380
Views
12
CrossRef citations to date
0
Altmetric
Review

An update on the use of non-ergot dopamine agonists for the treatment of Parkinson’s disease

&
Pages 2279-2291 | Received 17 Mar 2020, Accepted 31 Jul 2020, Published online: 17 Aug 2020

References

  • Grandas F Levodopa for the treatment of Parkinson’s disease: current perspectives. Aging health [Internet]. 2006 [cited 2020 Jul 28];2:101–109. Available from: https://www.futuremedicine.com/doi/10.2217/1745509X.2.1.101.
  • Cerri S, Siani F, Blandini F. Investigational drugs in Phase I and Phase II for Levodopa-induced dyskinesias. Expert Opin Investig Drugs. [ Internet]. 2017;26;777–791. Available from http://www.ncbi.nlm.nih.gov/pubmed/28535734
  • Blandini F, Armentero MT. Dopamine receptor agonists for Parkinson’s disease. Expert Opin Investig Drugs. 2014;23:387–410.
  • Fox SH, Katzenschlager R, Lim SY, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018;33:1248–1266.
  • Verschuur CVM, Suwijn SR, Boel JA, et al. Randomized delayed-start trial of levodopa in Parkinson’s disease. N Engl J Med. 2019;380:315–324.
  • Latt MD, Lewis S, Zekry O, et al. Factors to consider in the selection of dopamine agonists for older persons with Parkinson’s disease. Drugs Aging. [ Internet]. 2019;36:189–202.
  • Müller T, Öhm G, Eilert K, et al. Benefit on motor and non-motor behavior in a specialized unit for Parkinson’s disease. J Neural Transm. [ Internet]. 2017;124:715–720.
  • Dubaz OM, Wu S, Cubillos F, et al. Changes in prescribing practices of dopaminergic medications in individuals with Parkinson’s Disease by expert care centers from 2010 to 2017: the Parkinson’s foundation quality improvement initiative. Mov Disord Clin Pract. 2019;6:687–692.
  • Borkar N, Andersson DR, Yang M, et al. Efficacy of oral lipid-based formulations of apomorphine and its diester in a Parkinson’s disease rat model. J Pharm Pharmacol. 2017;69:1110–1115.
  • Tan JPK, Voo ZX, Lim S, et al. Effective encapsulation of apomorphine into biodegradable polymeric nanoparticles through a reversible chemical bond for delivery across the blood–brain barrier. Nanomedicine nanotechnology. BiolMed. [ Internet]. 2019;17:236–245.
  • Shaltiel-Karyo R, Tsarfati Y, Rubinski A, et al. Magnetic resonance imaging as a noninvasive method for longitudinal monitoring of infusion site reactions following administration of a novel apomorphine formulation. Toxicol Pathol. 2017;45:472–480.
  • Domenici RA, Campos ACP, Maciel ST, et al. Parkinson’s disease and pain: modulation of nociceptive circuitry in a rat model of nigrostriatal lesion. Exp Neurol. 2019;315:72–81.
  • Katzenschlager R, Poewe W, Rascol O, et al. Apomorphine subcutaneous infusion in patients with Parkinson’s disease with persistent motor fluctuations (TOLEDO): a multicentre, double-blind, randomised, placebo-controlled trial. Lancet Neurol. 2018;17:749–759.
  • Borgemeester RWK, van Laar T, Schuttelaar MLA. Cutaneous adverse drug reaction after apomorphine infusion, possibly caused by a systemic type IV hypersensitivity reaction to sodium metabisulfite: report of 2 cases. Contact Dermatitis. 2018;79:316–318.
  • Papuć E, Trzciniecka O, Rejdak K. Continuous subcutaneous apomorphine monotherapy in Parkinson’s disease. Ann Agric Environ Med. 2019;26:133–137.
  • Á S, Fernández-Pajarín G, Ares B, et al. Continuous subcutaneous apomorphine in advanced Parkinson’s disease patients treated with deep brain stimulation. J Neurol. 2019;266:659–666.
  • Li BD, Cui JJ, Song J, et al. Comparison of the efficacy of different drugs on non-motor symptoms of Parkinson’s disease: a network meta-analysis. Cell Physiol Biochem. 2018;45:119–130.
