We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

5-HT1A agonists for levodopa-induced dyskinesia in Parkinson's disease

    Jawad Al-Kassmy

    Royal College of Surgeons in Ireland, School of Medicine, Dublin, Ireland

    ,
    Christine Sun

    Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada

    &
    Philippe Huot

    *Author for correspondence: Tel.: +1 514 934 8026;

    E-mail Address: philippe.huot@mcgill.ca

    Neurodegenerative Disease Group, Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, H3A 2B4, Canada

    Movement Disorder Clinic, Division of Neurology, Department of Neurosciences, McGill University Health Centre, Montreal, QC, H3A 2B4, Canada

    Department of Neurology & Neurosurgery, McGill University, Montreal, QC, H3A 2B4, Canada

    Published Online:https://doi.org/10.2217/nmt-2022-0039

    Levodopa is the most effective agent for treating the symptoms of Parkinson's disease (PD). However, levodopa-induced dyskinesia remains a significant complication that manifests after few years of treatment, for which therapeutic options remain limited. Several agonists of the serotonin type 1A (5-HT1A) receptor with varying levels of efficacy and interaction at other sites, have been tested in the clinic. Clinical trials testing 5-HT1A agonists have yielded inconsistent results in alleviating dyskinesia, especially that the antidyskinetic benefit observed was often accompanied by an adverse effect on motor function. In this article, we summarize and analyze the various clinical trials performed with 5-HT1A agonists in PD patients with dyskinesia and offer perspectives on the future of this class of agents in PD.

