1
|
Lukoyanov AA, Aksenova SA, Tabolin AA, Sukhorukov AY. 3-Halo-5,6-dihydro-4 H-1,2-oxazine N-oxides as synthetic equivalents of unsaturated nitrile oxides in the [3 + 2]-cycloaddition with arynes: synthesis of substituted 3-vinyl-1,2-benzisoxazoles. Org Biomol Chem 2024; 22:3615-3621. [PMID: 38634451 DOI: 10.1039/d4ob00391h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
The reaction of 3-halo-5,6-dihydro-4H-1,2-oxazine N-oxides with arynes was studied. Arynes were generated from o-silylaryl triflates and underwent consecutive [3 + 2]-cycloaddition/[4 + 2]-cycloreversion with N-oxides leading to substituted 3-vinyl-benzisoxazoles in high yields. In the presented sequence, 1,2-oxazine N-oxides act as surrogates of rarely employed unsaturated nitrile oxides. A broad substrate scope was demonstrated. The influence of the substitution pattern of an aryne on the reaction outcome was determined. In the presence of bulky substituents, polycyclic 4,4a-dihydro-3H-benzofuro[3,2-c][1,2]oxazines were selectively formed. Mechanistic schemes for the observed reaction pathways were proposed. The synthetic utility of the products was demonstrated by their follow-up modifications.
Collapse
Affiliation(s)
- Alexander A Lukoyanov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, Moscow, 119991, Russian Federation.
| | - Svetlana A Aksenova
- A. N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilov str. 28, Moscow, 119334, Russian Federation
| | - Andrey A Tabolin
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, Moscow, 119991, Russian Federation.
| | - Alexey Yu Sukhorukov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prosp. 47, Moscow, 119991, Russian Federation.
| |
Collapse
|
2
|
Aboulatta L, Haidar L, Abou-Setta A, Askin N, Rabbani R, Lavu A, Peymani P, Zarychanski R, Eltonsy S. Efficacy and Safety of MAO-B Inhibitors Safinamide and Zonisamide in Parkinson's Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. CNS Drugs 2023; 37:941-956. [PMID: 37973769 DOI: 10.1007/s40263-023-01048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND OBJECTIVE In Parkinson's disease, safinamide and zonisamide are novel monoamine oxidase-B inhibitors with a dual mechanism of action involving the inhibition of sodium and calcium channels and the subsequent release of glutamate. The aim of this systematic review and meta-analysis was to examine the efficacy and safety of both drugs compared with placebo on motor symptoms, cognitive function, and quality of life in patients with Parkinson's disease. METHODS We searched MEDLINE, EMBASE, Cochrane Central, Scopus, PsycINFO, and trials registries up to March 2023 for randomized controlled trials of adults with Parkinson's disease administered either safinamide or zonisamide and published in English. We excluded single-arm trials or if neither the efficacy nor safety outcomes of interest were reported. Primary outcomes were the change from baseline in Unified Parkinson's Disease Rating Scale section III (UPDRS-III) and serious adverse events. Secondary outcomes included a change from baseline in OFF-time, Parkinson's Disease Questionnaire 39 to evaluate quality of life, and Mini-Mental State Examination for cognitive function assessment. The meta-analysis was conducted using Review Manager 5.4.1. Random-effect models were used to calculate the pooled mean differences (MDs) and risk ratios with 95% confidence intervals (CIs). Subgroup analyses by medication, doses, Parkinson's disease stage, and risk of bias were conducted. We assessed the risk of bias using the Cochrane's risk of bias tool. Sensitivity analysis was conducted, and publication bias were evaluated. This meta-analysis was not externally funded, and the protocol is available on the Open Science Framework Registration ( https://doi.org/10.17605/OSF.IO/AMNP5 ). RESULTS Of 3570 screened citations, 16 trials met inclusion criteria (4314 patients with Parkinson's disease). Ten safinamide trials were conducted in several countries. Six zonisamide trials were included, five of which were conducted in Japan and one in India. UPDRS Part III scores were significantly lower with both monoamine oxidase-B inhibitors than with placebo (MD = - 2.18; 95% CI - 2.88 to - 1.49; I 2 =63%; n = 14 studies). A subgroup analysis showed a significant improvement in UPDRS-III in safinamide (MD = - 2.10; 95% CI - 3.09 to - 1.11; I2 = 71%; n = 8 studies) and zonisamide (MD = - 2.31; 95% CI - 3.35 to - 1.27; I2 = 52%; n = 6 studies) compared with placebo. Monoamine oxidase-B inhibitors significantly decreased OFF-time compared with placebo. No significant differences in cognitive function (Mini-Mental State Examination), whereas an improvement in quality of life (Parkinson's Disease Questionnaire 39 scores) was observed. There was no significant difference in incidence rates of serious adverse events among all examined doses of zonisamide and safinamide compared with placebo. Two trials were reported as a high risk of bias and sensitivity analyses confirmed the primary analysis results. CONCLUSIONS Evidence suggests that novel monoamine oxidase-B inhibitors not only improve motor symptoms but also enhance patients' quality of life. The meta-analysis showed that both medications have a similar safety profile to placebo with regard to serious adverse events. The overall findings emphasize the effectiveness of safinamide and zonisamide in the treatment of Parkinson's disease as adjunct therapy. Further long-term studies examining the impact of these medications on motor and non-motor symptoms are necessary.