  • Martinez-Martin P, Reddy P, Katzenschlager R, et al. EuroInf: A multicenter comparative observational study of apomorphine and levodopa infusion in Parkinson’s disease. Mov Disord. 2015;30:510–516.
  • Houvenaghel JF, Drapier S, Duprez J, et al. Effects of continuous subcutaneous apomorphine infusion in Parkinson’s disease without cognitive impairment on motor, cognitive, psychiatric symptoms and quality of life. J Neurol Sci. [ Internet]. 2018;395:113–118.
  • Dafsari HS, Martinez-Martin P, Rizos A, et al. EuroInf 2: subthalamic stimulation, apomorphine, and levodopa infusion in Parkinson’s disease. Mov Disord. 2019;34:353–365.
  • Pieroni MA Investigation of apomorphine during sleep in Parkinson’s: improvement in UPDRS Scores. Neurol. Int. [ Internet]. 2019 [cited 2020 Jul 28];11. Available from: https://www.pagepress.org/journals/index.php/ni/article/view/8207.
  • Patricio F, Parra I, Martínez I, et al. Effectiveness of fragment C domain of tetanus toxin and pramipexole in an animal model of Parkinson’s disease. Neurotox. Res. [Internet]. 2019 [cited 2020 Jul 28];35:699–710. Available from: http://link.springer.com/10.1007/s12640-018-9990-3.
  • Wang Y, Yu X, Zhang P, et al. Neuroprotective effects of pramipexole transdermal patch in the MPTP-induced mouse model of Parkinson’s disease. J Pharmacol Sci. [ Internet]. 2018;138:5.
  • Li S, Liu J, Li G, et al. Near-infrared light-responsive, pramipexole-loaded biodegradable PLGA microspheres for therapeutic use in Parkinson’s disease. Eur J Pharm Biopharm. 2019;141:1–11.
  • Motyl J, Ł P, Boguszewski PM, et al. Pramipexole and Fingolimod exert neuroprotection in a mouse model of Parkinson’s disease by activation of sphingosine kinase 1 and Akt kinase. Neuropharmacology. 2018;135:139–150.
  • Holtz NA, Tedford SE, Persons AL, et al. Pharmacologically distinct pramipexole-mediated akinesia vs. risk-taking in a rat model of Parkinson’s disease. Prog. Neuro-Psychopharmacology Biol. Psychiatry [Internet]. 2016 [cited 2020 Jul 28];70:77–84. Available from: https://linkinghub.elsevier.com/retrieve/pii/S027858461630077X.
  • Raj R, Wairkar S, Sridhar V, et al. Pramipexole dihydrochloride loaded chitosan nanoparticles for nose to brain delivery: development, characterization and in vivo anti-Parkinson activity. Int J Biol Macromol. [ Internet]. 2018;109:27–35.
  • Berghauzen-Maciejewska K, Kuter K, Kolasiewicz W, et al. Pramipexole but not imipramine or fluoxetine reverses the “depressive-like” behaviour in a rat model of preclinical stages of Parkinson’s disease. Behav Brain Res. [ Internet]. 2014;271:343–353.
  • Favier M, Duran T, Carcenac C, et al. Pramipexole reverses Parkinson’s disease-related motivational deficits in rats. Mov Disord. 2014;29:912–920.
  • Mihaylova AS, Kostadinov ID, Doncheva ND, et al. Effects of pramipexole on learning and memory processes in naïve and haloperidol-challenged rats in active avoidance test. Folia Med (Plovdiv). 2019;61:258–265.
  • Wang Y, Sun S-G, Zhu S-Q, et al. Analysis of pramipexole dose–response relationships in Parkinson’s disease. Drug Des Devel Ther. [ Internet]. 2016;11:83–89. Available from: https://www.dovepress.com/analysis-of-pramipexole-dosendashresponse-relationships-in-parkinson39-peer-reviewed-article-DDDT
  • Olanow CW, Kieburtz K, Leinonen M, et al. A randomized trial of a low-dose Rasagiline and Pramipexole combination (P2B001) in early Parkinson’s disease. Mov Disord. 2017;32:783–789.