    Plain language summary

    After prolonged treatment with levodopa, patients with Parkinson's disease might start to experience abnormal involuntary movements, called ‘dyskinesias’. These abnormal movements may be difficult to cope with since they can occur for several hours during the day and can hamper the quality of life. A potential approach to reduce the severity of dyskinesia, which has been the focus of extensive research, consists of stimulating a target inside the brain called the 5-HT1A receptor. Several drugs harbouring this mechanism of action have been tested in clinical studies. Here, we provide an overview of these clinical studies and discuss their results.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Parent A, Hazrati LN. Functional anatomy of the basal ganglia. II. The place of subthalamic nucleus and external pallidum in basal ganglia circuitry. Brain Res. Rev. 20(1), 128–154 (1995).
    • 2. Parent A, Hazrati LN. Functional anatomy of the basal ganglia. I. The cortico-basal ganglia-thalamo-cortical loop. Brain Res. Rev. 20(1), 91–127 (1995).
    • 3. Kalia LV, Lang AE. Parkinson's disease. Lancet 386(9996), 896–912 (2015).
    • 4. Hornykiewicz O. A brief history of levodopa. J. Neurol. 257(Suppl. 2), S249–S252 (2010).
    • 5. Hornykiewicz O. L-DOPA: from a biologically inactive amino acid to a successful therapeutic agent. Amino Acids 23(1–3), 65–70 (2002).
    • 6. Lewitt PA. Levodopa for the treatment of Parkinson's disease. N. Engl. J. Med. 359(23), 2468–2476 (2008). • Seminal article detailing the use of levodopa in Parkinson's disease (PD), along with associated complications.
    • 7. Curtze C, Nutt JG, Carlson-Kuhta P, Mancini M, Horak FB. Levodopa is a double-edged sword for balance and gait in people with Parkinson's disease. Mov. Disord. 30(10), 1361–1370 (2015).
    • 8. Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson's disease who were treated with ropinirole or levodopa. 056 Study Group. N. Engl. J. Med. 342(20), 1484–1491 (2000).
    • 9. Parkinson Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized controlled trial. Parkinson study group. JAMA 284(15), 1931–1938 (2000).
    • 10. Hely MA, Morris JG, Reid WG, Trafficante R. Sydney Multicenter Study of Parkinson's disease: non-L-dopa-responsive problems dominate at 15 years. Mov. Disord. 20(2), 190–199 (2005).
    • 11. Verhagen Metman L, Espay AJ. Teaching Video NeuroImages: the underrecognized diphasic dyskinesia of Parkinson disease. Neurology 89(7), e83–e84 (2017).
    • 12. Cenci MA. Presynaptic mechanisms of L-DOPA-induced dyskinesia: the findings, the debate, and the therapeutic implications. Front. Neurol. 5, 242 (2014).
    • 13. Huot P, Johnston TH, Koprich JB, Fox SH, Brotchie JM. The pharmacology of L-DOPA-induced dyskinesia in Parkinson's disease. Pharmacol. Rev. 65(1), 171–222 (2013). • Exhaustive review on the pathophysiology and pharmacological aspects of levodopa-induced dyskinesia.
    • 14. Tran TN, Vo TNN, Frei K, Truong DD. Levodopa-induced dyskinesia: clinical features, incidence, and risk factors. J. Neural. Transm. (Vienna) 125(8), 1109–1117 (2018).
    • 15. Van Gerpen JA, Kumar N, Bower JH, Weigand S, Ahlskog JE. Levodopa-associated dyskinesia risk among Parkinson's disease patients in Olmsted County, Minnesota, 1976–1990. Arch. Neurol. 63(2), 205–209 (2006).
    • 16. Perez-Lloret S, Rascol O. Efficacy and safety of amantadine for the treatment of L-DOPA-induced dyskinesia. J. Neural. Transm. (Vienna) 125(8), 1237–1250 (2018).
    • 17. Durif F, Debilly B, Galitzky M et al. Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 62(3), 381–388 (2004).
    • 18. Alvir JM, Lieberman JA, Safferman AZ, Schwimmer JL, Schaaf JA. Clozapine-induced agranulocytosis. Incidence and risk factors in the United States. N. Engl. J. Med. 329(3), 162–167 (1993).
    • 19. Carta M, Carlsson T, Kirik D, Bjorklund A. Dopamine released from 5-HT terminals is the cause of L-DOPA-induced dyskinesia in parkinsonian rats. Brain 130(Pt 7), 1819–1833 (2007).