Collapse
Affiliation(s)
- Laila Aboulatta
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Lara Haidar
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ahmed Abou-Setta
- George & Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
- Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Nicole Askin
- WRHA Virtual Library, University of Manitoba, Winnipeg, MB, Canada
| | - Rasheda Rabbani
- George & Fay Yee Center for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
| | - Alekhya Lavu
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Payam Peymani
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada
| | - Ryan Zarychanski
- Community Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Cancer Care Manitoba, Winnipeg, MB, Canada
| | - Sherif Eltonsy
- College of Pharmacy, University of Manitoba, Winnipeg, MB, R3E 0T5, Canada.
| |
Collapse
|
3
|
Chen X, Feng Y, Quinn RJ, Pountney DL, Richardson DR, Mellick GD, Ma L. Potassium Channels in Parkinson's Disease: Potential Roles in Its Pathogenesis and Innovative Molecular Targets for Treatment. Pharmacol Rev 2023; 75:758-788. [PMID: 36918260 DOI: 10.1124/pharmrev.122.000743] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 01/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) region of the midbrain. The loss of neurons results in a subsequent reduction of dopamine in the striatum, which underlies the core motor symptoms of PD. To date, there are no effective treatments to stop, slow, or reverse the pathologic progression of dopaminergic neurodegeneration. This unfortunate predicament is because of the current early stages in understanding the biologic targets and pathways involved in PD pathogenesis. Ion channels have become emerging targets for new therapeutic development for PD due to their essential roles in neuronal function and neuroinflammation. Potassium channels are the most prominent ion channel family and have been shown to be critically important in PD pathology because of their roles in modulating neuronal excitability, neurotransmitter release, synaptic transmission, and neuroinflammation. In this review, members of the subfamilies of voltage-gated K+ channels, inward rectifying K+ channels, and Ca2+-activated K+ channels are described. Evidence of the role of these channels in PD etiology is discussed together with the latest views on related pathologic mechanisms and their potential as biologic targets for developing neuroprotective drugs for PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is the second most common neurodegenerative disorder, featuring progressive degeneration of dopaminergic neurons in the midbrain. It is a multifactorial disease involving multiple risk factors and complex pathobiological mechanisms. Mounting evidence suggests that ion channels play vital roles in the pathogenesis and progression of PD by regulating neuronal excitability and immune cell function. Therefore, they have become "hot" biological targets for PD, as demonstrated by multiple clinical trials of drug candidates targeting ion channels for PD therapy.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Yunjiang Feng
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Ronald J Quinn
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Dean L Pountney
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Des R Richardson
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - George D Mellick
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| | - Linlin Ma
- School of Environment and Science (Y.F., D.R.R., G.D.M., L.M.) and Centre for Cancer Cell Biology and Drug Discovery (D.R.R.), Griffith Institute for Drug Discovery (X.C., Y.F., R.J.Q., D.R.R., G.D.M., L.M.), Griffith University, Nathan, Brisbane, Queensland, Australia; and School of Pharmacy and Medical Science, Griffith University, Gold Coast, Queenslandstate, Australia (D.L.P.)
| |
Collapse
|
4
|
Fletcher EJR, Kaminski T, Williams G, Duty S. Drug repurposing strategies of relevance for Parkinson's disease. Pharmacol Res Perspect 2021; 9:e00841. [PMID: 34309236 PMCID: PMC8311732 DOI: 10.1002/prp2.841] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/29/2021] [Indexed: 01/01/2023] Open
Abstract
Parkinson's disease is a highly disabling, progressive neurodegenerative disease that manifests as a mix of motor and non-motor signs. Although we are equipped with some symptomatic treatments, especially for the motor signs of the disease, there are still no established disease-modifying drugs so the disease progresses unchecked. Standard drug discovery programs for disease-modifying therapies have provided key insights into the pathogenesis of Parkinson's disease but, of the many positive candidates identified in pre-clinical studies, none has yet translated into a successful clinically efficacious drug. Given the huge cost of drug discovery programs, it is not surprising that much attention has turned toward repurposing strategies. The trialing of an established therapeutic has the advantage of bypassing the need for preclinical safety testing and formulation optimization, thereby cutting both time and costs involved in getting a treatment to the clinic. Additional reduced failure rates for repurposed drugs are also a potential bonus. Many different strategies for drug repurposing are open to researchers in the Parkinson's disease field. Some of these have already proven effective in identifying suitable drugs for clinical trials, lending support to such approaches. In this review, we present a summary of the different strategies for drug repurposing, from large-scale epidemiological correlation analysis through to single-gene transcriptional approaches. We provide examples of past or ongoing studies adopting each strategy, where these exist. For strategies that have yet to be applied to Parkinson's disease, their utility is illustrated using examples taken from other disorders.