  • Pérez-Pérez J, Pagonabarraga J, Martínez-Horta S, et al. Head-to-head comparison of the neuropsychiatric effect of dopamine agonists in Parkinson’s disease: a prospective, cross-sectional study in non-demented patients. Drugs Aging. 2015;32:401–407.
  • Garcia-Ruiz PJ, Martinez Castrillo JC, Desojo LV. Creativity related to dopaminergic treatment: A multicenter study. Park Relat Disord. [ Internet]. 2019;63:169–173.
  • Pardeshi CV, VS B. N,N,N‑trimethyl chitosan modified flaxseed oil based mucoadhesive neuronanoemulsions for direct nose to brain drug delivery. Int J Biol Macromol. [ Internet]. 2018;120:2560–2571.
  • Mustafa G, Ahuja A, Al Rohaimi AH, et al. Nano-ropinirole for the management of Parkinsonism: blood-brain pharmacokinetics and carrier localization. Expert Rev Neurother. 2015;15:695–710.
  • Karavasili C, Bouropoulos N, Sygellou L, et al. PLGA/DPPC/trimethylchitosan spray-dried microparticles for the nasal delivery of ropinirole hydrochloride: in vitro, ex vivo and cytocompatibility assessment. Mater Sci Eng C. [ Internet]. 2016;59:1053–1062.
  • Jafarieh O, Md S, Ali M, et al. Design, characterization, and evaluation of intranasal delivery of ropinirole-loaded mucoadhesive nanoparticles for brain targeting. Drug Dev Ind Pharm. [ Internet]. 2015;41:1674–1681.
  • Patel P, Pol A, More S, et al. Colloidal soft nanocarrier for transdermal delivery of dopamine agonist: ex vivo and in vivo evaluation. J Biomed Nanotechnol. 2014;10:3291–3303.
  • Barcia E, Boeva L, García-García L, et al. Nanotechnology-based drug delivery of ropinirole for parkinson’s disease. Drug Deliv. [ Internet]. 2017;24:1112–1123.
  • Lai KL, Fang Y, Han H, et al. Orally-dissolving film for sublingual and buccal delivery of ropinirole. Colloids Surfaces B Biointerfaces [Internet]. 2018;163:9–18. doi: 10.1016/j.colsurfb.2017.12.015.
  • Zesiewicz TA, Chriscoe S, Jimenez T, et al. A fixed-dose, dose-response study of ropinirole prolonged release in early stage Parkinson’s disease. Neurodegener Dis Manag. 2017;7:49–59.
  • Zesiewicz TA, Chriscoe S, Jimenez T, et al. A randomized, fixed-dose, dose–response study of ropinirole prolonged release in advanced Parkinson’s disease. Neurodegener Dis Manag. [ Internet]. 2017;7:61–72. Available from: https://www.futuremedicine.com/doi/10.2217/nmt-2016-0038
  • Zhuo C, Zhu X, Jiang R, et al. Comparison for efficacy and tolerability among ten drugs for treatment of Parkinson’s disease: a network meta-analysis. Sci Rep. [ Internet]. 2017;8:1–14.
  • Ciurleo R, Bonanno L, Di Lorenzo G, et al. Metabolic changes in de novo Parkinson’s disease after dopaminergic therapy: A proton magnetic resonance spectroscopy study. Neurosci Lett. [ Internet]. 2015;599:55–60.
  • Yan X, Xu L, Bi C, et al. Lactoferrin-modified rotigotine nanoparticles for enhanced nose-to-brain delivery: LESA-MS/MS-based drug biodistribution, pharmacodynamics, and neuroprotective effects. Int J Nanomedicine. 2018;13:273–281.
  • Bhattamisra SK, Shak AT, Xi LW, et al. Nose to brain delivery of rotigotine loaded chitosan nanoparticles in human SH-SY5Y neuroblastoma cells and animal model of Parkinson’s disease. Int J Pharm. 2020;579:119148.
  • Adachi N, Yoshimura A, Chiba S, et al. Rotigotine, a dopamine receptor agonist, increased BDNF protein levels in the rat cortex and hippocampus. Neurosci Lett. [ Internet]. 2018;662:44–50.