    • 20. Nevalainen N, S Af Bjerken, Gerhardt GA, Stromberg I. Serotonergic nerve fibers in L-DOPA-derived dopamine release and dyskinesia. Neuroscience 260, 73–86 (2014).
    • 21. Bishop C, Krolewski DM, Eskow KL et al. Contribution of the striatum to the effects of 5-HT1A receptor stimulation in L-DOPA-treated hemiparkinsonian rats. J. Neurosci. Res. 87(7), 1645–1658 (2009).
    • 22. Huot P, Johnston TH, Fox SH, Newman-Tancredi A, Brotchie JM. The highly-selective 5-HT(1A) agonist F15599 reduces L-DOPA-induced dyskinesia without compromising anti-parkinsonian benefits in the MPTP-lesioned macaque. Neuropharmacology 97, 306–311 (2015).
    • 23. Gregoire L, Samadi P, Graham J, Bedard PJ, Bartoszyk GD, Di Paolo T. Low doses of sarizotan reduce dyskinesias and maintain antiparkinsonian efficacy of L-Dopa in parkinsonian monkeys. Parkinsonism & Related Disorders 15(6), 445–452 (2009).
    • 24. Depoortere R, Johnston TH, Fox SH, Brotchie JM, Newman-Tancredi A. The selective 5-HT1A receptor agonist, NLX-112, exerts anti-dyskinetic effects in MPTP-treated macaques. Parkinsonism Relat. Disord. 78, 151–157 (2020).
    • 25. Ashby CR Jr, Wang RY. Pharmacological actions of the atypical antipsychotic drug clozapine: a review. Synapse 24(4), 349–394 (1996).
    • 26. Anttila SA, Leinonen EV. A review of the pharmacological and clinical profile of mirtazapine. CNS Drug Rev. 7(3), 249–264 (2001).
    • 27. Meco G, Fabrizio E, Di Rezze S, Alessandri A, Pratesi L. Mirtazapine in L-dopa-induced dyskinesias. Clin. Neuropharmacol. 26(4), 179–181 (2003).
    • 28. Shapiro DA, Renock S, Arrington E et al. Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 28(8), 1400–1411 (2003).
    • 29. Bollinger S, Hubner H, Heinemann FW, Meyer K, Gmeiner P. Novel pyridylmethylamines as highly selective 5-HT(1A) superagonists. J. Med. Chem. 53(19), 7167–7179 (2010).
    • 30. Newman-Tancredi A, Martel JC, Cosi C et al. Distinctive in vitro signal transduction profile of NLX-112, a potent and efficacious serotonin 5-HT(1A) receptor agonist. J. Pharm. Pharmacol. 69(9), 1178–1190 (2017).
    • 31. Loane C, Politis M. Buspirone: what is it all about? Brain Res. 1461, 111–118 (2012).
    • 32. Tunnicliff G. Molecular basis of buspirone's anxiolytic action. Pharmacol. Toxicol. 69(3), 149–156 (1991).
    • 33. de Boer SF, Koolhaas JM. 5-HT1A and 5-HT1B receptor agonists and aggression: a pharmacological challenge of the serotonin deficiency hypothesis. Eur. J. Pharmacol. 526(1–3), 125–139 (2005).
    • 34. Schipper J, Tulp MT, Sijbesma H. Neurochemical profile of eltoprazine. Drug Metabol. Drug Interact. 8(1–2), 85–114 (1990).
    • 35. Napier C, Stewart M, Melrose H, Hopkins B, McHarg A, Wallis R. Characterisation of the 5-HT receptor binding profile of eletriptan and kinetics of [3H]eletriptan binding at human 5-HT1B and 5-HT1D receptors. Eur. J. Pharmacol. 368(2–3), 259–268 (1999).
    • 36. Wainscott DB, Krushinski JH Jr, Audia JE et al. [3H]LY334370, a novel radioligand for the 5-HT1F receptor. In vitro characterization of binding properties. Naunyn. Schmiedebergs Arch. Pharmacol. 371(3), 169–177 (2005).
    • 37. Glennon JC, Van Scharrenburg G, Ronken E et al. In vitro characterization of SLV308 (7-[4-methyl-1-piperazinyl]-2(3H)-benzoxazolone, monohydrochloride): a novel partial dopamine D2 and D3 receptor agonist and serotonin 5-HT1A receptor agonist. Synapse 60(8), 599–608 (2006).
    • 38. Rabiner EA, Gunn RN, Wilkins MR, Sedman E, Grasby PM. Evaluation of EMD 128 130 occupancy of the 5-HT1A and the D2 receptor: a human PET study with [11C]WAY-100635 and [11C]raclopride. J. Psychopharmacol. 16(3), 195–199 (2002).
    • 39. Kuzhikandathil EV, Bartoszyk GD. The novel antidyskinetic drug sarizotan elicits different functional responses at human D2-like dopamine receptors. Neuropharmacology 51(4), 873–884 (2006).
    • 40. Hamik A, Oksenberg D, Fischette C, Peroutka SJ. Analysis of tandospirone (SM-3997) interactions with neurotransmitter receptor binding sites. Biol. Psychiatry 28(2), 99–109 (1990).
