical parkinsonism. Mov Disord 2007; 22: 214955.Google Scholar
Lang, AE, Houeto, J, Krack, P, et al. Deep brain stimulation: preoperative issues. Mov Disord 2006; 21 (Suppl. 14): S17196.Google Scholar
Okun, MS, Rodriguez, RL, Mikos, A, et al. Deep brain stimulation and the role of the neuropsychologist. Clin Neuropsychol 2007; 21: 16289.Google Scholar
Anheim, M, Fraix, V, Chabardès, S, et al. Lifetime of Itrel II pulse generators for subthalamic nucleus stimulation in Parkinson's disease. Mov Disord 2007; 22: 24369.Google Scholar
Funkiewiez, A. Long term effects of bilateral subthalamic nucleus stimulation on cognitive function, mood, and behaviour in Parkinson's disease. J Neurol Neurosurg Psychiatry 2004; 75: 8349.Google Scholar
Parsons, TD, Rogers, SA, Braaten, AJ, Woods, SP, Tröster, AI. Cognitive sequelae of subthalamic nucleus deep brain stimulation in Parkinson's disease: a meta-analysis. Lancet Neurol 2006; 5: 57888.Google Scholar
Rieu, I, Derost, P, Ulla, M, et al. Body weight gain and deep brain stimulation. J Neurolog Sci 2011; 310: 26770.Google Scholar
Voon, V, Kubu, C, Krack, P, Houeto, J, Tröster, AI. Deep brain stimulation: neuropsychological and neuropsychiatric issues. Mov Disord 2006; 21 (Suppl. 14): S30527.Google Scholar
Voon, V, Krack, P, Lang, AE, et al. A multicentre study on suicide outcomes following subthalamic stimulation for Parkinson's disease. Brain 2008; 131: 27208.Google Scholar
Kleiner-Fisman, G, Herzog, J, Fisman, D, et al. Subthalamic nucleus deep brain stimulation: summary and meta-analysis of outcomes. Mov Disord 2006; 21 (Suppl. 14): S290304.Google Scholar
Appel-Cresswell, S, La Fuente-Fernandez de, R, Galley, S, McKeown, MJ. Imaging of compensatory mechanisms in Parkinson’s disease. Curr Opin Neurol 2010; 23: 40712.Google Scholar
Rodriguez-Oroz, MC. Bilateral deep brain stimulation in Parkinson's disease: a multicentre study with 4 years follow-up. Brain 2005; 128: 22409.Google Scholar
Wider, C, Pollo, C, Bloch, J, Burkhard, PR, Vingerhoets, FJG. Long-term outcome of 50 consecutive Parkinson's disease patients treated with subthalamic deep brain stimulation. Parkinsonism Relat Disord 2008; 14: 11419.Google Scholar
Zibetti, M, Merola, A, Rizzi, L,et al. Beyond nine years of continuous subthalamic nucleus deep brain stimulation in Parkinson's disease. Mov Disord 2011; 26: 232734.Google Scholar
Hilker, R, Portman, A, Voges, J, Staal, M. Disease progression continues in patients with advanced Parkinson's disease and effective subthalamic nucleus stimulation. J Neurol 2005; 76: 121721.Google Scholar
Maesawa, S, Kaneoke, Y, Kajita, Y, et al. Long-term stimulation of the subthalamic nucleus in hemiparkinsonian rats: neuroprotection of dopaminergic neurons. J Neurosurg 2004; 100: 67987.Google Scholar
Temel, Y, Visser-Vandewalle, V, Kaplan, S, et al. Protection of nigral cell death by bilateral subthalamic nucleus stimulation. Brain Res 2006; 1120: 1005.Google Scholar
Harnack, D, Meissner, W, Jira, J, et al. Placebo-controlled chronic high-frequency stimulation of the subthalamic nucleus preserves dopaminergic nigral neurons in a rat model of progressive Parkinsonism. Exp Neurol 2008; 210: 25760.Google Scholar
Wallace, BA, Ashkan, K, Heise, CE, et al. Survival of midbrain dopaminergic cells after lesion or deep brain stimulation of the subthalamic nucleus in MPTP-treated monkeys. Brain 2007; 130: 212945.Google Scholar
Spieles-Engemann, AL, Behbehani, MM, Collier, TJ, et al. Stimulation of the rat subthalamic nucleus is neuroprotective following significant nigral dopamine neuron loss. Neurobiol Dis 2010; 39: 10515.Google Scholar
Olanow, CW, Kieburtz, K, Schapira, AH. Why have we failed to achieve neuroprotection in Parkinson's disease? Ann Neurol 2008; 64(Supp. 2): S10110.Google Scholar
Moro, E, Lozano, A, Pollak, P, et al. Long-term results of a multicenter study on subthalamic and pallidal stimulation in Parkinson's disease. Mov Disord 2010; 25: 57886.Google Scholar
Castrioto, A, Lozano, A, Poon, Y, et al. Ten-year outcome of subthalamic stimulation in Parkinson disease: a blinded evaluation. Arch Neurol 2011; 68: 15506.Google Scholar
Ngoga, D, Mitchell, R, Kausar, J, et al. Deep brain stimulation improves survival in severe Parkinson's disease. J Neurol Neurosurg Psychiatry 2013; 85: 1722.Google Scholar
Aarsland, D, Marsh, L, Schrag, A. Neuropsychiatric symptoms in Parkinson's disease. Mov Disord 2009; 24: 217586.Google Scholar
Welter, ML, Houeto, JL, Tezenas du Montcel, S, et al. Clinical predictive factors of subthalamic stimulation in Parkinson's disease. Brain 2002; 125: 57583.Google Scholar
Rodriguez-Oroz, MC, Moro, E, Krack, P. Long-term outcomes of surgical therapies for Parkinson's disease. Mov Disord 2012; 27: 171828.Google Scholar
Schermer, M. Ethical issues in deep brain stimulation. Front Integr Neurosci 2011; 5: 17.Google Scholar
Bell, E, Bell, E, Maxwell, B, et al. Deep brain stimulation and ethics: perspectives from a multisite qualitative study of Canadian neurosurgical centers. World Neurosurg 2011; 76: 53747.Google Scholar
Ardouin, C, Pillon, B, Peiffer, E, et al. Bilateral subthalamic or pallidal stimulation for Parkinson's disease affects neither memory nor executive functions: a consecutive series of 62 patients. Ann Neurol 1999; 46: 21723.Google Scholar
Massano, J, Garrett, C. Deep brain stimulation and cognitive decline in Parkinson's disease: a clinical review. Front Neurol 2012; 3: 66.Google Scholar
