1
|
Das S, Mccloskey K, Nepal B, Kortagere S. EAAT2 Activation Regulates Glutamate Excitotoxicity and Reduces Impulsivity in a Rodent Model of Parkinson's Disease. Mol Neurobiol 2025; 62:5787-5803. [PMID: 39630405 PMCID: PMC11953204 DOI: 10.1007/s12035-024-04644-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/25/2024] [Indexed: 03/29/2025]
Abstract
Parkinson's disease (PD) is a systemic disease characterized by motor and nonmotor impairments. Loss of dopaminergic neurons in the substantia nigra pars compacta region in PD disrupts dopamine-glutamate homeostasis in the corticostriatal circuit, contributing to cognitive impairment. In addition, excitatory amino acid transporter-2 (EAAT2), localized predominantly to astrocytes and responsible for > 80% of synaptic glutamate clearance, is downregulated in PD, causing glutamate spillover and excitotoxicity. This altered dopamine-glutamate homeostasis and excitotoxicity may affect reward-mediated decision-making behaviors and promote impulsive behaviors in PD. In this study, we hypothesized that GTS467, a small-molecule activator of EAAT2, could effectively reduce excitotoxicity and treat cognitive impairment without promoting impulsive behavior in PD. Rats that were unilaterally lesioned with the 6-OHDA toxin to produce Parkinsonian symptoms were referred to as lesioned rats. Lesioned rats were trained to meet baseline criteria in a 5-choice serial reaction time task, and the chronic effects of GTS467 were assessed after 3 weeks of treatment. The results showed that chronic treatment with GTS467 significantly improved correct responses and reduced premature impulsive responses and omissions compared with saline treatment. This improvement in performance correlated with a reduction in glutamate levels, an increase in EAAT2 expression, and normalization of NMDA receptor subunit expression and signaling. Furthermore, transcriptomic studies on the prefrontal cortex tissue have shown the differential expression of genes involved in neuroprotection, neuroinflammation, learning, and memory. These results validate the role of glutamate excitotoxicity in promoting impulsive behaviors and suggest that GTS467 can be developed as a therapeutic agent to reduce cognitive impairment and impulsive behaviors in PD.
Collapse
Affiliation(s)
- Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Kyle Mccloskey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19129, USA.
| |
Collapse
|
2
|
Svenningsson P, Odin P, Bergquist F, Wirdefeldt K, Nyholm D, Andréasson M, Markaki I, Johansson AC, Jergil M, Jankosky C, Varney MA, Herbrecht F, Johnson SA, Newman-Tancredi A. NLX-112 Randomized Phase 2A Trial: Safety, Tolerability, Anti-Dyskinetic, and Anti-Parkinsonian Efficacy. Mov Disord 2025. [PMID: 40091754 DOI: 10.1002/mds.30175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Levodopa-induced dyskinesia (LID) in Parkinson's disease (PD) is associated with 'false neurotransmitter' release of dopamine from serotonin (5-HT) neurons. NLX-112 is a first-in-class, highly selective 5-HT1A receptor agonist which counteracts LIDs in experimental PD models. OBJECTIVES The primary objective was to evaluate the safety and tolerability of NLX-112 compared with placebo in people with PD. The secondary objective was to assess the preliminary efficacy of NLX-112 in reducing LID and its effects on PD symptoms. METHODS Participants received NLX-112 or placebo (2:1 ratio) alongside stable Parkinson's medications, with 22 participants completing the study. Dosing was up-titrated over 28 days to 2 mg/day (1 mg twice daily), stabilized for 14 days (to day 42), and down-titrated for 14 days. Efficacy was measured using the Unified Dyskinesia Rating Scale (UDysRS), Unified Parkinson's Disease Rating Scale (UPDRS), and Clinical Global Impression of Change (CGI-C) following a levodopa challenge (150% of usual dose). RESULTS Adverse events (AEs) were mainly central nervous system (CNS)-related and mostly occurred during up-titration, with no serious AEs in the NLX-112 group. There were no treatment-induced clinically significant changes in vital signs, electrocardiogram, or laboratory parameters. NLX-112 reduced LID from baseline levels: at day 42, UDysRS total score decreased by 6.3 points, whereas placebo group changes were not significant (-2.4). NLX-112 also reduced parkinsonism from baseline values: UPDRS Part 3 scores decreased by 3.7 points, whereas placebo group changes were non-significant (+0.1). In CGI-C assessment, the NLX-112 group showed greater improvement than the placebo group (53% vs. 29%). CONCLUSION These results support further clinical investigation of NLX-112 for treatment of PD LID. © 2025 Neurolixis SAS. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Per Odin
- Department of Clinical Sciences, Lund University, Skåne University Hospital, Lund, Sweden
| | - Filip Bergquist
- Sahlgrenska Hospital, Gothenburg, Sweden
- Department of Pharmacology, University of Gothenburg, Gothenburg, Sweden
| | - Karin Wirdefeldt
- Department of Neurology, Karolinska University Hospital, Solna, Sweden
| | | | | | | | | | - Måns Jergil
- Clinical Trial Consultants, AB, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
3
|
Isaacson SH, Jenner P. Moving to a non-dopaminergic approach for the treatment of OFF fluctuations in Parkinson's disease. Clin Park Relat Disord 2025; 12:100303. [PMID: 39968317 PMCID: PMC11834069 DOI: 10.1016/j.prdoa.2025.100303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