  • Isooka N, Miyazaki I, Kikuoka R, et al. Dopaminergic neuroprotective effects of rotigotine via 5-HT1A receptors: possibly involvement of metallothionein expression in astrocytes. Neurochem Int. [ Internet]. 2020;132:104608.
  • Rizos A, Sauerbier A, Falup-Pecurariu C, et al. Tolerability of non-ergot oral and transdermal dopamine agonists in younger and older Parkinson’s disease patients: an European multicentre survey. J Neural Transm. [ Internet]. 2020;127:875–879.
  • Müller T, Tolosa E, Badea L, et al. An observational study of rotigotine transdermal patch and other currently prescribed therapies in patients with Parkinson’s disease. J Neural Transm. [ Internet]. 2018;125:953–963.
  • Serrao M, Ranavolo A, Conte C, et al. Effect of 24-h continuous rotigotine treatment on stationary and non-stationary locomotion in de novo patients with Parkinson disease in an open-label uncontrolled study. J Neurol. 2015;262:2539–2547.
  • Ikeda K, Hirayama T, Takazawa T, et al. Transdermal patch of rotigotine attenuates freezing of gait in patients with Parkinson’s disease: an open-label comparative study of three non-ergot dopamine receptor agonists. Intern Med. 2016;55:2765–2769.
  • Nomoto M, Iwaki H, Kondo H, et al. Efficacy and safety of rotigotine in elderly patients with Parkinson’s disease in comparison with the non-elderly: a post hoc analysis of randomized, double-blind, placebo-controlled trials. J Neurol. 2018;265:253–265.
  • Giladi N, Asgharnejad M, Bauer L, et al. Rotigotine in combination with the MAO-B inhibitor selegiline in early Parkinson’s disease: A post hoc analysis. J Parkinsons Dis. 2016;6:401–411.
  • LeWitt PA, Poewe W, Elmer LW, et al. The efficacy profile of rotigotine during the waking hours in patients with advanced Parkinson’s disease. Clin Neuropharmacol. [ Internet]. 2016;39:88–93. Available from: http://journals.lww.com/00002826-201603000-00004
  • Wang H, Wang L, He Y, et al. Rotigotine transdermal patch for the treatment of neuropsychiatric symptoms in Parkinson’s disease: A meta-analysis of randomized placebo-controlled trials. J Neurol Sci. 2018;393:31–38.
  • Chung SJ, Asgharnejad M, Bauer L, et al. Evaluation of rotigotine transdermal patch for the treatment of depressive symptoms in patients with Parkinson’s disease. Expert Opin Pharmacother. 2016;17:1453–1461.
  • Hauser RA, Slawek J, Barone P, et al. Evaluation of rotigotine transdermal patch for the treatment of apathy and motor symptoms in Parkinson’s disease. BMC Neurol. [ Internet]. 2016;16:1–12.
  • Pierantozzi M, Placidi F, Liguori C, et al. Rotigotine may improve sleep architecture in Parkinson’s disease: A double-blind, Randomized, Placebo-controlled polysomnographic study. Sleep Med. [ Internet]. 2016;21:140–144.
  • Wang Y, Yang YC, Lan DM, et al. An observational clinical and video-polysomnographic study of the effects of rotigotine in sleep disorder in Parkinson’s disease. Sleep Breath. 2017;21:319–325.
  • Calandra-Buonaura G, Guaraldi P, Doria A, et al. Rotigotine objectively improves sleep in Parkinson’s disease: an open-label pilot study with actigraphic recording. Parkinsons Dis. 2016;2016:3724148.
  • Bhidayasiri R, Sringean J, Chaiwong S, et al. Rotigotine for nocturnal hypokinesia in Parkinson’s disease: quantitative analysis of efficacy from a randomized, placebo-controlled trial using an axial inertial sensor. Park Relat Disord. [ Internet]. 2017;44:124–128.
  • Wang Y, Yang Y, Wu H, et al. Effects of rotigotine on REM sleep behavior disorder in Parkinson disease. J Clin Sleep Med. 2016;12:1403–1409.
  • Hirano M, Isono C, Sakamoto H, et al. Rotigotine transdermal patch improves swallowing in dysphagic patients with Parkinson’s disease. Dysphagia. 2015;30:452–456.