Weintraub, D, Duda, JE, Carlson, K, et al. Suicide ideation and behaviours after STN and GPi DBS surgery for Parkinson's disease: results from a randomised, controlled trial. J Neurol Neurosurg Psychiatry 2013; 84: 111318.Google Scholar
Meissner, W, Schreiter, D, Volkmann, J, et al. Deep brain stimulation in late stage Parkinson's disease: a retrospective cost analysis in Germany. J Neurol 2005; 252: 21823.Google Scholar
Oyama, G, Okun, MS, Schmidt, P, et al. Deep brain stimulation may improve quality of life in people with Parkinson's disease without affecting caregiver burden. Neuromodulation 2013; 17: 12632.Google Scholar
Temel, Y, Kessels, A, Tan, S, et al. Behavioural changes after bilateral subthalamic stimulation in advanced Parkinson disease: a systematic review. Parkinsonism Relat Disord 2006; 12: 26572.Google Scholar
Kopin, IJ. MPTP: an industrial chemical and contaminant of illicit narcotics stimulates a new era in research on Parkinson's disease. Environ Health Perspect 1987; 75: 4551.Google Scholar
Schwarting, RK, Huston, JP. The unilateral 6-hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Prog Neurobiol 1996; 50: 275331.Google Scholar
Deep-Brain Stimulation for Parkinson's Disease Study Group. Deep-brain stimulation of the subthalamic nucleus or the pars interna of the globus pallidus in Parkinson's disease. N Engl J Med 2001; 345: 95663.Google Scholar
Luquin, M, Saldise, L, Guillén, J, et al. Does increased excitatory drive from the subthalamic nucleus contribute to dopaminergic neuronal death in Parkinson's disease? Exp Neurol 2006; 201: 40715.Google Scholar
Visser-Vandewalle, V, van der Linden, C, Temel, Y, et al. Long-term effects of bilateral subthalamic nucleus stimulation in advanced Parkinson disease: a four year follow-up study. Parkinsonism Relat Disord 2005; 11: 15765.Google Scholar
Rodriguez-Oroz, M, Obeso, J, Lang, A, et al. Bilateral deep brain stimulation in Parkinson's disease: a multicentre study with 4 years follow-up. Brain 2005; 128: 22409.Google Scholar
Cheng, H, Ulane, CM, Burke, RE. Clinical progression in Parkinson disease and the neurobiology of axons. Ann Neurol 2010; 67: 71525.Google Scholar
Kordower, JH, Olanow, CW, Dodiya, HB, et al. Disease duration and the integrity of the nigrostriatal system in Parkinson's disease. Brain 2013; 136: 241931.Google Scholar
Gill, CE, Allen, LA, Konrad, PE, et al. Deep brain stimulation for early-stage Parkinson's disease: an illustrative case. Neuromodulation 2011; 14: 51522.Google Scholar
Charles, PD, Dolhun, RM, Gill, CE, et al. Deep brain stimulation in early Parkinson's disease: enrollment experience from a pilot trial. Parkinsonism Relat Disord 2012; 18: 26873.Google Scholar
Charles, D, Tolleson, C, Davis, T, Gill, C. et al. Pilot study assessing the feasibility of applying bilateral subthalamic nucleus deep brain stimulation in very early stage Parkinson's disease: study design and rationale. J Parkinson's Dis 2012; 2: 21523.Google Scholar
Kahn, E, D'Haese, P-, Dawant, B, et al. Deep brain stimulation in early stage Parkinson's disease: operative experience from a prospective randomised clinical trial. J Neurol Neurosurg Psychiatry 2012; 83: 16470.Google Scholar
Tramontana, MG, Molinari, AL, Konrad, PE, et al. Neuropsychological effects of deep brain stimulation in early stage Parkinson's disease. J Parkinsons Dis 2015; 5: 15163.Google Scholar
Hacker, ML, Tonascia, J, Turchan, M, et al. Deep brain stimulation may reduce the relative risk of clinically important worsening in early stage Parkinson's disease. Parkinsonism Relat Disord 2015; 21: 117783.Google Scholar
Finder, SG, Bliton, MJ, Gill, CE, Davis, TL, Konrad, PE, Charles, PD. Potential subjects' responses to an ethics questionnaire in a phase I study of deep brain stimulation in early Parkinson's disease. J Clin Ethics 2012; 23: 20716.Google Scholar
Finder, SG, Bliton, MJ. Fortitude and Community: Response to Yee and Ford. J. Clin Ethics 2012; 23: 2213.Google Scholar
Charles, D, Konrad, PE, Davis, TL, Neimat, JS, Hacker, ML, Finder, SG. Deep brain stimulation in early stage Parkinson's disease. Parkinsonism Relat Disord 2015; 21: 3478.Google Scholar
Hacker, M, Charles, D, Finder, S. Deep brain stimulation in early stage Parkinson's disease may reduce the relative risk of symptom worsening. Parkinsonism Related Disord 2016; 22: 11213.Google Scholar

References

Emre, M, Aarsland, D, Brown, R, et al. Clinical diagnostic criteria for dementia associated with Parkinson's disease. Mov Disord 2007; 22: 1689707; quiz 1837.Google Scholar
Kehagia, AA, Barker, RA, Robbins, TW. Cognitive impairment in Parkinson's disease: the dual syndrome hypothesis. Neurodegener Dis 2013; 11: 7992.Google Scholar
Emre, M, Aarsland, D, Albanese, A, et al. Rivastigmine for dementia associated with Parkinson's disease. N Engl J Med 2004; 351: 250918.Google Scholar
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th edn. Arlington, VA: American Psychiatric Publishing, 2013.Google Scholar
Olin, JT, Aarsland, D, Meng, X. Rivastigmine in the treatment of dementia associated with Parkinson's disease: effects on activities of daily living. Dement Geriatr Cogn Disord 2010; 29: 51015.Google Scholar
Seppi, K, Weintraub, D, Coelho, M, 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): S4280.Google Scholar
Leroi, I, Brandt, J, Reich, SG, et al. Randomized placebo-controlled trial of donepezil in cognitive impairment in Parkinson's disease. Int J Geriatr Psychiatry 2004; 19: 18.Google Scholar
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 Psychiatry 2005; 76: 9349.Google Scholar