In levodopa treated patients with Parkinson's disease (PD), the standard approach to managing motor fluctuations is to adjust dopaminergic therapy. However, despite the availability of a wide armamentarium of dopaminergic medications, most patients treated with levodopa will still experience significant OFF time, and it is increasingly clear that motor fluctuations have a significant non-dopaminergic component. In this narrative review, we compare and contrast the therapeutic profiles of the only two non-dopaminergic medications approved in the US for the management of OFF time, namely amantadine and istradefylline. When compared against each other the two agents exemplify two different pharmacological approaches to treatment. Whereas amantadine has a multimodal pharmacology, istradefylline has highly specific actions at A2A receptors which are highly expressed in the indirect pathway of the basal ganglia. We discuss how both offer an important alternative approach to treatment, without increasing total dopaminergic load. Clinicians can also consider that amantadine and istradefylline each have overlapping indications with classic dopaminergic medications, but with distinct mechanisms of action that can complement each other to reduce motor complications in patients already being treated with other dopaminergic agents.
Collapse
Affiliation(s)
- Stuart H. Isaacson
- Parkinson’s Disease and Movement Disorders Center of Boca Raton Boca Raton FL USA
| | - Peter Jenner
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London London UK
| |
Collapse
|
4
|
Shuklinova O, Neuhoff S, Polak S. How PBPK Can Help to Understand Old Drugs and Inform their Dosing in Elderly: Amantadine Case Study. Clin Pharmacol Ther 2024; 116:225-234. [PMID: 38666589 DOI: 10.1002/cpt.3276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/17/2024] [Indexed: 06/18/2024]
Abstract
Amantadine, despite being on the market for 55 years, has several unknown aspects of its pharmacokinetics especially related to the influence of covariates such as age, disease, or interactions linked to amantadine's renal elimination. As amantadine is used in Parkinson's disease and is considered a potential candidate in COVID treatment and other diseases, there is an unmet need for thorough understanding of its pharmacokinetic in special populations, such as the elderly. We aimed to mechanistically describe amantadine pharmacokinetics in healthy subjects and shed some light on the differences in drug behavior between healthy volunteers (18-65 years) and an elderly/geriatric population (65-98 years) using PBPK modeling and simulation. The middle-out PBPK model includes mechanistic description of drug renal elimination, specifically an organic cation transporter (OCT)2-mediated electrogenic bidirectional transport (basolateral) and multidrug and toxic compound extrusion (MATE)1-mediated efflux (apical). The model performance was verified against plasma and urine data reported after single and multiple dose administration in healthy volunteers and elderly patients from 18 independent studies. The ratios of predicted vs. observed maximal plasma concentration and area under the concentration-time curve values were within 1.25-fold. The model illustrates that renal transporter activity is expected to decrease in healthy elderly compared to healthy volunteers, which is in line with literature proteomic data for OCT2. The model was applied to assess the potential of reaching toxicity-related plasma concentrations in different age groups of geriatric subjects.
Collapse
Affiliation(s)
- Olha Shuklinova
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Kraków, Poland
- Simcyp Division, Certara UK Limited, Sheffield, UK
| | | | - Sebastian Polak
- Simcyp Division, Certara UK Limited, Sheffield, UK
- Faculty of Pharmacy, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
5
|
Espay AJ, Ostrem JL, Formella AE, Tanner CM. Extended-release amantadine for OFF-related dystonia in Parkinson's disease. Parkinsonism Relat Disord 2024; 122:106088. [PMID: 38461688 DOI: 10.1016/j.parkreldis.2024.106088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/16/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Dystonia is a painful OFF-related complication in Parkinson's disease (PD) with limited treatment options. METHODS Post-hoc analysis using pooled data from two extended-release amantadine pivotal trials and follow-on open-label extension. Dystonia was assessed using the Unified Dyskinesia Rating Scale (UDysRS) Part 2 and the Movement Disorder Society-Unified PD Rating Scale (MDS-UPDRS) item 4.6. RESULTS Of 196 participants, 119 (60.7%) reported OFF-related dystonia at baseline per UDysRS. Twelve-week treatment with extended-release amantadine improved OFF dystonia (treatment differences vs placebo: UDysRS Part 2, -1.0 [-1.9,-0.1]; p = 0.03 and MDS-UPDRS Item 4.6, -0.3 [-0.6,-0.05]; p = 0.02). There was no correlation between changes in OFF time and changes in OFF dystonia. Double-blind improvements in OFF dystonia were sustained throughout the 2-year follow-up. CONCLUSIONS Extended-release amantadine yielded a sustained reduction in OFF-related dystonia in PD patients that was independent from a reduction in OFF time. A randomized controlled trial is warranted to confirm these findings.
