Photobiomodulation as part of a multi-disciplinary approach for the treatment of Parkinson’s disease symptoms

Main Article Content

Ann Liebert Anita Saltmarche Melissa McGonaghy Orla Hares Brian Bicknell Geoffrey Herkes

Abstract

Background: Parkinson’s disease is an increasingly common neurodegenerative disease that has a substantial personal, social and economic cost. Although the cardinal symptoms of the disease are motor impairment, non-motor symptoms are arguably just as important for their effect on quality of life. Parkinson’s disease also has a highly heterogeneous symptom presentation. The mainstay of treatment has for many years been levodopa, and more recently dopamine agonists, which can alleviate some of the motor symptoms, but can have severe side-effects, can lose effectiveness over time and do not have an effect on non-motor symptoms. Thus, new treatment modalities are required for management of this disease.


Objective: To describe the effect of a multidisciplinary approach for the treatment of the symptoms of Parkinson’s disease, incorporating photobiomodulation (PBM), exercise, diet and careful control of medication.


Methods: Participants presented to either a Parkinson’s clinic in Sydney, Australia or to a Parkinson’s rehabilitation centre in Hamilton, Canada. Participants were self-referred, referred by a neurologist, or had been enrolled in a clinical trial and had continued with treatment for two additional years. All participants received PBM therapy to the abdomen and neck and participated in a targeted Parkinson’s specific exercise program.


Results: All participants reported improvements in some of their Parkinson’s symptoms, both motor and non-motor, including mobility and gait, balance, fine motor control, cognition, quality of life, and sense of smell. These improvements were maintained over a number of years.


Conclusion: A multidisciplinary and individual approach to the treatment of the symptoms of Parkinson’s disease may be an effective way to improve some symptoms and to potentially delay disease progression.

Keywords: Parkinson’s disease, photobiomodulation, exercise, motor symptoms, non-motor symptoms, multidisciplinary

Article Details

How to Cite
LIEBERT, Ann et al. Photobiomodulation as part of a multi-disciplinary approach for the treatment of Parkinson’s disease symptoms. Medical Research Archives, [S.l.], v. 12, n. 9, sep. 2024. ISSN 2375-1924. Available at: <https://esmed.org/MRA/mra/article/view/5648>. Date accessed: 04 oct. 2024. doi: https://doi.org/10.18103/mra.v12i9.5648.
Section
Research Articles

References

1. Dorsey ER, Zafar M, Lettenberger SE, et al. Trichloroethylene: An Invisible Cause of Parkinson’s Disease? Journal of Parkinson's Disease. 2023;13: 203-218. doi:10.3233/JPD-225047

2. Marsden CD. Problems with long-term levodopa therapy for Parkinson's disease. Clin Neuropharmacol. 1994;17 Suppl 2:S32-44.

3. Antonini A, Emmi A, Campagnolo M. Beyond the dopaminergic system: lessons learned from levodopa resistant symptoms in Parkinson's disease. Movement disorders clinical practice. 2023;10(Suppl 2):S50.

4. Heimrich KG, Schönenberg A, Santos-García D, Mir P, Coppadis Study G, Prell T. The Impact of Nonmotor Symptoms on Health-Related Quality of Life in Parkinson's Disease: A Network Analysis Approach. J Clin Med. Mar 29 2023;12(7)doi:10 .3390/jcm12072573

5. Church FC. Treatment Options for Motor and Non-Motor Symptoms of Parkinson's Disease. Biomolecules. Apr 20 2021;11(4)doi:10.3390/biom 11040612

6. Pagano G, Taylor KI, Anzures Cabrera J, et al. Prasinezumab slows motor progression in rapidly progressing early-stage Parkinson’s disease. Nature Medicine. 2024/04/01 2024;30(4):1096-1103. doi:10.1038/s41591-024-02886-y

7. Foltynie T, Athauda D. Chapter 13 - Repurposing anti-diabetic drugs for the treatment of Parkinson's disease: Rationale and clinical experience. In: Björklund A, Cenci MA, eds. Progress in Brain Research. Elsevier; 2020:493-523.

8. Bicknell B, Liebert A, Herkes G. Parkinson’s Disease and Photobiomodulation: Potential for Treatment. Journal of Personalized Medicine. 2024;14(1):112.