  • Schirinzi T, Imbriani P, D’Elia A, et al. Rotigotine may control drooling in patients with Parkinson’s Disease: preliminary findings. Clin Neurol Neurosurg. [ Internet]. 2017;156:63–65.
  • Rascol O, Zesiewicz T, Chaudhuri KR, et al. A randomized controlled exploratory pilot study to evaluate the effect of rotigotine transdermal patch on Parkinson’s disease–associated chronic pain. J Clin Pharmacol. 2016;56:852–861.
  • Olanow CW, Standaert DG, Kieburtz K, et al. Once-weekly subcutaneous delivery of polymer-linked rotigotine (SER-214) provides continuous plasma levels in Parkinson’s disease patients. Mov Disord. 2020;35:1055–1061.
  • Gray R, Ives N, Rick C, et al. Long-term effectiveness of dopamine agonists and monoamine oxidase B inhibitors compared with levodopa as initial treatment for Parkinson’s disease (PD MED): A large, open-label, pragmatic randomised trial. Lancet. 2014;384:1196–1205.
  • Cilia R, Akpalu A, Sarfo FS, et al. The modern pre-levodopa era of Parkinson’s disease: insights into motor complications from sub-Saharan Africa. Brain. 2014;137:2731–2742.
  • Giannakis A, Chondrogiorgi M, Tsironis C, et al. Levodopa-induced dyskinesia in Parkinson’s disease: still no proof? A meta-analysis. J Neural Transm. 2018;125:1341–1349.
  • Lee JY, Jeon B, Koh SB, et al. Behavioural and trait changes in parkinsonian patients with impulse control disorder after switching from dopamine agonist to levodopa therapy: results of REIN-PD trial. J Neurol Neurosurg Psychiatry. 2019;90:30–37.
  • Vargas AP, Vaz LS, Reuter A, et al. Impulse control symptoms in patients with Parkinson’s disease: the influence of dopaminergic agonist. Park Relat Disord. 2019;68:17–21.
  • Corvol JC, Artaud F, Cormier-Dequaire F, et al. Longitudinal analysis of impulse control disorders in Parkinson disease. Neurology. 2018;91:e189–e201.
  • Weintraub D, Koester J, Potenza MN, et al. Impulse control disorders in Parkinson disease. Arch Neurol. [ Internet]. 2010;67. Available from http://archneur.jamanetwork.com/article.aspx?doi=10.1001/archneurol.2010.65
  • Weintraub D, Claassen DO. impulse control and related disorders in Parkinson’s disease. Int Rev Neurobiol. [ Internet]. 2017;133;679–717. Available from http://www.ncbi.nlm.nih.gov/pubmed/28802938
  • Baig F, Kelly MJ, Lawton MA, et al. Impulse control disorders in Parkinson disease and RBD. Neurology. [ Internet]. 2019;93:e675–e687. Available from: http://www.neurology.org/lookup/doi/10.1212/WNL.0000000000007942
  • Weintraub D, David AS, Evans AH, et al. Clinical spectrum of impulse control disorders in Parkinson’s disease. Mov Disord. [ Internet]. 2015;30:121–127. Available from: http://www.ncbi.nlm.nih.gov/pubmed/25370355
  • Leroi I, Harbishettar V, Andrews M, et al. Carer burden in apathy and impulse control disorders in Parkinson’s disease. Int J Geriatr Psychiatry. 2012;27:160–166.
  • Vela L, Martínez Castrillo JC, García Ruiz P, et al. The high prevalence of impulse control behaviors in patients with early-onset Parkinson’s disease: A cross-sectional multicenter study. J Neurol Sci. [ Internet]. 2016;368:150–154.
  • Rizos A, Sauerbier A, Antonini A, et al. A European multicentre survey of impulse control behaviours in Parkinson’s disease patients treated with short- and long-acting dopamine agonists. Eur J Neurol. 2016;23:1255–1261.
  • Antonini A, Barone P, Bonuccelli U, et al. ICARUS study: prevalence and clinical features of impulse control disorders in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2017;88:317–324.