Collapse
Affiliation(s)
- Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Cincinnati, OH, 45267, USA; Department of Neurology, University of Cincinnati, Cincinnati, OH, 45267, USA.
| | - Jill L Ostrem
- Movement Disorder and Neuromodulation Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94143, USA.
| | | | - Caroline M Tanner
- Movement Disorder and Neuromodulation Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
6
|
Gupta HV, Lenka A, Dhamija RK, Fasano A. A video-atlas of levodopa-induced dyskinesia in Parkinson's disease: terminology matters. Neurol Sci 2024; 45:1389-1397. [PMID: 37987930 DOI: 10.1007/s10072-023-07209-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Dyskinesia is a common complication of long-term levodopa therapy in patients with Parkinson's disease (PD), which often worsens the quality of life. It is usually dose-dependent and emerges possibly due to pulsatile stimulation of dopamine receptors. Delineating the pattern of dyskinesia is crucial for determining the most effective therapeutic approach, a task that often presents challenges for numerous neurologists. This article comprehensively describes various patterns of dyskinesia in PD patients and features video demonstration of some of the common forms of dyskinesia. We have used a real case scenario as an example to lead the discussion on the phenomenology, distinguishing features, and management of various types of dyskinesia. A comprehensive literature search was conducted in PubMed using "dyskinesia" as a keyword. The prototype case with videos highlights the differentiating features of dyskinesia along with the treatment strategies. A wide range of descriptive rubrics have been used for certain dyskinesia which are described in detail in this article. The newer types of dyskinesia associated with continuous dopaminergic stimulation in patients with advanced PD and their implications have been described. As there are distinct ways of managing various types of dyskinesia, understanding the phenomenology and chronology of dyskinesia is vital for the optimal management of dyskinetic PD patients. We suggest that dyskinesia should be classified broadly into peak-dose dyskinesia (PDD), biphasic dyskinesia (BD), and OFF-period dystonia. The occurrence of low-dose dyskinesia and complex dyskinesia of continuous dopaminergic treatments should be known to specialists and will require additional studies.
Collapse
Affiliation(s)
- Harsh V Gupta
- Department of Neurology, Memorial Healthcare System, Hollywood, FL, USA.
| | - Abhishek Lenka
- Department of Neurology, Baylor College of Medicine, Houston, TX, USA
| | - Rajinder K Dhamija
- Department of Neurology, Institute of Human Behaviour and Allied Sciences, New Delhi, India
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, UHN, Toronto, ON, Canada
- Division of Neurology, University of Toronto, Toronto, ON, Canada
- Krembil Brain Institute, Toronto, ON, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, Canada
| |
Collapse
|
7
|
Malar DS, Thitilertdecha P, Ruckvongacheep KS, Brimson S, Tencomnao T, Brimson JM. Targeting Sigma Receptors for the Treatment of Neurodegenerative and Neurodevelopmental Disorders. CNS Drugs 2023; 37:399-440. [PMID: 37166702 PMCID: PMC10173947 DOI: 10.1007/s40263-023-01007-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
The sigma-1 receptor is a 223 amino acid-long protein with a recently identified structure. The sigma-2 receptor is a genetically unrelated protein with a similarly shaped binding pocket and acts to influence cellular activities similar to the sigma-1 receptor. Both proteins are highly expressed in neuronal tissues. As such, they have become targets for treating neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), Parkinson's disease (PD), multiple sclerosis (MS), Rett syndrome (RS), developmental and epileptic encephalopathies (DEE), and motor neuron disease/amyotrophic lateral sclerosis (MND/ALS). In recent years, there have been many pre-clinical and clinical studies of sigma receptor (1 and 2) ligands for treating neurological disease. Drugs such as blarcamesine, dextromethorphan and pridopidine, which have sigma-1 receptor activity as part of their pharmacological profile, are effective in treating multiple aspects of several neurological diseases. Furthermore, several sigma-2 receptor ligands are under investigation, including CT1812, rivastigmine and SAS0132. This review aims to provide a current and up-to-date analysis of the current clinical and pre-clinical data of drugs with sigma receptor activities for treating neurological disease.
Collapse
Affiliation(s)
- Dicson S Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kanokphorn S Ruckvongacheep
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - James M Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand.