9. Herkes G, McGee C, Liebert A, et al. A novel transcranial photobiomodulation device to address motor signs of Parkinson’s disease: a parallel randomised feasibility study. EClinicalMedicine. 2023;66

10. Liebert A, Bicknell B, Laakso E-L, et al. Remote Photobiomodulation Treatment for the Clinical Signs of Parkinson's Disease: A Case Series Conducted During COVID-19. Photobiomodulation, photome dicine, and laser surgery. 2022;40(2):112-122.

11. Liebert A, Bicknell B, Laakso EL, et al. Improvements in clinical signs of Parkinson’s disease using photobiomodulation: a prospective proof-of-concept study. BMC Neurology. 2021/07/ 02 2021;21(1):256. doi:10.1186/s12883-021-02248-y

12. McGee C, Liebert A, Bicknell B, et al. A Randomized Placebo-Controlled Study of a Transcranial Photobiomodulation Helmet in Parkinson’s Disease: Post-Hoc Analysis of Motor Outcomes. J Clin Med. 2023;12(8):2846.

13. Liebert A, Bicknell B, Laakso E-L, et al. Improvements in the clinical signs of Parkinson’s disease using photobiomodulation: a 3-year follow-up case series. Medical Research Archives. 2023;11(3)doi:doi.org/10.18103/mra.v11i3.3690

14. Hamblin MR. Mechanisms and Mitochondrial Redox Signaling in Photobiomodulation. Photochemistry and Photobiology. 2018;94(2):199-212. doi:doi:10.1111/php.12864

15. Mester E, Szende B, P. G. The effect of laser beams on the growth of hair in mice. Radiobiology and Radiotherapy 1968;9(5):621-626.

16. E M, G L, M S, B S, J. T. The simulating effect of low power laser rays on biological systems. Laser Review. 1968;1:3.

17. Dompe C, Moncrieff L, Matys J, et al. Photobiomodulation-Underlying Mechanism and Clinical Applications. J Clin Med. Jun 3 2020;9(6):1724. doi:10.3390/jcm9061724

18. Glass GE. Photobiomodulation: the clinical applications of low-level light therapy. Aesthetic Surgery Journal. 2021;41(6):723-738.

19. González-Muñoz A, Cuevas-Cervera M, Pérez-Montilla JJ, et al. Efficacy of Photobiomodulation Therapy in the Treatment of Pain and Inflammation: A Literature Review. Healthcare. 2023;11(7):938.

20. Kim H, Kim MJ, Kwon YW, et al. Benefits of a Skull-Interfaced Flexible and Implantable Multilight Emitting Diode Array for Photobiomodulation in Ischemic Stroke. Advanced Science. 2022;9(11):21 04629. doi: https://doi.org/10.1002/advs.202104629

21. Casalechi HL, Dumont AJL, Ferreira LAB, et al. Acute effects of photobiomodulation therapy and magnetic field on functional mobility in stroke survivors: a randomized, sham-controlled, triple-blind, crossover, clinical trial. Lasers in medical science. 2020;35(6):1253-1262.

22. Longo F, Gabriela M, Tan CO, et al. Effect of Transcranial Low-Level Light Therapy vs Sham Therapy Among Patients With Moderate Traumatic Brain Injury: A Randomized Clinical Trial. JAMA Network Open. 2020;3(9):e2017337-e2017337. doi:10.1001/jamanetworkopen.2020.17337

23. Naeser MA, Martin PI, Ho MD, et al. Transcranial, Red/Near-Infrared Light-Emitting Diode Therapy to Improve Cognition in Chronic Traumatic Brain Injury. Photomed Laser Surg. Dec 2016;34(12):610-626. doi:10.1089/pho.2015.4037

24. Naeser MA, Martin PI, Ho MD, et al. Transcranial Photobiomodulation Treatment: Significant Improvements in Four Ex-Football Players with Possible Chronic Traumatic Encephalopathy. Journal of Alzheimer's Disease Reports. 2023;7:77-105. doi:10.3233/ADR-220022

25. Li Y, Dong Y, Yang L, et al. Transcranial photobiomodulation prevents PTSD-like comorbidities in rats experiencing underwater trauma. Translational Psychiatry. 2021;11(1):270.