Aarsland, D, Laake, K, Larsen, JP, Janvin, C. Donepezil for cognitive impairment in Parkinson's disease: a randomised controlled study. J Neurol Neurosurg Psychiatry 2002; 72: 70812.Google Scholar
Dubois, B, Tolosa, E, Katzenschlager, R, et al. Donepezil in Parkinson's disease dementia: a randomized, double-blind efficacy and safety study. Mov Disord 2012; 27: 12308.Google Scholar
Lipton, SA, Rosenberg, PA. Excitatory amino acids as a final common pathway for neurologic disorders. N Engl J Med 1994; 330: 61322.Google Scholar
McShane, R, Areosa Sastre, A, Minakaran, N. Memantine for dementia. Cochrane Database Syst Rev 2006; (2): CD003154.Google Scholar
Leroi, I, Overshott, R, Byrne, EJ, Daniel, E, Burns, A. Randomized controlled trial of memantine in dementia associated with Parkinson's disease. Mov Disord 2009; 24: 121721.Google Scholar
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: 96977.Google Scholar
Weintraub, D, Mavandadi, S, Mamikonyan, E, et al. Atomoxetine for depression and other neuropsychiatric symptoms in Parkinson disease. Neurology 2010; 75: 44855.Google Scholar
Trillo, L, Das, D, Hsieh, W, et al. Ascending monoaminergic systems alterations in Alzheimer's disease. translating basic science into clinical care. Neurosci Biobehav Rev 2013; 37: 136379.Google Scholar
Del Tredici, K, Braak, H. Dysfunction of the locus coeruleus-norepinephrine system and related circuitry in Parkinson's disease-related dementia. J Neurol Neurosurg Psychiatry 2013; 84: 77483.Google Scholar
Pedersen, KF, Larsen, JP, Tysnes, OB, Alves, G. Prognosis of mild cognitive impairment in early Parkinson disease: the Norwegian ParkWest study. JAMA Neurol 2013; 70: 5806.Google Scholar
Hanagasi, HA, Gurvit, H, Unsalan, P, et al. The effects of rasagiline on cognitive deficits in Parkinson's disease patients without dementia: a randomized, double-blind, placebo-controlled, multicenter study. Mov Disord 2011; 26: 18518.Google Scholar
US National Institutes of Health. Parallel-group study to assess the effect of rasagiline on cognition in patients with Parkinson's disease. Available at: http://clinicaltrials.gov/show/NCT01723228.Google Scholar
Marsh, L, Biglan, K, Gerstenhaber, M, Williams, JR. Atomoxetine for the treatment of executive dysfunction in Parkinson's disease: a pilot open-label study. Mov Disord 2009; 24: 27782.Google Scholar
Sammer, G, Reuter, I, Hullmann, K, Kaps, M, Vaitl, D. Training of executive functions in Parkinson's disease. J Neurol Sci 2006; 248: 11519.Google Scholar
Edwards, JD, Hauser, RA, O'Connor, ML, et al. Randomized trial of cognitive speed of processing training in Parkinson disease. Neurology 2013; 81: 128490.Google Scholar
Marsh, L, McDonald, WM, Cummings, J, Ravina, B, NINDS/NIMH Work Group on Depression and Parkinson's Disease. Provisional diagnostic criteria for depression in Parkinson's disease: report of an NINDS/NIMH Work Group. Mov Disord 2006; 21: 14858.Google Scholar
Weintraub, D, Morales, KH, Moberg, PJ, et al. Antidepressant studies in Parkinson's disease: a review and meta-analysis. Mov Disord 2005; 20: 11619.Google Scholar
Richard, IH, McDermott, MP, Kurlan, R, et al. A randomized, double-blind, placebo-controlled trial of antidepressants in Parkinson disease. Neurology 2012; 78: 122936.Google Scholar
Menza, M, Dobkin, RD, Marin, H, et al. A controlled trial of antidepressants in patients with Parkinson disease and depression. Neurology 2009; 72: 88692.Google Scholar
Devos, D, Dujardin, K, Poirot, I, et al. Comparison of desipramine and citalopram treatments for depression in Parkinson's disease: a double-blind, randomized, placebo-controlled study. Mov Disord 2008; 23: 8507.Google Scholar
Barone, P, Poewe, W, Albrecht, S, et al. Pramipexole for the treatment of depressive symptoms in patients with Parkinson's disease: a randomised, double-blind, placebo-controlled trial. Lancet Neurol 2010; 9: 57380.Google Scholar
Barone, P, Scarzella, L, Marconi, R, et al. Pramipexole versus sertraline in the treatment of depression in Parkinson's disease: a national multicenter parallel-group randomized study. J Neurol 2006; 253: 6017.Google Scholar
Pahwa, R, Stacy, MA, Factor, SA, et al. Ropinirole 24-hour prolonged release: randomized, controlled study in advanced Parkinson disease. Neurology 2007; 68: 110815.Google Scholar
Rascol, O, Brooks, DJ, Melamed, E, et al. Rasagiline as an adjunct to levodopa in patients with Parkinson's disease and motor fluctuations (LARGO, Lasting effect in Adjunct therapy with Rasagiline Given Once daily, study): a randomised, double-blind, parallel-group trial. Lancet 2005; 365: 94754.Google Scholar
Korchounov, A, Winter, Y, Rossy, W. Combined beneficial effect of rasagiline on motor function and depression in de novo PD. Clin Neuropharmacol 2012; 35: 1214.Google Scholar
Olanow, CW, Rascol, O, Hauser, R, et al. A double-blind, delayed-start trial of rasagiline in Parkinson's disease. N Engl J Med 2009; 361: 126878.Google Scholar
Dobkin, RD, Menza, M, Allen, LA, et al. Cognitive-behavioral therapy for depression in Parkinson's disease: a randomized, controlled trial. Am J Psychiatry 2011; 168: 106674.Google Scholar
Follett, KA, Weaver, FM, Stern, M, et al. Pallidal versus subthalamic deep-brain stimulation for Parkinson's disease. N Engl J Med 2010; 362: 207791.Google Scholar
Forsaa, EB, Larsen, JP, Wentzel-Larsen, T, et al. A 12-year population-based study of psychosis in Parkinson disease. Arch Neurol 2010; 67: 9961001.Google Scholar