- Research, Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Room 409, ChulaPat-1 Building, 154 Rama 1 Road, Bangkok, 10330, Thailand.
| |
Collapse
|
8
|
Caulfield ME, Vander Werp MJ, Stancati JA, Collier TJ, Sortwell CE, Sandoval IM, Manfredsson FP, Steece-Collier K. Downregulation of striatal CaV1.3 inhibits the escalation of levodopa-induced dyskinesia in male and female parkinsonian rats of advanced age. Neurobiol Dis 2023; 181:106111. [PMID: 37001610 DOI: 10.1016/j.nbd.2023.106111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
In the past 25 years, the prevalence of Parkinson's disease (PD) has nearly doubled. Age remains the primary risk factor for PD and as the global aging population increases this trend is predicted to continue. Even when treated with levodopa, the gold standard dopamine (DA) replacement therapy, individuals with PD frequently develop therapeutic side effects. Levodopa-induced dyskinesia (LID), a common side effect of long-term levodopa use, represents a significant unmet clinical need in the treatment of PD. Previously, in young adult (3-month-old) male parkinsonian rats, we demonstrated that the silencing of CaV1.3 (Cacan1d) L-type voltage-gated calcium channels via striatal delivery of rAAV-CaV1.3-shRNA provides uniform protection against the induction of LID, and significant reduction of established severe LID. With the goal of more closely replicating a clinical demographic, the current study examined the effects of CaV1.3-targeted gene therapy on LID escalation in male and female parkinsonian rats of advanced age (18-month-old at study completion). We tested the hypothesis that silencing aberrant CaV1.3 channel activity in the parkinsonian striatum would prevent moderate to severe dyskinesia with levodopa dose escalation. To test this hypothesis, 15-month-old male and female F344 rats were rendered unilaterally parkinsonian and primed with low-dose (3-4 mg/kg) levodopa. Following the establishment of stable, mild dyskinesias, rats received an intrastriatal injection of either the Cacna1d-specific rAAV-CaV1.3-shRNA vector (CAV-shRNA), or the scramble control rAAV-SCR-shRNA vector (SCR-shRNA). Daily (M-Fr) low-dose levodopa was maintained for 4 weeks during the vector transduction and gene silencing window followed by escalation to 6 mg/kg, then to 12 mg/kg levodopa. SCR-shRNA-shRNA rats showed stable LID expression with low-dose levodopa and the predicted escalation of LID severity with increased levodopa doses. Conversely, complex behavioral responses were observed in aged rats receiving CAV-shRNA, with approximately half of the male and female subjects-therapeutic 'Responders'-demonstrating protection against LID escalation, while the remaining half-therapeutic 'Non-Responders'-showed LID escalation similar to SCR-shRNA rats. Post-mortem histological analyses revealed individual variability in the detection of Cacna1d regulation in the DA-depleted striatum of aged rats. However, taken together, male and female therapeutic 'Responder' rats receiving CAV-shRNA had significantly less striatal Cacna1d in their vector-injected striatum relative to contralateral striatum than those with SCR-shRNA. The current data suggest that mRNA-level silencing of striatal CaV1.3 channels maintains potency in a clinically relevant in vivo scenario by preventing dose-dependent dyskinesia escalation in rats of advanced age. As compared to the uniform response previously reported in young male rats, there was notable variability between individual aged rats, particularly females, in the current study. Future investigations are needed to derive the sex-specific and age-related mechanisms which underlie variable responses to gene therapy and to elucidate factors which determine the therapeutic efficacy of treatment for PD.
Collapse
|
9
|
Zhu W, Fan Y, Li Y, Peng L, Li Y, Yan F, Zhao J, Zhang L, Kurihara H, He R, Chen H. Hybridization of Amantadine with Gardenamide A Enhances NMDA Antagonism and in vivo Anti-PD Effects. Bioorg Chem 2022; 130:106223. [DOI: 10.1016/j.bioorg.2022.106223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
|
10
|
Amantadine extended release capsules (GOCOVRI®) in Parkinson’s disease: a profile of its use in the USA. DRUGS & THERAPY PERSPECTIVES 2022. [DOI: 10.1007/s40267-022-00912-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
11
|
Bandopadhyay R, Mishra N, Rana R, Kaur G, Ghoneim MM, Alshehri S, Mustafa G, Ahmad J, Alhakamy NA, Mishra A. Molecular Mechanisms and Therapeutic Strategies for Levodopa-Induced Dyskinesia in Parkinson's Disease: A Perspective Through Preclinical and Clinical Evidence. Front Pharmacol 2022; 13:805388. [PMID: 35462934 PMCID: PMC9021725 DOI: 10.3389/fphar.2022.805388] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/21/2022] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the second leading neurodegenerative disease that is characterized by severe locomotor abnormalities. Levodopa (L-DOPA) treatment has been considered a mainstay for the management of PD; however, its prolonged treatment is often associated with abnormal involuntary movements and results in L-DOPA-induced dyskinesia (LID). Although LID is encountered after chronic administration of L-DOPA, the appearance of dyskinesia after weeks or months of the L-DOPA treatment has complicated our understanding of its pathogenesis. Pathophysiology of LID is mainly associated with alteration of direct and indirect pathways of the cortico-basal ganglia-thalamic loop, which regulates normal fine motor movements. Hypersensitivity of dopamine receptors has been involved in the development of LID; moreover, these symptoms are worsened by concurrent non-dopaminergic innervations including glutamatergic, serotonergic, and peptidergic neurotransmission. The present study is focused on discussing the recent updates in molecular mechanisms and therapeutic approaches for the effective management of LID in PD patients.