26. Cassano P, Petrie SR, Mischoulon D, et al. Transcranial Photobiomodulation for the Treatment of Major Depressive Disorder. The ELATED-2 Pilot Trial. Photomed Laser Surg. Dec 2018;36(12):634-646. doi:10.1089/pho.2018.4490

27. Saltmarche AE, Naeser MA, Ho KF, Hamblin MR, Lim L. Significant improvement in cognition in mild to moderately severe dementia cases treated with transcranial plus intranasal photobiomodulation: case series report. Photomedicine and laser surgery. 2017;35(8):432-441.

28. Chao LL. Effects of Home Photobiomodulation Treatments on Cognitive and Behavioral Function, Cerebral Perfusion, and Resting-State Functional Connectivity in Patients with Dementia: A Pilot Trial. Photobiomodul Photomed Laser Surg. Mar 2019;37(3):133-141. doi:10.1089/photob.2018.4555

29. Ceranoglu TA, Cassano P, Hoskova B, et al. Transcranial Photobiomodulation in Adults with High-Functioning Autism Spectrum Disorder: Positive Findings from a Proof-of-Concept Study. Photobiomodulation, Photomedicine, and Laser Surgery. 2022/01/01 2021;40(1):4-12. doi:10.1089 /photob.2020.4986

30. Schiffer F, Khan A, Bolger E, Flynn E, Seltzer WP, Teicher MH. An Effective and Safe Novel Treatment of Opioid Use Disorder: Unilateral Transcranial Photobiomodulation. Front Psychiatry. 2021;12:713686. doi:10.3389/fpsyt.2021.713686

31. de Pauli Paglioni M, Araújo ALD, Arboleda LPA, et al. Tumor safety and side effects of photobiomodulation therapy used for prevention and management of cancer treatment toxicities. A systematic review. Oral oncology. 2019;93:21-28.

32. Cassano P, Norton R, Caldieraro MA, et al. Tolerability and safety of transcranial photobiomodulation for mood and anxiety disorders. Photonics. 2022;9(8):507.

33. Wang EB, Kaur R, Fierro M, Austin E, Jones LR, Jagdeo J. Chapter 5 - Safety and penetration of light into the brain. In: Hamblin MR, Huang Y-Y, eds. Photobiomodulation in the Brain. Academic Press; 2019:49-66.

34. Cassano P, Caldieraro MA, Norton R, et al. Reported Side Effects, Weight and Blood Pressure, After Repeated Sessions of Transcranial Photobiomodulation. Photobiomodulation, photo medicine, and laser surgery. 2019;37(10):651-656.

35. Sharma SK, Sardana S, Hamblin MR. Role of opsins and light or heat activated transient receptor potential ion channels in the mechanisms of photobiomodulation and infrared therapy. Journal of Photochemistry and Photobiology. 2023/02/01/ 2023;13:100160. doi: https://doi.org/10.1016/j.jpap.2023.100160

36. Liebert A, Capon W, Pang V, et al. Photophysical Mechanisms of Photobiomodulation Therapy as Precision Medicine. Biomedicines. 2023;11(2):237.

37. Hamblin MR. Mechanisms and applications of the anti-inflammatory effects of photobio modulation. AIMS biophysics. 05/19 2017;4(3): 337-361. doi:10.3934/biophy.2017.3.337

38. Cardoso FDS, Salehpour F, Coimbra NC, Gonzalez-Lima F, Gomes da Silva S. Photobiomodulation for the treatment of neuroinflammation: A systematic review of controlled laboratory animal studies. Front Neurosci. 2022;16:1006031. doi:10.3389/fnins.20 22.1006031

39. Hamblin MR. Chapter 8 - Mechanisms of photobiomodulation in the brain. In: Hamblin MR, Huang Y-Y, eds. Photobiomodulation in the Brain. Academic Press; 2019:97-110.

40. Shamloo S, Defensor E, Ciari P, et al. The anti-inflammatory effects of photobiomodulation are mediated by cytokines: Evidence from a mouse model of inflammation. Front Neurosci. 2023; 17:1150156. doi:10.3389/fnins.2023.1150156

41. Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Near-infrared light via light-emitting diode treatment is therapeutic against rotenone-and 1-methyl-4-phenylpyridinium ion-induced neurotoxicity. Neuroscience. 2008;153(4):963-974.