Ravina, B, Marder, K, Fernandez, HH, et al. Diagnostic criteria for psychosis in Parkinson's disease: report of an NINDS, NIMH work group. Mov Disord 2007; 22: 10618.Google Scholar
Fenelon, G, Alves, G. Epidemiology of psychosis in Parkinson's disease. J Neurol Sci 2010; 289: 1217.Google Scholar
Parkinson Study Group. Low-dose clozapine for the treatment of drug-induced psychosis in Parkinson's disease. N Engl J Med 1999; 340: 75763.Google Scholar
Weintraub, D, Chen, P, Ignacio, RV, Mamikonyan, E, Kales, HC. Patterns and trends in antipsychotic prescribing for Parkinson disease psychosis. Arch Neurol 2011; 68: 899904.Google Scholar
Friedman, JH, Berman, RM, Goetz, CG, et al. Open-label flexible-dose pilot study to evaluate the safety and tolerability of aripiprazole in patients with psychosis associated with Parkinson's disease. Mov Disord 2006; 21: 207881.Google Scholar
Marras, C, Gruneir, A, Wang, X, et al. Antipsychotics and mortality in parkinsonism. Am J Geriatr Psychiatry 2012; 20: 14958.Google Scholar
Cummings, J, Isaacson, S, Mills, R, et al. Pimavanserin for patients with Parkinson's disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet 2013; 383: 53340.Google Scholar
Weintraub, D, Koester, J, Potenza, MN, et al. Impulse control disorders in Parkinson disease: a cross-sectional study of 3090 patients. Arch Neurol 2010; 67: 58995.Google Scholar
Weintraub, D, Papay, K, Siderowf, A, Parkinson's Progression Markers Initiative. Screening for impulse control symptoms in patients with de novo Parkinson disease: a case–control study. Neurology 2013; 80: 17680.Google Scholar
Okai, D, Askey-Jones, S, Samuel, M, et al. Trial of CBT for impulse control behaviors affecting Parkinson patients and their caregivers. Neurology 2013; 80: 7929.Google Scholar
Troeung, L, Egan, SJ, Gasson, N. A meta-analysis of randomised placebo-controlled treatment trials for depression and anxiety in Parkinson's disease. PLoS One 2013; 8: e79510.Google Scholar
Devos, D, Moreau, C, Maltete, D, et al. Rivastigmine in apathetic but dementia and depression-free patients with Parkinson's disease: a double-blind, placebo-controlled, randomised clinical trial. J Neurol Neurosurg Psychiatry 2013; 85: 66874.Google Scholar
Thobois, S, Lhommee, E, Klinger, H, et al. Parkinsonian apathy responds to dopaminergic stimulation of D2/D3 receptors with piribedil. Brain 2013; 136: 156877.Google Scholar
Friedman, JH, Ravina, B, Mills, R. A multicenter, placebo controlled, double blind trial to examine the safety and efficacy of pimavanserin in the treatment of psychosis in Parkinson's disease. Neurology 2010; 74: A299.Google Scholar
Walsh, BT, Seidman, SN, Sysko, R, Gould, M. Placebo response in studies of major depression: variable, substantial, and growing. JAMA 2002; 287: 18407.Google Scholar
Goetz, CG, Wuu, J, McDermott, MP, et al. Placebo response in Parkinson's disease: comparisons among 11 trials covering medical and surgical interventions. Mov Disord 2008; 23: 6909.Google Scholar
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, multicentre trial. Lancet Neurol 2009; 8: 61318.Google Scholar
Michael J. Fox Foundation. Fox Trial Finder. Available at: https://foxtrialfinder.michaeljfox.org/.Google Scholar
Starkstein, SE, Dragovic, M, Dujardin, K, et al. Anxiety has specific syndromal profiles in Parkinson disease: a data-driven approach. Am J Geriatr Psychiatry 2013; 22: 141017.Google Scholar
Dubois, B, Burn, D, Goetz, C, et al. Diagnostic procedures for Parkinson's disease dementia: recommendations from the Movement Disorder Society task force. Mov Disord 2007; 22: 231424.Google Scholar
Litvan, I, Aarsland, D, Adler, CH, et al. MDS Task Force on mild cognitive impairment in Parkinson's disease: critical review of PD-MCI. Mov Disord 2011; 26: 181424.Google Scholar
Robert, P, Onyike, CU, Leentjens, AF, et al. Proposed diagnostic criteria for apathy in Alzheimer's disease and other neuropsychiatric disorders. Eur Psychiatry 2009; 24: 98104.Google Scholar
Drijgers, RL, Dujardin, K, Reijnders, JS, Defebvre, L, Leentjens, AF. Validation of diagnostic criteria for apathy in Parkinson's disease. Parkinsonism Relat Disord 2010; 16: 65660.Google Scholar
Weintraub, D, Mamikonyan, E, Papay, K, et al. Questionnaire for impulsive-compulsive disorders in Parkinson's disease – rating scale. Mov Disord 2012; 27: 2427.Google Scholar
Giovannoni, G, O'sullivan, JD, Turner, K, Manson, AJ, Lees, AJ. Hedonistic homeostatic dysregulation in patients with Parkinson's disease on dopamine replacement therapies. J Neurol Neurosurg Psychiatry 2000; 68: 4238.Google Scholar
Barton, B, Grabli, D, Bernard, B, et al. Clinical validation of Movement Disorder Society-recommended diagnostic criteria for Parkinson's disease with dementia. Mov Disord 2012; 27: 24853.Google Scholar
Schrag, A, Barone, P, Brown, RG, et al. Depression rating scales in Parkinson's disease: critique and recommendations. Mov Disord 2007; 22: 107792.Google Scholar
Fernandez, HH, Aarsland, D, Fenelon, G, et al. Scales to assess psychosis in Parkinson's disease: Critique and recommendations. Mov Disord 2008; 23: 484500.Google Scholar
Leentjens, AF, Dujardin, K, Marsh, L, et al. Apathy and anhedonia rating scales in Parkinson's disease: critique and recommendations. Mov Disord 2008; 23: 200414.Google Scholar
Leentjens, AF, Dujardin, K, Marsh, L, et al. Anxiety rating scales in Parkinson's disease: critique and recommendations. Mov Disord 2008; 23: 201525.Google Scholar