Collapse
Affiliation(s)
- Ritam Bandopadhyay
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Nainshi Mishra
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Ruhi Rana
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Gagandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Gulam Mustafa
- College of Pharmacy (Boys), Al-Dawadmi Campus, Shaqra University, Riyadh, Saudi Arabia
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Nabil. A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)—Guwahati, Guwahati, India
| |
Collapse
|
12
|
Hauser RA, Lytle J, Formella AE, Tanner CM. Amantadine delayed release/extended release capsules significantly reduce OFF time in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:29. [PMID: 35304480 PMCID: PMC8933492 DOI: 10.1038/s41531-022-00291-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 02/17/2022] [Indexed: 11/16/2022] Open
Abstract
Maintaining consistent levodopa benefits while simultaneously controlling dyskinesia can be difficult. Recently, an amantadine delayed release/extended release (DR/ER) formulation (Gocovri®) indicated for dyskinesia received additional FDA approval as an adjunct to levodopa for the treatment of OFF episodes. We evaluated OFF time reductions with amantadine-DR/ER in a pooled analysis of two phase III amantadine-DR/ER trials (NCT02136914, NCT02274766) followed by a 2-year open-label extension trial (NCT02202551). OFF outcomes were analyzed for the mITT population, as well as stratified by baseline OFF time of ≥2.5 h/day or <2.5 h/day. At Week 12, mean placebo-subtracted treatment difference in OFF time was −1.00 [−1.57, −0.44] h in the mITT population (n = 196), −1.2 [−2.08, −0.32] h in the ≥2.5 h subgroup (n = 102) and −0.77 [−1.49, −0.06] in the <2.5 h subgroup (n = 94). Amantadine-DR/ER-treated participants showed reduced MDS-UPDRS Part IV motor fluctuation subscores by week 2 that were maintained below baseline to Week 100.
Collapse
Affiliation(s)
| | - Judy Lytle
- Adamas Pharmaceuticals, Inc, Emeryville, CA, USA
| | | | | |
Collapse
|
13
|
Battista VD, Hey-Hawkins E. Development of Prodrugs for Treatment of Parkinson's Disease: New Inorganic Scaffolds for Blood-Brain Barrier Permeation. J Pharm Sci 2022; 111:1262-1279. [PMID: 35182542 DOI: 10.1016/j.xphs.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/13/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
Abstract
The treatment of Parkinson's disease (PD) has not been consistently modified for more than 60 years. L-DOPA, the blood-brain barrier permeable precursor prodrug of dopamine, is to date the only effective therapy on the market. However, it is well known that prolonged treatment with L-DOPA leads to several side effects, which may affect the patient's life expectancy (i.e., the wearing-off phenomenon, on-off fluctuations, and dyskinesia). For this reason, modifications, and supplements to L-DOPA treatment have been and are being studied, which, however, have not yet resulted in a valid alternative to the cornerstone drug. This review aims to summarize the main formulations currently in use for PD treatment, explaining advantages and disadvantages for each class. The attention will be focused on the promising prodrug concept, aimed at finding a suitable L-DOPA substitute with improved pharmacokinetic behavior. In this respect, new potential candidates which show interesting properties for the intended scope, the so-called dicarba-closo-dodecaboranes(12) (carboranes), will be discussed. Carboranes are inorganic molecular icosahedral boron-carbon clusters with 12 vertices and 20 deltahedral faces. They have been extensively studied for applications in medicine as potential pharmacophores, reagents in boron neutron capture therapy (BNCT) and radiotherapy. Here, we discuss them as inorganic scaffolds for dopamine delivery at the central nervous system (CNS) level.