    • 41. Meco G, Stirpe P, Edito F et al. Aripiprazole in L-dopa-induced dyskinesias: a one-year open-label pilot study. J. Neural. Transm. (Vienna) 116(7), 881–884 (2009).
    • 42. Fernandez HH, Trieschmann ME, Friedman JH. Aripiprazole for drug-induced psychosis in Parkinson disease: preliminary experience. Clin. Neuropharmacol. 27(1), 4–5 (2004).
    • 43. 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. 21(12), 2078–2081 (2006). •• Study with aripiprazole in PD patients that found the drug to be generally poorly tolerated by subjects, as it deteriorated their motor function.
    • 44. Hammerstad JP, Carter J, Nutt JG et al. Buspirone in Parkinson's disease. Clin. Neuropharmacol. 9(6), 556–560 (1986). •• Initial study conducted with buspirone in PD patients.
    • 45. Kleedorfer B, Lees AJ, Stern GM. Buspirone in the treatment of levodopa induced dyskinesias. J. Neurol. Neurosurg. Psychiatry 54(4), 376–377 (1991).
    • 46. Bonifati V, Fabrizio E, Cipriani R, Vanacore N, Meco G. Buspirone in levodopa-induced dyskinesias. Clin. Neuropharmacol. 17(1), 73–82 (1994).
    • 47. Ludwig CL, Weinberger DR, Bruno G et al. Buspirone, Parkinson's disease, and the locus ceruleus. Clin. Neuropharmacol. 9(4), 373–378 (1986).
    • 48. Schneider RB, Auinger P, Tarolli CG, Iourinets J, Gil-Diaz MC, Richard IH. A trial of buspirone for anxiety in Parkinson's disease: safety and tolerability. Parkinsonism Relat. Disord. 81, 69–74 (2020).
    • 49. Scholtissen B, Verhey FR, Adam JJ, Weber W, Leentjens AF. Challenging the serotonergic system in Parkinson disease patients: effects on cognition, mood, and motor performance. Clin. Neuropharmacol. 29(5), 276–285 (2006).
    • 50. Svenningsson P, Rosenblad C, Af Edholm Arvidsson K et al. Eltoprazine counteracts l-DOPA-induced dyskinesias in Parkinson's disease: a dose-finding study. Brain 138(Pt 4), 963–973 (2015). •• Phase II trial in which eltoprazine diminished the severity of levodopa-induced dyskinesia, without interfering with the antiparkinsonian action of levodopa.
    • 51. McFarthing K, Prakash N, Simuni T. Clinical trial highlights – dyskinesia. J. Parkinsons Dis. 9(3), 449–465 (2019).
    • 52. Fabbrini A, Guerra A. Pathophysiological mechanisms and experimental pharmacotherapy for L-Dopa-induced dyskinesia. J. Exp. Pharmacol. 13, 469–485 (2021).
    • 53. Rascol O, Bronzova J, Hauser RA et al. Pardoprunox as adjunct therapy to levodopa in patients with Parkinson's disease experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled, trial. Parkinsonism Relat. Disord. 18(4), 370–376 (2012).
    • 54. Sampaio C, Bronzova J, Hauser RA et al. Pardoprunox in early Parkinson's disease: results from 2 large, randomized double-blind trials. Mov. Disord. 26(8), 1464–1476 (2011). • Results of two phase III trials with pardoprunox in PD showing that the drug is generally poorly tolerated, leading to several adverse events, possibly because of too rapid titration.
    • 55. Bronzova J, Sampaio C, Hauser RA et al. Double-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson's disease. Mov. Disord. 25(6), 738–746 (2010). • Phase III trial in which pardoprunox extended duration of on-time without troublesome dyskinesia. However, there was a high drop-out rate, because of adverse events.
    • 56. Hauser RA, Bronzova J, Sampaio C et al. Safety and tolerability of pardoprunox, a new partial dopamine agonist, in a randomized, controlled study of patients with advanced Parkinson's disease. Eur. Neurol. 62(1), 40–48 (2009).
    • 57. Goetz CG, Damier P, Hicking C et al. Sarizotan as a treatment for dyskinesias in Parkinson's disease: a double-blind placebo-controlled trial. Mov. Disord. 22(2), 179–186 (2007). • Phase III trial in which sarizotan reduced some measures of dyskinesia, yet also exacerbated parkinsonian features.
    • 58. Goetz CG, Laska E, Hicking C et al. Placebo influences on dyskinesia in Parkinson's disease. Mov. Disord. 23(5), 700–707 (2008).