Voss, T, Bahr, D, Cummings, J, et al. Performance of a shortened Scale for Assessment of Positive Symptoms for Parkinson's disease psychosis. Parkinsonism Relat Disord 2013; 19: 2959.Google Scholar
Martinez-Martin, P, Rojo-Abuin, JM, Dujardin, K, et al. Designing a new scale to measure anxiety symptoms in Parkinson's disease: item selection based on canonical correlation analysis. Eur J Neurol 2013; 20: 1198203.Google Scholar
Schneider, LS, Tariot, PN, Dagerman, KS, et al. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer's disease. N Engl J Med 2006; 355: 152538.Google Scholar

References

Marks, L, Turner, K, O'Sulliva, J, Deighton, B, Lees, A. Drooling in Parkinson's disease: a novel speech and language therapy intervention. Int J Lang Commun Disord 2001; 36 (Suppl.): 2827.Google Scholar
Mancini, F, Zangaglia, R, Cristina, S, et al. Double-blind, placebo-controlled study to evaluate the efficacy and safety of botulinum toxin type A in the treatment of drooling in parkinsonism. Mov Disord 2003; 18: 6858.Google Scholar
Lagalla, G, Millevolte, M, Capecci, M, Provinciali, L, Ceravolo, MG. Botulinum toxin type A for drooling in Parkinson's disease: a double-blind, randomized, placebo-controlled study. Mov Disord 2006; 21: 7047.Google Scholar
Ondo, WG, Hunter, C, Moore, W. A double-blind placebo-controlled trial of botulinum toxin B for sialorrhea in Parkinson's disease. Neurology 2004; 62: 3740.Google Scholar
Lagalla, G, Millevolte, M, Capecci, M, Provinciali, L, Ceravolo, MG. Long-lasting benefits of botulinum toxin type B in Parkinson's disease-related drooling. J Neurol 2009; 256: 5637.Google Scholar
Chinnapongse, R, Gullo, K, Nemeth, P, Zhang, Y, Griggs, L. Safety and efficacy of botulinum toxin type B for treatment of sialorrhea in Parkinson's disease: a prospective double-blind trial. Mov Disord 2012; 27: 21926.Google Scholar
Thomsen, TR, Galpern, WR, Asante, A, Arenovich, T, Fox, SH. Ipratropium bromide spray as treatment for sialorrhea in Parkinson's disease. Mov Disord 2007; 22: 226873.Google Scholar
Arbouw, ME, Movig, KL, Koopmann, M, et al. Glycopyrrolate for sialorrhea in Parkinson disease: a randomized, double-blind, crossover trial. Neurology 2010; 74: 12037.Google Scholar
Lloret, SP, Nano, G, Carrosella, A, Gamzu, E, Merello, M. A double-blind, placebo-controlled, randomized, crossover pilot study of the safety and efficacy of multiple doses of intra-oral tropicamide films for the short-term relief of sialorrhea symptoms in Parkinson's disease patients. J Neurolog Sci 2011; 310: 24850.Google Scholar
Troche, MS, Okun, MS, Rosenbek, JC, et al. Aspiration and swallowing in Parkinson disease and rehabilitation with EMST A randomized trial. Neurology 2010; 75: 191219.Google Scholar
Manor, Y, Mootanah, R, Freud, D, Giladi, N, Cohen, JT. Video-assisted swallowing therapy for patients with Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 20711.Google Scholar
Baijens, LW, Speyer, R, Passos, VL, et al. The effect of surface electrical stimulation on swallowing in dysphagic Parkinson patients. Dysphagia 2012; 27: 52837.Google Scholar
Heijnen, BJ, Speyer, R, Baijens, LW, Bogaardt, HC. Neuromuscular electrical stimulation versus traditional therapy in patients with Parkinson's disease and oropharyngeal dysphagia: effects on quality of life. Dysphagia 2012; 27: 33645.Google Scholar
Sullivan, KL, Staffetti, JF, Hauser, RA, Dunne, PB, Zesiewicz, TA. Tegaserod (Zelnorm) for the treatment of constipation in Parkinson's disease. Mov Disord 2006; 21: 11516.Google Scholar
Zangaglia, R, Martignoni, E, Glorioso, M, et al. Macrogol for the treatment of constipation in Parkinson's disease. A randomized placebo-controlled study. Mov Disord 2007; 22: 123944.Google Scholar
Ondo, WG, Kenney, C, Sullivan, K, et al. Placebo-controlled trial of lubiprostone for constipation associated with Parkinson disease. Neurology 2012; 78: 16504.Google Scholar
Locke, MC, Wu, SS, Foote, KD, et al. Weight changes in subthalamic nucleus vs globus pallidus internus deep brain stimulation: results from the COMPARE Parkinson disease deep brain stimulation cohort. Neurosurgery 2011; 68: 12337.Google Scholar
Kabay, SC, Kabay, S, Yucel, M, Ozden, H. Acute urodynamic effects of percutaneous posterior tibial nerve stimulation on neurogenic detrusor overactivity in patients with Parkinson's disease. Neurourol Urodyn 2009; 28: 627.Google Scholar
Kulaksizoglu, H, Parman, Y. Use of botulinim toxin-A for the treatment of overactive bladder symptoms in patients with Parkinson's disease. Parkinsonism Relat Disord 2010; 16: 5314.Google Scholar
Vaughan, CP, Juncos, JL, Burgio, KL, et al. Behavioral therapy to treat urinary incontinence in Parkinson disease. Neurology 2011; 76: 16314.Google Scholar
Hussain, IF, Brady, CM, Swinn, MJ, Mathias, CJ, Fowler, CJ. 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: 3714.Google Scholar
Safarinejad, MR, Taghva, A, Shekarchi, B, Safarinejad, S. Safety and efficacy of sildenafil citrate in the treatment of Parkinson-emergent erectile dysfunction: a double-blind, placebo-controlled, randomized study. Int J Impot Res 2010; 22: 32535. Retraction: Int J Impot Res 2011; 23: 94.Google Scholar
Abate, G, Polimeni, RM, Cuccurullo, F, Puddu, P, Lenzi, S. Effects of indomethacin on postural hypotension in Parkinsonism. Br Med J 1979; 2: 1466.Google Scholar
Schoffer, KL, Henderson, RD, O'Maley, K, O'sullivan, JD. Nonpharmacological treatment, fludrocortisone, and domperidone for orthostatic hypotension in Parkinson's disease. Mov Disord 2007; 22: 15439.Google Scholar