  • Voon V, Napier TC, Frank MJ, et al. Impulse control disorders and levodopa-induced dyskinesias in Parkinson’s disease: an update. Lancet Neurol. [ Internet]. 2017;16:238–250.
  • Weintraub D, Mamikonyan E. Impulse control disorders in Parkinson’s disease. Am J Psychiatry. [ Internet]. 2019;176;5–11. Available from http://www.ncbi.nlm.nih.gov/pubmed/30848949
  • Marín-Lahoz J, Sampedro F, Martinez-Horta S, et al. Depression as a risk factor for impulse control disorders in Parkinson disease. Ann Neurol. 2019;86:762–769.
  • Erga AH, Dalen I, Ushakova A, et al. Dopaminergic and opioid pathways associated with impulse control disorders in Parkinson’s disease. Front Neurol. 2018;9:1–9.
  • Castro-Martínez XH, García-Ruiz PJ, Martínez-García C, et al. Behavioral addictions in early-onset Parkinson disease are associated with DRD3 variants. Park Relat Disord. [ Internet]. 2018;49:100–103.
  • Krishnamoorthy S, Rajan R, Banerjee M, et al. Dopamine D3 receptor Ser9Gly variant is associated with impulse control disorders in Parkinson’s disease patients. Park Relat Disord. [ Internet]. 2016;30:13–17.
  • Kraemmer J, Smith K, Weintraub D, et al. Clinical-genetic model predicts incident impulse control disorders in Parkinson’s disease. J Neurol Neurosurg Psychiatry. [ Internet]. 2016;87:1106–1111.
  • MacDonald HJ, Stinear CM, Ren A, et al. Dopamine gene profiling to predict impulse control and effects of dopamine agonist ropinirole. J Cogn Neurosci. [ Internet]. 2016;28:909–919. .
  • Ramírez Gómez CC, Serrano Dueñas M, Bernal O, et al. A multicenter comparative study of impulse control disorder in Latin American patients with Parkinson disease. Clin Neuropharmacol. [ Internet]. 2017;40:51–55. Available from: http://journals.lww.com/00002826-201703000-00001
  • Engeln M, Ansquer S, Dugast E, et al. Multi-facetted impulsivity following nigral degeneration and dopamine replacement therapy. Neuropharmacology. [ Internet]. 2016;109:69–77.
  • Fluchère F, Burle B, Vidal F, et al. Subthalamic nucleus stimulation, dopaminergic treatment and impulsivity in Parkinson’s disease. Neuropsychologia. 2018;117:167–177.
  • Béreau M, Krack P, Brüggemann N, et al. Neurobiology and clinical features of impulse control failure in Parkinson’s disease. Neurol Res Pract. [ Internet]. 2019;1:9.
  • Martini A, Dal Lago D, Edelstyn NMJ, et al. Dopaminergic neurotransmission in patients with Parkinson’s disease and impulse control disorders: a systematic review and meta-analysis of PET and SPECT studies. Front Neurol. 2018;9:1018.
  • Kon T, Ueno T, Haga R, et al. The factors associated with impulse control behaviors in Parkinson’s disease: A 2-year longitudinal retrospective cohort study. Brain Behav. 2018;8:1–11.
  • Tran T, Brophy JM, Suissa S, et al. Risks of cardiac valve regurgitation and heart failure associated with ergot- and non-ergot-derived dopamine agonist use in patients with Parkinson’s disease: a systematic review of observational studies. CNS Drugs. 2015;29:985–998.
  • De Vecchis R, Cantatrione C, Mazzei D, et al. Non-ergot dopamine agonists do not increase the risk of heart failure in Parkinson’s disease patients: a meta-analysis of randomized controlled trials. J Clin Med Res. 2016;8:449–460.
  • Montastruc F, Moulis F, Araujo M, et al. Ergot and non-ergot dopamine agonists and heart failure in patients with Parkinson’s disease. Eur J Clin Pharmacol. 2017;73:99–103.
  • Erken Pamukcu H, Gerede Uludağ DM, Tekin Tak B, et al. Evaluation of the effect of non-ergot dopamine agonists on left ventricular systolic function with speckle tracking echocardiography. Anatol J Cardiol. 2018;20:213–219.