    • 59. Bara-Jimenez W, Bibbiani F, Morris MJ et al. Effects of serotonin 5-HT1A agonist in advanced Parkinson's disease. Mov. Disord. 20(8), 932–936 (2005). • Phase II trial in which sarizotan reduced the severity of levodopa-induced dyskinesia without hindering the antiparkinsonian action of levodopa.
    • 60. Olanow CW, Damier P, Goetz CG et al. Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin. Neuropharmacol. 27(2), 58–62 (2004).
    • 61. Krosser S, Neugebauer R, Chassard D, Kovar A. Investigation of the impact of sarizotan on the pharmacokinetics of levodopa. Biopharm. Drug Dispos. 28(7), 339–347 (2007).
    • 62. Kannari K, Kurahashi K, Tomiyama M et al. Tandospirone citrate, a selective 5-HT1A agonist, alleviates L-DOPA-induced dyskinesia in patients with Parkinson's disease. No To Shinkei 54(2), 133–137 (2002). •• Phase II trial that found that tandospirone alleviated dyskinesia, yet also exacerbated parkinsonism.
    • 63. Nomoto M, Iwata S, Kaseda S. A 5-HT1A receptor agonist, tandospirone improves gait disturbance of patients with Parkinson's disease. J. Mov. Disord. Disability 7, 65–70 (1997).
    • 64. Cenci MA, Riggare S, Pahwa R, Eidelberg D, Hauser RA. Dyskinesia matters. Mov. Disord. 35(3), 392–396 (2020).
    • 65. Hung SW, Adeli GM, Arenovich T, Fox SH, Lang AE. Patient perception of dyskinesia in Parkinson's disease. J. Neurol. Neurosurg. Psychiatry 81(10), 1112–1115 (2010).
    • 66. Eskow KL, Dupre KB, Barnum CJ, Dickinson SO, Park JY, Bishop C. The role of the dorsal raphe nucleus in the development, expression, and treatment of L-dopa-induced dyskinesia in hemiparkinsonian rats. Synapse 63(7), 610–620 (2009).
    • 67. Dupre KB, Ostock CY, Eskow Jaunarajs KL et al. Local modulation of striatal glutamate efflux by serotonin 1A receptor stimulation in dyskinetic, hemiparkinsonian rats. Exp. Neurol. 229(2), 288–299 (2011).
    • 68. Marin C, Aguilar E, Rodriguez-Oroz MC, Bartoszyk GD, Obeso JA. Local administration of sarizotan into the subthalamic nucleus attenuates levodopa-induced dyskinesias in 6-OHDA-lesioned rats. Psychopharmacology (Berl.) 204(2), 241–250 (2009).
    • 69. Klawans HL Jr, Weiner WJ. Attempted use of haloperidol in the treatment of L-dopa induced dyskinesias. J. Neurol. Neurosurg. Psychiatry 37(4), 427–430 (1974).
    • 70. Jackson MJ, Al-Barghouthy G, Pearce RK, Smith L, Hagan JJ, Jenner P. Effect of 5-HT1B/D receptor agonist and antagonist administration on motor function in haloperidol and MPTP-treated common marmosets. Pharmacol. Biochem. Behav. 79(3), 391–400 (2004).
    • 71. Fisher R, Hikima A, Morris R et al. The selective 5-HT1A receptor agonist, NLX-112, exerts anti-dyskinetic and anti-parkinsonian-like effects in MPTP-treated marmosets. Neuropharmacology 167, 107997 (2020).
    • 72. Huot P, Fox SH, Newman-Tancredi A, Brotchie JM. Anatomically selective serotonergic type 1A and serotonergic type 2A therapies for Parkinson's disease: an approach to reducing dyskinesia without exacerbating parkinsonism? J. Pharmacol. Exp. Ther. 339(1), 2–8 (2011). • Article discussing the therapeutic potential of biased 5-HT1A agonists for the treatment of levodopa-induced dyskinesia.
    • 73. Goetz CG, Stebbins GT, Chung KA et al. Which dyskinesia scale best detects treatment response? Mov. Disord. 28(3), 341–346 (2013).
    • 74. Goetz CG, Nutt JG, Stebbins GT. The Unified Dyskinesia Rating Scale: presentation and clinimetric profile. Mov. Disord. 23(16), 2398–2403 (2008).
    • 75. Lohle M, Bremer A, Gandor F et al. Validation of the PD home diary for assessment of motor fluctuations in advanced Parkinson's disease. NPJ Parkinsons Dis. 8(1), 69 (2022).
    • 76. Timpka J, Lohle M, Bremer A et al. Objective observer vs. patient motor state assessments using the PD home diary in advanced Parkinson's disease. Front Neurol. 13, 935664 (2022).
    • 77. Iderberg H, McCreary AC, Varney MA, Cenci MA, Newman-Tancredi A. Activity of serotonin 5-HT(1A) receptor ‘biased agonists’ in rat models of Parkinson's disease and L-DOPA-induced dyskinesia. Neuropharmacology 93, 52–67 (2015).