Djaldetti, R, Yust-Katz, S, Kolianov, V, Melamed, E, Dabby, R. The effect of duloxetine on primary pain symptoms in Parkinson disease. Clin Neuropharmacol 2007; 30: 2015.Google Scholar
Dellapina, E, Ory-Magne, F, Regragui, W, et al. Effect of subthalamic deep brain stimulation on pain in Parkinson's disease. Pain 2012; 153: 226773.Google Scholar
Marques, A, Chassin, O, Morand, D, et al. Central pain modulation after subthalamic nucleus stimulation: a crossover randomized trial. Neurology 2013; 81: 63340.Google Scholar
Högl, B, Saletu, M, Brandauer, E, 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: 9059.Google Scholar
Adler, CH, Caviness, JN, Hentz, JG, Lind, M, Tiede, J. Randomized trial of modafinil for treating subjective daytime sleepiness in patients with Parkinson's disease. Mov Disord 2003; 18: 28793.Google Scholar
Ondo, WG, Fayle, R, Atassi, F, Jankovic, J. Modafinil for daytime somnolence in Parkinson's disease: double blind, placebo controlled parallel trial. J Neurol Neurosurg Psychiatry 2005; 76: 16369.Google Scholar
Postuma, RB, Lang, AE, Munhoz, RP, et al. Caffeine for treatment of Parkinson disease A randomized controlled trial. Neurology 2012; 79: 6518.Google Scholar
Medeiros, CA, Carvalhedo de Bruin, PF, Lopes, LA, et al. Effect of exogenous melatonin on sleep and motor dysfunction in Parkinson's disease. J Neurol 2007; 254: 45964.Google Scholar
Dowling, GA, Mastick, J, Colling, E, et al. Melatonin for sleep disturbances in Parkinson's disease. Sleep Med 2005; 6: 45966.Google Scholar
Menza, M, Dobkin, RD, Marin, H, et al. Treatment of insomnia in Parkinson's disease: a controlled trial of eszopiclone and placebo. Mov Disord 2010; 25: 170814.Google Scholar
Larsson, V, Aarsland, D, Ballard, C, Minthon, L, Londos, E. The effect of memantine on sleep behaviour in dementia with Lewy bodies and Parkinson's disease dementia. Int J Geriatr Psychiatry 2010; 25: 10308.Google Scholar
Di Giacopo, R, Fasano, A, Quaranta, D, et al. Rivastigmine as alternative treatment for refractory REM behavior disorder in Parkinson's disease. Mov Disord 2012; 27: 55961.Google Scholar

References

Ashburn, A, Fazakarley, L, Ballinger, C, et al. A randomised controlled trial of a home based exercise programme to reduce the risk of falling among people with Parkinson's disease. J Neurol Neurosurg Psychiatry 2007; 78: 67884.Google Scholar
Kluding, P, McGinnis, PQ. Multidimensional exercise for people with Parkinson's disease: a case report. Physiother Theory Pract 2006; 22: 15362.Google Scholar
Kwolek, A. [Rehabilitation of patients with Parkinson disease]. Neurol Neurochir Pol 2003; 37 (Suppl. 5): 21120 (in Polish).Google Scholar
Schenkman, M, Hall, D, Kumar, R, Kohrt, WM. Endurance exercise training to improve economy of movement of people with Parkinson disease: three case reports. Phys Ther 2008; 88: 6376.Google Scholar
Bergen, JL, Toole, T, Elliott, RG 3rd, et al. Aerobic exercise intervention improves aerobic capacity and movement initiation in Parkinson's disease patients. NeuroRehabilitation 2002; 17: 1618.Google Scholar
Schenkman, M, Hall, DA, Baron, AE, et al. Exercise for people in early- or mid-stage Parkinson disease: a 16-month randomized controlled trial. Phys Ther 2012; 92: 1395410.Google Scholar
Burini, D, Farabollini, B, Iacucci, S, et al. A randomised controlled cross-over trial of aerobic training versus Qigong in advanced Parkinson's disease. Eura Medicophys 2006; 42: 2318.Google Scholar
Tillerson, JL, Caudle, WM, Reveron, ME, Miller, GW. Exercise induces behavioral recovery and attenuates neurochemical deficits in rodent models of Parkinson's disease. Neuroscience 2003; 119: 899911.Google Scholar
Rochester, L, Burn, DJ, Woods, G, Godwin, J, Nieuwboer, A. Does auditory rhythmical cueing improve gait in people with Parkinson's disease and cognitive impairment? A feasibility study. Mov Disord 2009; 24: 83945.Google Scholar
Poulton, NP, Muir, GD. Treadmill training ameliorates dopamine loss but not behavioral deficits in hemi-parkinsonian rats. Exp Neurol 2005; 193: 18197.Google Scholar
Ridgel, AL, Vitek, JL, Alberts, JL. Forced, not voluntary, exercise improves motor function in Parkinson's disease patients. Neurorehabil Neural Repair 2009; 23: 6008.Google Scholar
Miyai, I, Fujimoto, Y, Yamamoto, H, et al. Long-term effect of body weight-supported treadmill training in Parkinson's disease: a randomized controlled trial. Arch Phys Med Rehabil 2002; 83: 13703.Google Scholar
Protas, EJ, Mitchell, K, Williams, A, et al. Gait and step training to reduce falls in Parkinson's disease. NeuroRehabilitation 2005; 20: 18390.Google Scholar
Pohl, M, Rockstroh, G, Ruckriem, S, Mrass, G, Mehrholz, J. Immediate effects of speed-dependent treadmill training on gait parameters in early Parkinson's disease. Arch Phys Med Rehabil 2003; 84: 17606.Google Scholar
Baddeley, A. Working memory: looking back and looking forward. Nat Rev Neurosci 2003; 4: 82939.Google Scholar
Altgassen, M, Phillips, L, Kopp, U, Kliegel, M. Role of working memory components in planning performance of individuals with Parkinson's disease. Neuropsychologia 2007; 45: 23937.Google Scholar
Koerts, J, Leenders, KL, Brouwer, WH. Cognitive dysfunction in non-demented Parkinson's disease patients: controlled and automatic behavior. Cortex 2009; 45: 9229.Google Scholar
Muslimovic, D, Post, B, Speelman, JD, Schmand, B. Cognitive profile of patients with newly diagnosed Parkinson disease. Neurology 2005; 65: 123945.Google Scholar