Collapse
Key Words
- %F, Oral Bioavailability
- 5-HTP, L-5-Hydroxy-Tryptophan
- AADC, Aromatic L-Amino Acid Decarboxylase
- AGPs, Arabinogalactan Proteins
- AUC, Area Under the Plasma Concentration Curve
- Abbreviations
- BBB, Blood–Brain Barrier
- BNCT, Boron Neutron Capture Therapy
- CNS, Central Nervous System
- COMT, Catechol-O-Methyltransferase
- DBS, Deep Brain Stimulation
- DDC, Dopamine Decarboxylase
- DMSO, Dimethylsulfoxide
- FAD, Flavin Adenine Dinucleotide
- FDA, Food and Drug Administration
- GPCRs, G-Protein-Coupled Receptors
- HIV, Human Immunodeficiency Virus
- HSA, Human Serum Albumin
- ICT, Intramolecular Charge Transfer
- IPG, Implanted Pulse Generator
- IUPAC, International Union of Pure and Applied Chemistry
- IV, Intravenous Injection
- LDEE, L-DOPA Ethyl Ester
- LNAA, Large Neutral Amino Acid transport system
- MAO-A/B, Monoamine Oxidase-A/B
- MPO, Multiparameter Optimization
- Mw, Molecular Weight
- NMDAR, N-Methyl D-Aspartate Receptor
- P, Partition Coefficient
- PAMPA, Parallel Artificial Membrane Permeability Assay
- PD, Parkinson's Disease
- PLP, Pyridoxal Phosphate
- PNS, Peripheral Nervous System
- Parkinson's disease, Dopamine, Blood–brain barrier, Permeability, Bioavailability, L-DOPA, Prodrugs, Inorganic scaffold, Icosahedral carborane
- SAM, S-Adenosyl L-Methionine
- STN, Subthalamic Nucleus
- TBP, Tetrahydrobiopterin
- UPDRS, Unified Parkinson's Disease Rating Scale
- VTA, Ventral Tegmental Are
- hBMECs, human Brain Microvascular Endothelial Cells
Collapse
Affiliation(s)
- Veronica Di Battista
- Leipzig University, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany
| | - Evamarie Hey-Hawkins
- Leipzig University, Faculty of Chemistry and Mineralogy, Institute of Inorganic Chemistry, Johannisallee 29, 04103 Leipzig, Germany.
| |
Collapse
|
14
|
Hauser RA, Goud S, Formella AE. Potential utility of amantadine DR/ER in persons with Parkinson’s disease meeting 5-2-1 criteria for device aided therapy. Clin Park Relat Disord 2022; 6:100123. [PMID: 35059622 PMCID: PMC8760552 DOI: 10.1016/j.prdoa.2021.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
5-2-1 criteria help identify PD patients who may benefit from device aided therapy. Amantadine-DR/ER increased ‘good’ ON time in patients meeting 5-2-1 criteria. Increased good ON time resulted from reductions in troublesome dyskinesia and OFF. Amantadine-DR/ER efficacy was maintained over 100 weeks in this advanced population.
Background The 5-2-1 criteria (≥5 levodopa doses/day, ≥2 h OFF/day, and ≥ 1-hour dyskinesia/day) propose to identify people with Parkinson’s disease (PD) who are poorly controlled on oral therapies and who may therefore benefit from device-aided therapies. Amantadine-DR/ER is the only medication FDA-approved for both dyskinesia and OFF episodes in levodopa-treated patients. In this post-hoc analysis of phase 3 clinical trials, we evaluated the efficacy and safety of amantadine-DR/ER in patients meeting 5-2-1 criteria. Methods Week-12 treatment differences (Amantadine-DR/ER − placebo) in the Unified Dyskinesia Rating Scale (UDysRS) and PD motor states (patient diaries) were evaluated in pooled, phase-3, double-blind trial participants meeting 5-2-1 criteria at baseline. This 5-2-1 cohort was followed into a 2-year open-label trial, where Movement Disorder Society - Unified Parkinson’s Disease Rate Scale (MDS-UPDRS) Part IV scores were assessed relative to double-blind baseline. Results Of 198 enrolled participants in the phase 3 trials, 65 (33%; n = 29 placebo; n = 36 amantadine-DR/ER) comprised the 5-2-1 cohort. At Week-12 endpoint, amantadine-DR/ER significantly improved UDysRS scores (treatment difference of 9.57 ± 3.15 points, p = 0.004) and ON time without troublesome dyskinesia (‘good ON’, treatment difference of 2.9 ± 0.90 h/day, p = 0.002). Improvements in good ON time resulted from significant reductions in both troublesome dyskinesia and OFF time. Treatment benefit on MDS-UPDRS-Part IV was sustained through open-label, follow-up. The most common adverse events in patients who met 5-2-1 criteria and were treated with amantadine-DR/ER included falls and peripheral edema. Conclusions Findings suggest Amantadine-DR/ER should be considered as an option for people with PD who meet 5-2-1 criteria.
Collapse
|
15
|
Oh T, Daadi ES, Kim J, Daadi EW, Chen PJ, Roy-Choudhury G, Bohmann J, Blass BE, Daadi MM. Dopamine D3 receptor ligand suppresses the expression of levodopa-induced dyskinesia in nonhuman primate model of parkinson's disease. Exp Neurol 2022; 347:113920. [PMID: 34762921 DOI: 10.1016/j.expneurol.2021.113920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 11/04/2022]
Abstract
Parkinson's disease (PD) is a complex multisystem, chronic and so far incurable disease with significant unmet medical needs. The incidence of PD increases with aging and the expected burden will continue to escalate with our aging population. Since its discovery in the 1961 levodopa has remained the gold standard pharmacotherapy for PD. However, the progressive nature of the neurodegenerative process in and beyond the nigrostriatal system causes a multitude of side effects, including levodopa-induced dyskinesia within 5 years of therapy. Attenuating dyskinesia has been a significant challenge in the clinical management of PD. We report on a small molecule that eliminates the expression of levodopa-induced dyskinesia and significantly improves PD-like symptoms. The lead compound PD13R we discovered is a dopamine D3 receptor partial agonist with high affinity and selectivity, orally active and with desirable drug-like properties. Future studies are aimed at developing this lead compound for treating PD patients with dyskinesia.