  • Crispo JAG, Willis AW, Thibault DP, et al. Associations between cardiovascular events and nonergot dopamine agonists in Parkinson’s disease. Mov Disord Clin Pract. 2016;3:257–267.
  • Shen Z, Kong D. Meta-analysis of the adverse events associated with extended-release versus standard immediate-release pramipexole in Parkinson disease. Med (United States). 2018;97:1–6.
  • Li BD, Bi ZY, Liu JF, et al. Adverse effects produced by different drugs used in the treatment of Parkinson’s disease: A mixed treatment comparison. CNS Neurosci Ther. 2017;23:827–842.
  • Makumi CW, Asgharian A, Ellis J, et al. Long-term, open-label, safety study of once-daily ropinirole extended/prolonged release in early and advanced Parkinson’s disease. Int J Neurosci. 2016;126:30–38.
  • Xiang W, Sun YQ, Teoh HC. Comparison of nocturnal symptoms in advanced Parkinson’s disease patients with sleep disturbances: pramipexole sustained release versus immediate release formulations. Drug Des Devel Ther. 2018;12:2017–2024.
  • Antonini A, Bauer L, Dohin E, et al. Effects of rotigotine transdermal patch in patients with Parkinson’s disease presenting with non-motor symptoms - results of a double-blind, randomized, placebo-controlled trial. Eur J Neurol. [ Internet]. 2015;22:1400–1407.
  • Zhang Z-X, Shang H-F, Hu X, et al. Rotigotine transdermal patch in Chinese patients with early Parkinson’s disease: A randomized, double-blind, placebo-controlled pivotal study. Parkinsonism Relat Disord. [ Internet]. 2016;28:49–55. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1353802016301262
  • Zhang Z-X, Liu C-F, Tao E-X, et al. Rotigotine transdermal patch in Chinese patients with advanced Parkinson’s disease: A randomized, double-blind, placebo-controlled pivotal study. Parkinsonism Relat. Disord. [ Internet]. 2017 [cited 2020 Jul 28];44:6–12. https://linkinghub.elsevier.com/retrieve/pii/S1353802017302973.
  • Hindmarsh J, Hindmarsh S, Lee M, et al. The combination of levomepromazine (methotrimeprazine) and rotigotine enables the safe and effective management of refractory nausea and vomiting in a patient with idiopathic Parkinson’s disease. Palliat Med. 2019;33:109–113.
  • Olanow CW, Factor SA, Espay AJ, et al. Apomorphine sublingual film for off episodes in Parkinson’s disease: a randomised, double-blind, placebo-controlled phase 3 study. Lancet Neurol. [ Internet]. 2020;19:135–144. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1474442219303965
  • Hauser RA, Olanow CW, Dzyngel B, et al. Sublingual apomorphine (APL-130277) for the acute conversion of OFF to ON in Parkinson’s disease. Mov Disord. [ Internet]. 2016;31:1366–1372.
  • Zhao H, Ning Y, Cooper J, et al. Indirect comparison of ropinirole and pramipexole as levodopa adjunctive therapy in advanced Parkinson’s disease: a systematic review and network meta-analysis. Adv Ther [ Internet]. 2019;36:1252–1265. doi:10.1007/s12325-019-00938-1.
  • Dogan B, Akyol A, Memis CO, et al. The relationship between temperament and depression in Parkinson’s disease patients under dopaminergic treatment. Psychogeriatrics. 2019;19:73–79.
  • Espay AJ, Foster ED, Coffey CS, et al. Lack of independent mood-enhancing effect for dopaminergic medications in early Parkinson’s disease. J Neurol Sci. [ Internet]. 2019;402:81–85. .
  • Cicero CE, Nicoletti A, Mostile G, et al. A case of severe leg oedema in a patient with Parkinson’s disease treated with pramipexole. Postgrad Med J. [ Internet]. 2016;92:484.
  • Martini ML, Ray C, Yu X, et al. Designing FUNCTIONALLY SELECTIVE NONCATECHOL DOPAMINE D 1 receptor agonists with potent in vivo antiparkinsonian activity. ACS Chem Neurosci. [ Internet]. 2019;10:4160–4182. Available from: https://pubs.acs.org/doi/10.1021/acschemneuro.9b00410
  • Davoren JE, Nason D, Coe J, et al. Discovery and lead optimization of atropisomer D1 agonists with reduced desensitization. J Med Chem. [ Internet]. 2018;61:11384–11397.