Collapse
Affiliation(s)
- Edward J. R. Fletcher
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Thomas Kaminski
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Gareth Williams
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| | - Susan Duty
- King’s College LondonInstitute of Psychiatry, Psychology & NeuroscienceWolfson Centre for Age‐Related DiseasesLondonUK
| |
Collapse
|
5
|
Li C, Xue L, Liu Y, Yang Z, Chi S, Xie A. Zonisamide for the Treatment of Parkinson Disease: A Current Update. Front Neurosci 2020; 14:574652. [PMID: 33408605 PMCID: PMC7779619 DOI: 10.3389/fnins.2020.574652] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022] Open
Abstract
Zonisamide has been used as an add-on treatment in order to overcome the deficiencies of the general therapies currently used to resolve the motor complications and non-motor symptoms of Parkinson disease. Various trials have been designed to investigate the mechanism of action and treatment effects of zonisamide in this condition. Most clinical trials of zonisamide in Parkinson disease were from Japan. The vast majority of studies used changes in the Unified Parkinson’s Disease Rating Scale (UPDRS) scores and daily “OFF” time as primary endpoints. Based on adequate randomized controlled trials, zonisamide is considered a safe and efficacious add-on treatment in Parkinson disease. The most convincing proof is available for a dosage of 25–50 mg, which was shown to lead to a significant reduction in the UPDRS III score and daily “OFF” time, without increasing disabling dyskinesia. Furthermore, zonisamide may play a beneficial role in improving non-motor symptoms in PD, including impulsive–compulsive disorder, rapid eye movement sleep behavior disorder, and dementia. Among the various mechanisms reported, inhibition of monoamine oxidase-B, blocking of T-type calcium channels, modulation of the levodopa–dopamine metabolism, modulation of receptor expression, and neuroprotection are the most often cited. The mechanisms underlying neuroprotection, including modulation of dopamine turnover, induction of neurotrophic factor expression, inhibition of oxidative stress and apoptosis, inhibition of neuroinflammation, modulation of synaptic transmission, and modulation of gene expression, have been most extensively studied. This review focuses on structure, pharmacokinetics, mechanisms, therapeutic effectiveness, and safety and tolerability of zonisamide in patients with Parkinson disease.
Collapse
Affiliation(s)
- Chengqian Li
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Xue
- Department of Medical Record, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yumei Liu
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhengjie Yang
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Tripathi AC, Upadhyay S, Paliwal S, Saraf SK. Privileged scaffolds as MAO inhibitors: Retrospect and prospects. Eur J Med Chem 2018; 145:445-497. [PMID: 29335210 DOI: 10.1016/j.ejmech.2018.01.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/01/2017] [Accepted: 01/01/2018] [Indexed: 12/24/2022]
Abstract
This review aims to be a comprehensive, authoritative, critical, and readable review of general interest to the medicinal chemistry community because it focuses on the pharmacological, chemical, structural and computational aspects of diverse chemical categories as monoamine oxidase inhibitors (MAOIs). Monoamine oxidases (MAOs), namely MAO-A and MAO-B represent an enormously valuable class of neuronal enzymes embodying neurobiological origin and functions, serving as potential therapeutic target in neuronal pharmacotherapy, and hence we have coined the term "Neurozymes" which is being introduced for the first time ever. Nowadays, therapeutic attention on MAOIs engrosses two imperative categories; MAO-A inhibitors, in certain mental disorders such as depression and anxiety, and MAO-B inhibitors, in neurodegenerative disorders like Alzheimer's disease (AD) and Parkinson's disease (PD). The use of MAOIs declined due to some potential side effects, food and drug interactions, and introduction of other classes of drugs. However, curiosity in MAOIs is reviving and the recent developments of new generation of highly selective and reversible MAOIs, have renewed the therapeutic prospective of these compounds. The initial section of the review emphasizes on the detailed classification, structural and binding characteristics, therapeutic potential, current status and future challenges of the privileged pharmacophores. However, the chemical prospective of privileged scaffolds such as; aliphatic and aromatic amines, amides, hydrazines, azoles, diazoles, tetrazoles, indoles, azines, diazines, xanthenes, tricyclics, benzopyrones, and more interestingly natural products, along with their conclusive SARs have been discussed in the later segment of review. The last segment of the article encompasses some patents granted in the field of MAOIs, in a simplistic way.