42. Ying R, Liang HL, Whelan HT, Eells JT, Wong-Riley MT. Pretreatment with near-infrared light via light-emitting diode provides added benefit against rotenone- and MPP+-induced neurotoxicity. Brain Res. Dec 3 2008;1243:167-73. doi:10.1016/j.brainres.2008.09.057

43. Shaw VE, Spana S, Ashkan K, et al. Neuroprotection of midbrain dopaminergic cells in MPTP‐treated mice after near‐infrared light treatment. Journal of Comparative Neurology. 2010;518(1):25-40.

44. Peoples C, Spana S, Ashkan K, et al. Photobiomodulation enhances nigral dopaminergic cell survival in a chronic MPTP mouse model of Parkinson’s disease. Parkinsonism & related disorders. 2012;18(5):469-476.

45. Moro C, Torres N, El Massri N, et al. Photobiomodulation preserves behaviour and midbrain dopaminergic cells from MPTP toxicity: evidence from two mouse strains. BMC neuroscience. 2013;14(1):40.

46. Reinhart F, El Massri N, Darlot F, et al. 810 nm near-infrared light offers neuroprotection and improves locomotor activity in MPTP-treated mice. Neuroscience Research. 2015;92:86-90.

47. El Massri N, Moro C, Torres N, et al. Near-infrared light treatment reduces astrogliosis in MPTP-treated monkeys. Experimental brain research. 2016;234(11):3225-3232.

48. Reinhart F, Massri NE, Torres N, et al. The behavioural and neuroprotective outcomes when 670nm and 810nm near infrared light are applied together in MPTP-treated mice. Neurosci Res. Apr 2017;117:42-47. doi:10.1016/j.neures.2016.11.006

49. Reinhart F, El Massri N, Chabrol C, et al. Intracranial application of near-infrared light in a hemi-parkinsonian rat model: the impact on behavior and cell survival. Journal of Neurosurgery. 2016;124(6):1829-1841.

50. Moro C, El Massri N, Torres N, et al. Photobiomodulation inside the brain: a novel method of applying near-infrared light intracranially and its impact on dopaminergic cell survival in MPTP-treated mice. Journal of Neurosurgery. 2014;120(3):670-683. doi:doi:10.31 71/2013.9.JNS13423

51. El Massri N, Lemgruber AP, Rowe IJ, et al. Photobiomodulation-induced changes in a monkey model of Parkinson’s disease: changes in tyrosine hydroxylase cells and GDNF expression in the striatum. Experimental brain research. 2017;235(6):1861-1874.

52. El Massri N, Cullen KM, Stefani S, et al. Evidence for encephalopsin immunoreactivity in interneurones and striosomes of the monkey striatum. Exp Brain Res. 04 2018;236(4):955-961. doi:10.1007/s00221-018-5191-9

53. Moro C, El Massri N, Darlot F, et al. Effects of a higher dose of near-infrared light on clinical signs and neuroprotection in a monkey model of Parkinson's disease. Brain Research. 2016;1648:19-26.

54. Purushothuman S, Nandasena C, Johnstone DM, Stone J, Mitrofanis J. The impact of near-infrared light on dopaminergic cell survival in a transgenic mouse model of parkinsonism. Brain research. 2013;1535:61-70.

55. Gordon LC, Martin KL, Torres N, et al. Remote photobiomodulation targeted at the abdomen or legs provides effective neuroprotection against parkinsonian MPTP insult. European Journal of Neuroscience. 2023;57(9):1611-1624.

56. Stone J, Johnstone D, Mitrofanis J. The helmet experiment in Parkinson's disease: an observation of the mechanism of neuroprotection by near infra-red light. 2013:

57. Johnstone DM, Mitrofanis J, Stone J. Targeting the body to protect the brain: inducing neuroprotection with remotely-applied near infrared light. Neural Regen Res. Mar 2015;10 (3):349-51. doi:10.4103/1673-5374.153673

58. Tuby H, Maltz L, Oron U. Induction of Autologous Mesenchymal Stem Cells in the Bone Marrow by Low-Level Laser Therapy Has Profound Beneficial Effects on the Infarcted Rat Heart. Lasers in surgery and medicine. 2011;43:401-409.

59. Bicknell B, Liebert A, Borody T, Herkes G, McLachlan C, Kiat H. Neurodegenerative and Neurodevelopmental Diseases and the Gut-Brain Axis: The Potential of Therapeutic Targeting of the Microbiome. International Journal of Molecular Sciences. 2023;24(11):9577.