Collapse
Affiliation(s)
- Thomas Oh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Elyas S Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Jeffrey Kim
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA; Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA
| | - Etienne W Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Peng-Jen Chen
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Gourav Roy-Choudhury
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Benjamin E Blass
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Marcel M Daadi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA; Cell Systems & Anatomy, University of Texas Health at San Antonio, San Antonio, TX, USA; Radiology, Long School of Medicine, University of Texas Health at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
16
|
Marmol S, Feldman M, Singer C, Margolesky J. Amantadine Revisited: A Contender for Initial Treatment in Parkinson's Disease? CNS Drugs 2021; 35:1141-1152. [PMID: 34648150 DOI: 10.1007/s40263-021-00862-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
The best practice for the initiation of symptomatic motor treatment for Parkinson's disease is an ongoing topic of debate. Fueled by interpretation of the results of the LEAP and MED Parkinson's disease studies, many practitioners opt for early initiation of levodopa formulations, avoiding dopamine agonists to circumvent potential deleterious side effects, namely impulse control disorder. Compared with levodopa, monoamine oxidase inhibitors may lack necessary potency. Ignored in this academic debate is another therapeutic option for patients with Parkinson's disease requiring treatment initiation: amantadine. Amantadine was first reported effective in the treatment of Parkinson's disease in 1969 and several studies were published in the 1970s supporting its efficacy. Currently, amantadine is mainly utilized as an add-on therapy to mitigate levodopa-related dyskinesia and, more recently, new long-acting amantadine formulations have been developed, with new indications to treat motor fluctuations. Amantadine has not been reported to cause dyskinesia and is rarely implicated in impulse control disorder.
Collapse
Affiliation(s)
- Sarah Marmol
- Department of Neurology, University of Miami Miller School of Medicine, 1150 NW 14th Street, Suite 609, Miami, FL, 33136, USA
| | - Matthew Feldman
- Department of Neurology, University of Miami Miller School of Medicine, 1150 NW 14th Street, Suite 609, Miami, FL, 33136, USA
| | - Carlos Singer
- Department of Neurology, University of Miami Miller School of Medicine, 1150 NW 14th Street, Suite 609, Miami, FL, 33136, USA
| | - Jason Margolesky
- Department of Neurology, University of Miami Miller School of Medicine, 1150 NW 14th Street, Suite 609, Miami, FL, 33136, USA.
| |
Collapse
|
17
|
Hutny M, Hofman J, Klimkowicz-Mrowiec A, Gorzkowska A. Current Knowledge on the Background, Pathophysiology and Treatment of Levodopa-Induced Dyskinesia-Literature Review. J Clin Med 2021; 10:jcm10194377. [PMID: 34640395 PMCID: PMC8509231 DOI: 10.3390/jcm10194377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/02/2021] [Accepted: 09/22/2021] [Indexed: 02/07/2023] Open
Abstract
Levodopa remains the primary drug for controlling motor symptoms in Parkinson’s disease through the whole course, but over time, complications develop in the form of dyskinesias, which gradually become more frequent and severe. These abnormal, involuntary, hyperkinetic movements are mainly characteristic of the ON phase and are triggered by excess exogenous levodopa. They may also occur during the OFF phase, or in both phases. Over the past 10 years, the issue of levodopa-induced dyskinesia has been the subject of research into both the substrate of this pathology and potential remedial strategies. The purpose of the present study was to review the results of recent research on the background and treatment of dyskinesia. To this end, databases were reviewed using a search strategy that included both relevant keywords related to the topic and appropriate filters to limit results to English language literature published since 2010. Based on the selected papers, the current state of knowledge on the morphological, functional, genetic and clinical features of levodopa-induced dyskinesia, as well as pharmacological, genetic treatment and other therapies such as deep brain stimulation, are described.