Collapse
Affiliation(s)
- Avinash C Tripathi
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Savita Upadhyay
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Sarvesh Paliwal
- Pharmacy Department, Banasthali Vidyapith, Banasthali, Tonk 304022, Rajasthan, India
| | - Shailendra K Saraf
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India.
| |
Collapse
|
7
|
Bentea E, Van Liefferinge J, Verbruggen L, Martens K, Kobayashi S, Deneyer L, Demuyser T, Albertini G, Maes K, Sato H, Smolders I, Lewerenz J, Massie A. Zonisamide attenuates lactacystin-induced parkinsonism in mice without affecting system x c<sup/>. Exp Neurol 2016; 290:15-28. [PMID: 28024798 DOI: 10.1016/j.expneurol.2016.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 11/29/2022]
Abstract
Zonisamide (ZNS), an anticonvulsant drug exhibiting symptomatic effects in Parkinson's disease (PD), was recently reported to exert neuroprotection in rodent models. One of the proposed neuroprotective mechanisms involves increased protein expression of xCT, the specific subunit of the cystine/glutamate antiporter system xc-, inducing glutathione (GSH) synthesis. Here, we investigated the outcome of ZNS treatment in a mouse model of PD based on intranigral proteasome inhibition, and whether the observed effects would be mediated by system xc-. The proteasome inhibitor lactacystin (LAC) was administered intranigrally to male C57BL/6J mice receiving repeated intraperitoneal injections of either ZNS 30mgkg-1 or vehicle. Drug administration was initiated three days prior to stereotaxic LAC injection and was maintained until six days post-surgery. One week after lesion, mice were behaviorally assessed and investigated in terms of nigrostriatal neurodegeneration and molecular changes at the level of the basal ganglia, including expression levels of xCT. ZNS reduced the loss of nigral dopaminergic neurons following LAC injection and the degree of sensorimotor impairment. ZNS failed, however, to modulate xCT expression in basal ganglia of lesioned mice. In a separate set of experiments, the impact of ZNS treatment on system xc- was investigated in control conditions in vivo as well as in vitro. Similarly, ZNS did not influence xCT or glutathione levels in naive male C57BL/6J mice, nor did it alter system xc- activity or glutathione content in vitro. Taken together, these results demonstrate that ZNS treatment provides neuroprotection and behavioral improvement in a PD mouse model based on proteasome inhibition via system xc- independent mechanisms.
Collapse
Affiliation(s)
- Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lise Verbruggen
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katleen Martens
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sho Kobayashi
- Department of Food and Applied Life Sciences, Yamagata University, Yamagata, Japan
| | - Lauren Deneyer
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Demuyser
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katrien Maes
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hideyo Sato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology, Niigata University, Niigata, Japan
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
|
9
|
Abdel-Salam OME. Drug therapy for Parkinson’s disease: An update. World J Pharmacol 2015; 4:117. [DOI: 10.5497/wjp.v4.i1.117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Revised: 01/26/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023] Open
|
10
|
Binda C, Aldeco M, Mattevi A, Edmondson DE. Interactions of monoamine oxidases with the antiepileptic drug zonisamide: specificity of inhibition and structure of the human monoamine oxidase B complex. J Med Chem 2010; 54:909-12. [PMID: 21175212 DOI: 10.1021/jm101359c] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The binding of zonisamide to purified, recombinant monoamine oxidases (MAOs) has been investigated. It is a competitive inhibitor of human MAO B (K(i) = 3.1 ± 0.3 μM), of rat MAO B (K(i) = 2.9 ± 0.5 μM), and of zebrafish MAO (K(i) = 30.8 ± 5.3 μM). No inhibition is observed with purified human or rat MAO A. The 1.8 Å structure of the MAO B complex demonstrates that it binds within the substrate cavity.
Collapse
Affiliation(s)
- Claudia Binda
- Department Genetics and Microbiology, University of Pavia, via Ferrata 1, Pavia 27100, Italy
| | | | | | | |
Collapse
|
11
|
Yang LP, Perry CM. Zonisamide in Parkinsonʼs Disease†. Drugs Aging 2010; 27:73-5. [DOI: 10.2165/11203950-000000000-00000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
12
|
Verdier MC, Bentué-Ferrer D, Tribut O. Suivi thérapeutique pharmacologique du zonisamide. Therapie 2010; 65:29-34. [DOI: 10.2515/therapie/2009062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 12/01/2009] [Accepted: 12/01/2009] [Indexed: 11/20/2022]
|