60. Bicknell B, Liebert A, Johnstone D, Kiat H. Photobiomodulation of the microbiome: implications for metabolic and inflammatory diseases. Lasers in medical science. 2018;34(2):317-327.

61. Salehpour F, Sadigh-Eteghad s, Mahmoudi J, Kamari F, Cassano P, Hamblin M. Photobio modulation Therapy for Parkinson’s Disease. Photo biomodulation for the Brain Photobiomodulation Therapy in Neurology and Neuropsychiatry. Springer; 2023:chap 10. Synthesis Lectures on Biomedical Engineering.

62. Tajiri N, Yasuhara T, Shingo T, et al. Exercise exerts neuroprotective effects on Parkinson's disease model of rats. Brain research. 2010;1310 :200-207.

63. Svensson M, Lexell J, Deierborg T. Effects of physical exercise on neuroinflammation, neuroplasticity, neurodegeneration, and behavior: what we can learn from animal models in clinical settings. Neurorehabilitation and neural repair. 2015;29(6):577-589.

64. Ferreira AFF, Binda KH, Real CC. The effects of treadmill exercise in animal models of Parkinson’s disease: A systematic review. Neuroscience & Biobehavioral Reviews. 2021;131 :1056-1075.

65. Chahine LM, Darweesh SKL. Physical Activity and the Risk of Parkinson Disease. Neurology. 2023;101(4):151-152. doi:10.1212/WNL.00000000 00207527

66. Fang X, Han D, Cheng Q, et al. Association of levels of physical activity with risk of Parkinson disease: a systematic review and meta-analysis. JAMA network open. 2018;1(5):e182421-e182421.

67. Choi H-y, Cho K-H, Jin C, et al. Exercise Therapies for Parkinson’s Disease: A Systematic Review and Meta-Analysis. Parkinson’s Disease. 2020/09/08 2020;2020:2565320. doi:10.1155/202 0/2565320

68. Gamborg M, Hvid LG, Dalgas U, Langeskov‐Christensen M. Parkinson’s disease and intensive exercise therapy—An updated systematic review and meta‐analysis. Acta Neurologica Scandinavica. 2022;145(5):504-528.

69. Zhen K, Zhang S, Tao X, Li G, Lv Y, Yu L. A systematic review and meta-analysis on effects of aerobic exercise in people with Parkinson's disease. NPJ Parkinsons Dis. Oct 31 2022;8(1):146. doi:10.1038/s41531-022-00418-4

70. Shulman LM, Katzel LI, Ivey FM, et al. Randomized clinical trial of 3 types of physical exercise for patients with Parkinson disease. JAMA neurology. 2013;70(2):183-190.

71. Corcos DM, Robichaud JA, David FJ, et al. A two‐year randomized controlled trial of progressive resistance exercise for Parkinson's disease. Movement Disorders. 2013;28(9):1230-1240.

72. Schenkman M, Moore CG, Kohrt WM, et al. Effect of High-Intensity Treadmill Exercise on Motor Symptoms in Patients With De Novo Parkinson Disease: A Phase 2 Randomized Clinical Trial. JAMA Neurol. Feb 1 2018;75(2):219-226. doi:10.1001/jamaneurol.2017.3517

73. Tucak C, Chih H, Mastaglia F, Rodrigues J. The ‘PD Warrior’exercise program improves motor outcomes and quality of life in patients with early Parkinson's disease: results of a pilot study. Internal Medicine Journal. 2023;

74. van der Kolk NM, de Vries NM, Kessels RP, et al. Effectiveness of home-based and remotely supervised aerobic exercise in Parkinson's disease: a double-blind, randomised controlled trial. The Lancet Neurology. 2019;18(11):998-1008.

75. Johansson ME, Cameron IGM, Van der Kolk NM, et al. Aerobic Exercise Alters Brain Function and Structure in Parkinson's Disease: A Randomized Controlled Trial. Annals of Neurology. 2022/02/01 2022;91(2):203-216. doi: https://doi.org/10.1002/ana.26291

76. de Laat B, Hoye J, Stanley G, et al. Intense exercise increases dopamine transporter and neuromelanin concentrations in the substantia nigra in Parkinson’s disease. npj Parkinson's Disease. 2024/02/09 2024;10(1):34. doi:10.1038/s 41531-024-00641-1

77. Boulos C, Yaghi N, El Hayeck R, Heraoui GN, Fakhoury-Sayegh N. Nutritional risk factors, microbiota and Parkinson’s disease: what is the current evidence? Nutrients. 2019;11(8):1896.