  • Sohur US, Gray DL, Duvvuri S, et al. Phase 1 Parkinson’s disease studies show the dopamine D1/D5 agonist PF-06649751 is safe and well tolerated. Neurol Ther. [ Internet]. 2018;7:307–319.
  • Young D, Popiolek M, Trapa P, et al. D1 agonist improved movement of Parkinsonian nonhuman primates with limited dyskinesia side effects. ACS Chem Neurosci. 2020;11:560–566.
  • Papapetropoulos S, Liu W, Duvvuri S, et al. Evaluation of D1/D5 Partial Agonist PF-06412562 in Parkinson’s Disease following Oral Administration. Neurodegener Dis. 2018;02139:262–269.
  • Gurrell R, Duvvuri S, Sun P, et al. A Phase I study of the safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel dopamine D1 receptor partial agonist, PF-06669571, in subjects with idiopathic Parkinson’s disease. Clin Drug Investig. [ Internet]. 2018;38:509–517.
  • Mao Q, Qin W, Zhang A, et al. Recent advances in dopaminergic strategies for the treatment of Parkinson’s disease. Acta Pharmacol Sin. [ Internet]. 2020;41:471–482. Available from: http://www.nature.com/articles/s41401-020-0365-y
  • Voshavar C, Shah M, Xu L, et al. Assessment of protective role of multifunctional dopamine agonist D-512 against oxidative stress produced by depletion of glutathione in PC12 cells: implication in neuroprotective therapy for Parkinson’s disease. Neurotox Res [ Internet]. 2015;28:302–318. doi:10.1007/s12640-015-9548-6.
  • Lindenbach D, Das B, Conti MM, et al. D-512, a novel dopamine D2/3 receptor agonist, demonstrates greater anti-Parkinsonian efficacy than ropinirole in Parkinsonian rats. Br J Pharmacol. 2017;174:3058–3071.
  • Das B, Vedachalam S, Luo D, et al. Development of a highly potent D 2/D 3 agonist and a partial agonist from structure–activity relationship study of N 6 -(2-(4-(1 H -Indol-5-yl)piperazin-1-yl)ethyl)- N 6 -propyl-4,5,6,7-tetrahydrobenzo[d]thiazole-2,6-diamine analogues: implication in t. J Med Chem. [ Internet]. 2015;58:9179–9195.
  • Das B, Kandegedara A, Xu L, et al. A Novel Iron(II) Preferring Dopamine Agonist Chelator as Potential Symptomatic and Neuroprotective Therapeutic Agent for Parkinson’s Disease. ACS Chem Neurosci. 2017;8:723–730.
  • Das B, Rajagopalan S, Joshi GS, et al. A novel iron (II) preferring dopamine agonist chelator D-607 significantly suppresses α-syn- and MPTP-induced toxicities in vivo. Neuropharmacology. [ Internet]. 2017;123:88–99. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0028390817302319
  • Paudel P, Seong SH, Wu S, et al. Eckol as a potential therapeutic against neurodegenerative diseases targeting dopamine D3/D4 receptors. Mar Drugs. 2019;17:108.
  • Paudel P, Seong SH, Jung HA, et al. Characterizing fucoxanthin as a selective dopamine D3/D4 receptor agonist: relevance to Parkinson’s disease. Chem. Biol. Interact. [ Internet]. 2019;310:108757.
  • Seong SH, Paudel P, Choi J-W, et al. Probing multi-target action of phlorotannins as new monoamine oxidase inhibitors and dopaminergic receptor modulators with the potential for treatment of neuronal disorders. Mar Drugs. [ Internet]. 2019;17. Available from http://www.ncbi.nlm.nih.gov/pubmed/31238535
  • Paudel P, Park SE, Seong SH, et al. Bromophenols from symphyocladia latiuscula target human monoamine oxidase and dopaminergic receptors for the management of neurodegenerative diseases. J Agric Food Chem. [ Internet]. 2020;68:2426–2436. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32011134

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.