Collapse
Affiliation(s)
- Michał Hutny
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
- Correspondence:
| | - Jagoda Hofman
- Students’ Scientific Society, Department of Neurorehabilitation, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Aleksandra Klimkowicz-Mrowiec
- Department of Internal Medicine and Gerontology, Faculty of Medicine, Medical College, Jagiellonian University, 30-688 Kraków, Poland;
| | - Agnieszka Gorzkowska
- Department of Neurorehabilitation, Faculty of Medical Sciences, School of Medicine, Medical University of Silesia, 40-752 Katowice, Poland;
| |
Collapse
|
18
|
Fabbrini A, Guerra A. Pathophysiological Mechanisms and Experimental Pharmacotherapy for L-Dopa-Induced Dyskinesia. J Exp Pharmacol 2021; 13:469-485. [PMID: 33953618 PMCID: PMC8092630 DOI: 10.2147/jep.s265282] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/21/2022] Open
Abstract
L-dopa-induced dyskinesia (LID) is the most frequent motor complication associated with chronic L-dopa treatment in Parkinson’s disease (PD). Recent advances in the understanding of the pathophysiological mechanisms underlying LID suggest that abnormalities in multiple neurotransmitter systems, in addition to dopaminergic nigrostriatal denervation and altered dopamine release and reuptake dynamics at the synaptic level, are involved in LID development. Increased knowledge of neurobiological LID substrates has led to the development of several drug candidates to alleviate this motor complication. However, with the exception of amantadine, none of the pharmacological therapies tested in humans have demonstrated clinically relevant beneficial effects. Therefore, LID management is still one of the most challenging problems in the treatment of PD patients. In this review, we first describe the known pathophysiological mechanisms of LID. We then provide an updated report of experimental pharmacotherapies tested in clinical trials of PD patients and drugs currently under study to alleviate LID. Finally, we discuss available pharmacological LID treatment approaches and offer our opinion of possible issues to be clarified and future therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Fabbrini
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
19
|
Hauser RA, Walsh RR, Pahwa R, Chernick D, Formella AE. Amantadine ER (Gocovri ®) Significantly Increases ON Time Without Any Dyskinesia: Pooled Analyses From Pivotal Trials in Parkinson's Disease. Front Neurol 2021; 12:645706. [PMID: 33841311 PMCID: PMC8032973 DOI: 10.3389/fneur.2021.645706] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/22/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Clinical trials for antiparkinsonian drugs aimed at managing motor complications typically use patient diaries to divide levodopa-induced dyskinesias (LID) into "troublesome" and "non-troublesome" categories. Yet, given the choice, most patients would prefer to live without experiencing any dyskinesia. However, the concept of evaluating time spent ON without any dyskinesia as an outcome has never been tested. We conducted analyses of pooled Gocovri pivotal trial data in order to evaluate the extent to which Gocovri increased the time PD patients spent ON without dyskinesia (troublesome or non-troublesome), beyond its already identified improvement in reducing troublesome dyskinesia. Methods: Patients enrolled in phase 3 trials (EASE LID [NCT02136914] or EASE LID 3 [NCT02274766]) recorded time spent in the following PD diary states at baseline and Week 12 (endpoint): asleep, OFF, ON with troublesome dyskinesia, ON with non-troublesome dyskinesia, and ON without dyskinesia. Mixed model repeated measures analyses with estimated Cohen D effect sizes were performed on the modified intent to treat population to evaluate changes in time spent in these states. Results: Patients randomized to receive Gocovri showed an increase in ON time without dyskinesia and corresponding decreases in ON time with dyskinesia and OFF time vs. placebo. Treatment effects were statistically significant for Gocovri vs. placebo starting at Week 2 and were sustained until Week 12. On MMRM analysis at Week 12, patients in the Gocovri group showed an adjusted mean ± SE increase over placebo of 2.9 ± 0.6 h in ON time without dyskinesia (Cohen D effect size 0.79) and an adjusted mean ± SE decrease of -1.9 ± 0.6 h in ON time with dyskinesia (troublesome + non-troublesome) (Cohen D effect size 0.49), that included a -1.5 ± 0.4 h placebo-adjusted reduction in ON time with troublesome dyskinesia and a -0.6 ± 0.4 h reduction in ON time with non-troublesome dyskinesia. OFF time was reduced by -1.0 ± 0.3 h compared to placebo. Conclusions: Gocovri treatment more than doubled the daily time patients spent ON without dyskinesia. These results suggest that the Gocovri treatment effect was driven by a reduction in overall motor complications including ON time with both troublesome and non-troublesome dyskinesia as well as time spent OFF.
Collapse
Affiliation(s)
- Robert A Hauser
- Department of Neurology, University of South Florida, Tampa, FL, United States
| | - Ryan R Walsh
- Muhammad Ali Parkinson Center at Barrow Neurological Institute, Phoenix, AZ, United States
| | - Rajesh Pahwa
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | | | | |
Collapse
|
20
|
Isaacson SH, Lyons KE, Amjad F, Pahwa R. Development, Efficacy and Safety of Once-daily, Bedtime, Extended-release Amantadine (Gocovri®) to Treat Dyskinesia and OFF Time in Parkinson’s Disease. Neurology 2021. [DOI: 10.17925/usn.2021.17.1.36] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
|
21
|
AlShimemeri S, Fox SH, Visanji NP. Emerging drugs for the treatment of L-DOPA-induced dyskinesia: an update. Expert Opin Emerg Drugs 2020; 25:131-144. [DOI: 10.1080/14728214.2020.1763954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Sohaila AlShimemeri
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
- Division of Neurology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Susan H Fox
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| | - Naomi P Visanji
- Edmond J Safra Program in Parkinson Disease & Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, Toronto, ON, Canada
| |
Collapse
|