Kramer, AF, Erickson, KI. Capitalizing on cortical plasticity: influence of physical activity on cognition and brain function. Trends Cogn Sci 2007; 11: 3428.Google Scholar
Kramer, AF, Hahn, S, Cohen, NJ, et al. Ageing, fitness and neurocognitive function. Nature 1999; 400: 41819.Google Scholar
Colcombe, SJ, Erickson, KI, Scalf, PE, et al. Aerobic exercise training increases brain volume in aging humans. J Gerontol A Biol Sci Med Sci 2006; 61: 116670.Google Scholar
Raz, N, Williamson, A, Gunning-Dixon, F, Head, D, Acker, JD. Neuroanatomical and cognitive correlates of adult age differences in acquisition of a perceptual-motor skill. Microsc Res Tech. 2000; 51: 8593.Google Scholar
Nocera, JR, Altmann, LJ, Sapienza, C, Okun, MS, Hass, CJ. Can exercise improve language and cognition in Parkinson's disease? A case report. Neurocase 2010; 16: 3016.Google Scholar
Latham, NK, Bennett, DA, Stretton, CM, Anderson, CS. Systematic review of progressive resistance strength training in older adults. J Gerontol A Biol Sci Med Sci 2004; 59: 4861.Google Scholar
Mian, OS, Baltzopoulos, V, Minetti, AE, Narici, MV. The impact of physical training on locomotor function in older people. Sports Med 2007; 37: 683701.Google Scholar
David, FJ, Rafferty, MR, Robichaud, JA, et al. Progressive resistance exercise and Parkinson's disease: a review of potential mechanisms. Parkinsons Dis 2012; 2012: 124527.Google Scholar
Glendinning, DS, Enoka, RM. Motor unit behavior in Parkinson's disease. Phys Ther 1994; 74: 6170.Google Scholar
Corcos, DM, Robichaud, JA, David, FJ, et al. A two-year randomized controlled trial of progressive resistance exercise for Parkinson's disease. Mov Disord 2013; 28: 123040.Google Scholar
Nocera, J, Horvat, M, Ray, CT. Effects of home-based exercise on postural control and sensory organization in individuals with Parkinson disease. Parkinsonism Relat Disord 2009; 15: 7425.Google Scholar
Hass, CJ, Buckley, TA, Pitsikoulis, C, Barthelemy, EJ. Progressive resistance training improves gait initiation in individuals with Parkinson's disease. Gait Posture 2012; 35: 66973.Google Scholar
Hass, CJ, Collins, MA, Juncos, JL. Resistance training with creatine monohydrate improves upper-body strength in patients with Parkinson disease: a randomized trial. Neurorehabil Neural Repair 2007; 21: 10715.Google Scholar
Li, F, Harmer, P, Fisher, KJ, et al. Tai Chi-based exercise for older adults with Parkinson's disease: a pilot-program evaluation. J Aging Phys Act 2007; 15: 13951.Google Scholar
Hackney, ME, Earhart, GM. Tai Chi improves balance and mobility in people with Parkinson disease. Gait Posture 2008; 28: 45660.Google Scholar
Li, F, Harmer, P, Liu, Y, et al. A randomized controlled trial of patient-reported outcomes with tai chi exercise in Parkinson's disease. Mov Disord 2014; 29: 53945.Google Scholar
Amano, S, Nocera, JR, Vallabhajosula, S, et al. The effect of Tai Chi exercise on gait initiation and gait performance in persons with Parkinson's disease. Parkinsonism Relat Disord 2013; 19: 95560.Google Scholar
Lam, LC, Chau, RC, Wong, BM, et al. A 1-year randomized controlled trial comparing mind body exercise (Tai Chi) with stretching and toning exercise on cognitive function in older Chinese adults at risk of cognitive decline. J Am Med Dir Assoc 2012; 13: 568.e15–20.Google Scholar
Nocera, JR, Amano, S, Vallabhajosula, S, Hass, CJ. Tai Chi exercise to improve non-motor symptoms of Parkinson's disease. J Yoga Phys Ther 2013; 3. DOI:10.4172/2157-7595.1000137.Google Scholar
Rajendran, PR, Thompson, RE, Reich, SG. The use of alternative therapies by patients with Parkinson's disease. Neurology 2001; 57: 7904.Google Scholar
Donoyama, N, Ohkoshi, N. Effects of traditional Japanese massage therapy on various symptoms in patients with Parkinson's disease: a case-series study. J Altern Complement Med 2012; 18: 2949.Google Scholar
Zeng, BY, Salvage, S, Jenner, P. Current development of acupuncture research in Parkinson's disease. Int Rev Neurobiol 2013; 111: 14158.Google Scholar
Cho, SY, Shim, SR, Rhee, HY, et al. Effectiveness of acupuncture and bee venom acupuncture in idiopathic Parkinson's disease. Parkinsonism Relat Disord 2012; 18: 94852.Google Scholar
Lee, HS, Park, HL, Lee, SJ, et al. Scalp acupuncture for Parkinson's disease: a systematic review of randomized controlled trials. Chin J Integr Med 2013; 19: 297306.Google Scholar
Nieuwboer, A, Rochester, L, Muncks, L, Swinnen, SP. Motor learning in Parkinson's disease: limitations and potential for rehabilitation. Parkinsonism Relat Disord 2009; 15 (Suppl. 3): S538.Google Scholar
Kadivar, Z, Corcos, DM, Foto, J, Hondzinski, JM. Effect of step training and rhythmic auditory stimulation on functional performance in Parkinson patients. Neurorehabil Neural Repair 2011; 25: 62635.Google Scholar
Dibble, LE, Nicholson, DE, Shultz, B, et al. Sensory cueing effects on maximal speed gait initiation in persons with Parkinson's disease and healthy elders. Gait Posture 2004; 19: 21525.Google Scholar
Jiang, Y, Norman, KE. Effects of visual and auditory cues on gait initiation in people with Parkinson's disease. Clin Rehabil 2006; 20: 3645.Google Scholar
Ballanger, B, Thobois, S, Baraduc, P, et al. “Paradoxical kinesis” is not a hallmark of Parkinson's disease but a general property of the motor system. Mov Disord 2006; 21: 14905.Google Scholar
Howe, TE, Lovgreen, B, Cody, FW, Ashton, VJ, Oldham, JA. Auditory cues can modify the gait of persons with early-stage Parkinson's disease: a method for enhancing parkinsonian walking performance? Clin Rehabil 2003; 17: 3637.Google Scholar