78. Knight E, Geetha T, Burnett D, Babu JR. The Role of Diet and Dietary Patterns in Parkinson’s Disease. Nutrients. 2022;14(21):4472.

79. Hegelmaier T, Lebbing M, Duscha A, et al. Interventional Influence of the Intestinal Microbiome Through Dietary Intervention and Bowel Cleansing Might Improve Motor Symptoms in Parkinson’s Disease. Cells. 2020;9(2):376.

80. Shumway-Cook A, Brauer S, Woollacott M. Predicting the Probability for Falls in Community-Dwelling Older Adults Using the Timed Up & Go Test. Physical Therapy. 2000;80(9):896-903. doi:10.1093/ptj/80.9.896

81. Lang JT, Kassan TO, Devaney LL, Colon-Semenza C, Joseph MF. Test-Retest Reliability and Minimal Detectable Change for the 10-Meter Walk Test in Older Adults With Parkinson's disease. Journal of Geriatric Physical Therapy. 2016;39(4) doi:https://doi.org/10.1519/JPT.0000000000000068

82. Hill K, Bernhardt J, McGann A, Maltese D, Berkovits D. A new test of dynamic standing balance for stroke patients: reliability, validity, and quantitative clinical tests. Physiotherapy Canada. 1996;47:257-262.

83. Pullman SL. Spiral Analysis: A New Technique for Measuring Tremor With a Digitizing Tablet. Movement Disorders. 1998;13(S3):85-89. doi:10.1 002/mds.870131315

84. Smithson F, Morris ME, Iansek R. Performance on Clinical Tests of Balance in Parkinson's Disease. Physical Therapy. 1998;78(6):577-592. doi:10.1093 /ptj/78.6.577

85. Gill DJ, Freshman A, Blender JA, Ravina B. The Montreal cognitive assessment as a screening tool for cognitive impairment in Parkinson's disease. Movement Disorders. 2008;23(7):1043-1046.

86. Peto V, Jenkinson C, Fitzpatrick R. PDQ-39: a review of the development, validation and application of a Parkinson’s disease quality of life questionnaire and its associated measures. Journal of Neurology. 1998/04/01 1998;245(1):S10-S14. doi:10.1007/PL00007730

87. 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. Dec 2002;73(6):629-35. doi:10.1136/ jnnp.73.6.629

88. Ray Chaudhuri K, Martinez-Martin P, Rolfe KA, et al. Improvements in nocturnal symptoms with ropinirole prolonged release in patients with advanced Parkinson’s disease. European Journal of Neurology. 2012;19(1):105-113. doi:https://doi.org/10.1111/j.1468-1331.2011.03442.x

89. Lidstone SC. Great Expectations: The Placebo Effect in Parkinson’s Disease. In: Benedetti F, Enck P, Frisaldi E, Schedlowski M, eds. Placebo. Springer Berlin Heidelberg; 2014:139-147.

90. Quattrone A, Barbagallo G, Cerasa A, Stoessl AJ. Neurobiology of placebo effect in Parkinson's disease: What we have learned and where we are going. Movement Disorders. 2018;33(8):1213-1227. doi:https://doi.org/10.1002/mds.27438

91. Wager TD, Atlas LY. The neuroscience of placebo effects: connecting context, learning and health. Nat Rev Neurosci. Jul 2015;16(7):403-18. doi:10.1038/nrn3976

92. Goetz CG, Leurgans S, Raman R. Placebo-associated improvements in motor function: Comparison of subjective and objective sections of the UPDRS in early Parkinson's disease. Movement Disorders. 2002;17(2):283-288. doi: https://doi.org/10.1002/mds.10024

93. Wilson J, Alcock L, Yarnall AJ, et al. Gait Progression Over 6 Years in Parkinson's Disease: Effects of Age, Medication, and Pathology. Front Aging Neurosci. 2020;12:577435. doi:10.3389/fna gi.2020.577435

94. Schrag A, Dodel R, Spottke A, Bornschein B, Siebert U, Quinn NP. Rate of clinical progression in Parkinson's disease. A prospective study. Movement disorders. 2007;22(7):938-945. doi: https://doi.org/10.1002/mds.21429

95. Lawton M, Ben-Shlomo Y, May MT, et al. Developing and validating Parkinson’s disease subtypes and their motor and cognitive progression. Journal of Neurology, Neurosurgery & Psychiatry. 2018;89(12):1279-1287.