Nanhoe-Mahabier, W, Delval, A, Snijders, AH, et al. The possible price of auditory cueing: influence on obstacle avoidance in Parkinson's disease. Mov Disord 2012; 27: 5748.Google Scholar
Elefant, C, Baker, FA, Lotan, M, Lagesen, SK, Skeie, GO. The effect of group music therapy on mood, speech, and singing in individuals with Parkinson's disease – a feasibility study. J Music Ther 2012; 49: 278302.CrossRefGoogle ScholarPubMed
Shih, LC, Piel, J, Warren, A, et al. Singing in groups for Parkinson's disease (SING-PD): a pilot study of group singing therapy for PD-related voice/speech disorders. Parkinsonism Relat Disord 2012; 18: 54852.Google Scholar
Clair, A, Lyons, KE, Hamburg, J. A feasibility study of the effects of music and movement of physical function, quality of life, depression, and anxiety in patients with Parkinson's Disease. Music Med 2011; 4: 4955.Google Scholar
Pacchetti, C, Mancini, F, Aglieri, R, et al. Active music therapy in Parkinson's disease: an integrative method for motor and emotional rehabilitation. Psychosom Med 2000; 62: 38693.Google Scholar
Toth, MJ, Fishman, PS, Poehlman, ET. Free-living daily energy expenditure in patients with Parkinson's disease. Neurology 1997; 48: 8891.Google Scholar
Hirsch, MA, Toole, T, Maitland, CG, Rider, RA. The effects of balance training and high-intensity resistance training on persons with idiopathic Parkinson's disease. Arch Phys Med Rehabil 2003; 84: 110917.Google Scholar
Federici, A, Bellagamba, S, Rocchi, MB. Does dance-based training improve balance in adult and young old subjects? A pilot randomized controlled trial. Aging Clin Exp Res 2005; 17: 3859.Google Scholar
Palo-Bengtsson, L, Winblad, B, Ekman, SL. Social dancing: a way to support intellectual, emotional and motor functions in persons with dementia. J Psychiatr Ment Health Nurs 1998; 5: 54554.CrossRefGoogle ScholarPubMed
Song, R, June, KJ, Kim, CG, Jeon, MY. Comparisons of motivation, health behaviors, and functional status among elders in residential homes in Korea. Public Health Nurs. 2004; 21: 36171.Google Scholar
McKinley, P, Jacobson, A, Leroux, A, et al. Effect of a community-based Argentine tango dance program on functional balance and confidence in older adults. J Aging Phys Act 2008; 16: 43553.Google Scholar
Hackney, ME, Earhart, GM. Health-related quality of life and alternative forms of exercise in Parkinson disease. Parkinsonism Relat Disord 2009; 15: 6448.Google Scholar
Hackney, ME, Earhart, GM. Effects of dance on gait and balance in Parkinson's disease: a comparison of partnered and nonpartnered dance movement. Neurorehabil Neural Repair 2010; 24: 38492.Google Scholar
Hackney, ME, Kantorovich, S, Levin, R, Earhart, GM. Effects of tango on functional mobility in Parkinson's disease: a preliminary study. J Neurol Phys Ther 2007; 31: 1739.Google Scholar
Duncan, RP, Earhart, GM. Randomized controlled trial of community-based dancing to modify disease progression in Parkinson disease. Neurorehabil Neural Repair 2012; 26: 13243.Google Scholar
McKee, KE, Hackney, ME. The effects of adapted tango on spatial cognition and disease severity in Parkinson's disease. J Mot Behav 2013; 45: 51929.Google Scholar
Volpe, D, Signorini, M, Marchetto, A, Lynch, T, Morris, ME. A comparison of Irish set dancing and exercises for people with Parkinson's disease: a phase II feasibility study. BMC Geriatr 2013; 13: 54.CrossRefGoogle ScholarPubMed
Hackney, ME, Earhart, GM. Short duration, intensive tango dancing for Parkinson disease: an uncontrolled pilot study. Complement Ther Med 2009; 17: 2037.Google Scholar
Marchant, D, Sylvester, JL, Earhart, GM. Effects of a short duration, high dose contact improvisation dance workshop on Parkinson disease: a pilot study. Complement Ther Med 2010; 18: 18490.Google Scholar
Heiberger, L, Maurer, C, Amtage, F, et al. Impact of a weekly dance class on the functional mobility and on the quality of life of individuals with Parkinson's disease. Front Aging Neurosci 2011; 3: 14.Google Scholar
Hackney, ME, Earhart, GM. Effects of dance on balance and gait in severe Parkinson disease: a case study. Disabil Rehabil 2010; 32: 67984.Google Scholar
Houston, S, McGill, A. A mixed-methods study into ballet for people living with Parkinson's. Arts Health 2013; 5: 10319.Google Scholar
Morris, ME, Iansek, R, Kirkwood, B. A randomized controlled trial of movement strategies compared with exercise for people with Parkinson's disease. Mov Disord 2009; 24: 6471.Google Scholar
Morris, ME, Huxham, F, McGinley, J, Dodd, K, Iansek, R. The biomechanics and motor control of gait in Parkinson disease. Clin Biomech (Bristol, Avon) 2001; 16: 45970.Google Scholar
Foster, ER, Golden, L, Duncan, RP, Earhart, GM. Community-based Argentine tango dance program is associated with increased activity participation among individuals with Parkinson's disease. Arch Phys Med Rehabil 2013; 94: 2409.Google Scholar
Dereli, EE, Yaliman, A. Comparison of the effects of a physiotherapist-supervised exercise programme and a self-supervised exercise programme on quality of life in patients with Parkinson's disease. Clin Rehabil 2010; 24: 35262.Google Scholar
Elkis-Abuhoff, DL, Goldblatt, RB, Gaydos, M, Convery, C. A pilot study to determine the psychological effects of manipulation of therapeutic art forms among patients with Parkinson's disease. Int J Art Ther 2013;18: 11321.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×