96. Alves G, Wentzel-Larsen T, Aarsland D, Larsen JP. Progression of motor impairment and disability in Parkinson disease. Neurology. 2005;65(9):1436-1441. doi:10.1212/01.wnl.0000183359.50822.f2

97. Holden SK, Finseth T, Sillau SH, Berman BD. Progression of MDS-UPDRS Scores Over Five Years in De Novo Parkinson Disease from the Parkinson's Progression Markers Initiative Cohort. Movement Disorders Clinical Practice. 2018/01/01 2018;5(1) :47-53. doi: https://doi.org/10.1002/mdc3.12553

98. Shalash A, Helmy A, Salama M, Gaber A, El-Belkimy M, Hamid E. A 6-month longitudinal study on worsening of Parkinson's disease during the COVID-19 pandemic. NPJ Parkinsons Dis. Aug 31 2022;8(1):111. doi:10.1038/s41531-022-00376-x

99. Johansson C, Lindström B, Forsgren L, Johansson GM. Balance and mobility in patients with newly diagnosed Parkinson’s disease – a five-year follow-up of a cohort in northern Sweden. Disability and Rehabilitation. 2020/03/12 2020;42 (6):770-778. doi:10.1080/09638288.2018.1509240

100. Morris S, Morris ME, Iansek R. Reliability of measurements obtained with the Timed “Up & Go” test in people with Parkinson disease. Physical therapy. 2001;81(2):810-818. doi: https://doi.org/10.1093/ptj/81.2.810

101. 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. Physical therapy. 2008;88(6):733-746. doi: https://doi.org/10.2522/ptj.20070214

102. Vance RC, Healy DG, Galvin R, French HP. Dual tasking with the timed “up & go” test improves detection of risk of falls in people with Parkinson disease. Physical therapy. 2015;95(1):95-102.

103. Nocera JR, Stegemöller EL, Malaty IA, et al. Using the Timed Up & Go test in a clinical setting to predict falling in Parkinson's disease. Archives of physical medicine and rehabilitation. 2013;94 (7):1300-1305.

104. Allen NE, Schwarzel AK, Canning CG. Recurrent falls in Parkinson’s disease: a systematic review. Parkinson’s disease. 2013;2013 doi: https://doi.org/10.1155/2013/906274

105. Huang Y-C, Hong C-T, Chi W-C, et al. Deterioration of fine motor skills and functional disability in patients with moderate-to-advanced Parkinson disease: A longitudinal follow-up study. Archives of Gerontology and Geriatrics. 2024/06/01/ 2024;121:105366. doi:https://doi.org/10.1016/j.archger.2024.105366

106. Wills A-MA, Elm JJ, Ye R, et al. Cognitive function in 1736 participants in NINDS Exploratory Trials in PD Long-term Study-1. Parkinsonism & Related Disorders. 2016/12/01/ 2016;33:127-133. doi:https://doi.org/10.1016/j.parkreldis.2016.10.005

107. Aarsland D, Andersen K, Larsen JP, et al. The Rate of Cognitive Decline in Parkinson Disease. Archives of Neurology. 2004;61(12):1906-1911. doi:10.1001/archneur.61.12.1906

108. Greenland JC, Camacho M, Williams-Gray CH. Chapter 12 - The dilemma between milestones of progression versus clinical scales in Parkinson's disease. In: Espay AJ, ed. Handbook of Clinical Neurology. Elsevier; 2023:169-185.

109. Tarakad A, Jankovic J. Chapter Seventeen - Anosmia and Ageusia in Parkinson's Disease. In: Chaudhuri KR, Titova N, eds. International Review of Neurobiology. Academic Press; 2017:541-556.

110. Palacios N, Gao X, Schwarzschild M, Ascherio A. Declining quality of life in Parkinson disease before and after diagnosis. J Parkinsons Dis. 2012;2(2):153-60. doi:10.3233/jpd-2012-12083.