1
|
Pinheiro F, Pallarès I, Peccati F, Sánchez-Morales A, Varejão N, Bezerra F, Ortega-Alarcon D, Gonzalez D, Osorio M, Navarro S, Velázquez-Campoy A, Almeida MR, Reverter D, Busqué F, Alibés R, Sodupe M, Ventura S. Development of a Highly Potent Transthyretin Amyloidogenesis Inhibitor: Design, Synthesis, and Evaluation. J Med Chem 2022; 65:14673-14691. [PMID: 36306808 PMCID: PMC9661476 DOI: 10.1021/acs.jmedchem.2c01195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Transthyretin amyloidosis
(ATTR) is a group of fatal diseases described
by the misfolding and amyloid deposition of transthyretin (TTR). Discovering
small molecules that bind and stabilize the TTR tetramer, preventing
its dissociation and subsequent aggregation, is a therapeutic strategy
for these pathologies. Departing from the crystal structure of TTR
in complex with tolcapone, a potent binder in clinical trials for
ATTR, we combined rational design and molecular dynamics (MD) simulations
to generate a series of novel halogenated kinetic stabilizers. Among
them, M-23 displays one of the highest affinities for
TTR described so far. The TTR/M-23 crystal structure
confirmed the formation of unprecedented protein–ligand contacts,
as predicted by MD simulations, leading to an enhanced tetramer stability
both in vitro and in whole serum. We demonstrate
that MD-assisted design of TTR ligands constitutes a new avenue for
discovering molecules that, like M-23, hold the potential
to become highly potent drugs to treat ATTR.
Collapse
Affiliation(s)
- Francisca Pinheiro
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Irantzu Pallarès
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Francesca Peccati
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Adrià Sánchez-Morales
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Nathalia Varejão
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Filipa Bezerra
- Molecular Neurobiology Group, i3S−Instituto de Investigação e Inovação em Saúde, IBMC−Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS−Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - David Ortega-Alarcon
- Department of Biochemistry and Molecular & Cellular Biology, and Institute for Biocomputation eand Physics of Complex Systems (BIFI), Joint Unit GBsC-CSIC-BIFI, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Institute for Health Research, 50009 Zaragoza, Spain
- Biomedical Research Network Center in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain
| | - Danilo Gonzalez
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Marcelo Osorio
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Susanna Navarro
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Adrián Velázquez-Campoy
- Department of Biochemistry and Molecular & Cellular Biology, and Institute for Biocomputation eand Physics of Complex Systems (BIFI), Joint Unit GBsC-CSIC-BIFI, Universidad de Zaragoza, 50018 Zaragoza, Spain
- Aragon Institute for Health Research, 50009 Zaragoza, Spain
- Biomedical Research Network Center in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain
| | - Maria Rosário Almeida
- Molecular Neurobiology Group, i3S−Instituto de Investigação e Inovação em Saúde, IBMC−Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
- Departamento de Biologia Molecular, ICBAS−Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - David Reverter
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Félix Busqué
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Ramon Alibés
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Mariona Sodupe
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i Biomedicina and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona 08193, Spain
- ICREA, Passeig Lluis Companys 23, E-08010 Barcelona, Spain
| |
Collapse
|
2
|
Akhtar MJ, Yar MS, Grover G, Nath R. Neurological and psychiatric management using COMT inhibitors: A review. Bioorg Chem 2020; 94:103418. [DOI: 10.1016/j.bioorg.2019.103418] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/27/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022]
|
3
|
Pallesen J, Møllerud S, Frydenvang K, Pickering DS, Bornholdt J, Nielsen B, Pasini D, Han L, Marconi L, Kastrup JS, Johansen TN. N1-Substituted Quinoxaline-2,3-diones as Kainate Receptor Antagonists: X-ray Crystallography, Structure-Affinity Relationships, and in Vitro Pharmacology. ACS Chem Neurosci 2019; 10:1841-1853. [PMID: 30620174 DOI: 10.1021/acschemneuro.8b00726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Among the ionotropic glutamate receptors, the physiological role of kainate receptors is less well understood. Although ligands with selectivity toward the kainate receptor subtype GluK1 are available, tool compounds with selectivity at the remaining kainate receptor subtypes are sparse. Here, we have synthesized a series of quinoxaline-2,3-diones with substitutions in the N1-, 6-, and 7-position to investigate the structure-activity relationship (SAR) at GluK1-3 and GluK5. Pharmacological characterization at native and recombinant kainate and AMPA receptors revealed that compound 37 had a GluK3-binding affinity ( Ki) of 0.142 μM and 8-fold preference for GluK3 over GluK1. Despite lower binding affinity of 22 at GluK3 ( Ki = 2.91 μM), its preference for GluK3 over GluK1 and GluK2 was >30-fold. Compound 37 was crystallized with the GluK1 ligand-binding domain to understand the SAR. The X-ray structure showed that 37 stabilized the protein in an open conformation, consistent with an antagonist binding mode.
Collapse
Affiliation(s)
- Jakob Pallesen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Stine Møllerud
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Karla Frydenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Darryl S. Pickering
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jan Bornholdt
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birgitte Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Diletta Pasini
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Liwei Han
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Laura Marconi
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jette Sandholm Kastrup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Tommy N. Johansen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
4
|
Rakshit S, Lakshminarasimhan T, Guturi S, Kanagavel K, Kanusu UR, Niyogi AG, Sidar S, Luzung MR, Schmidt MA, Zheng B, Eastgate MD, Vaidyanathan R. Nitration Using Fuming HNO3 in Sulfolane: Synthesis of 6-Nitrovanillin in Flow Mode. Org Process Res Dev 2018. [DOI: 10.1021/acs.oprd.8b00020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Souvik Rakshit
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Thirumalai Lakshminarasimhan
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Sivakrishna Guturi
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Kishorekumar Kanagavel
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Umamaheswara Rao Kanusu
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Ankita G. Niyogi
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Somprabha Sidar
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| | - Michael R. Luzung
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Michael A. Schmidt
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Bin Zheng
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Martin D. Eastgate
- Chemical and Synthetic Development, Bristol-Myers Squibb, 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Rajappa Vaidyanathan
- Chemical Development and API Supply, Biocon Bristol-Myers Squibb Research and Development Center, Biocon Park, Jigani Link Road, Bommasandra IV, Bangalore 560099, India
| |
Collapse
|
5
|
Conroy T, Manohar M, Gong Y, Wilkinson SM, Webster M, Lieberman BP, Banister SD, Reekie TA, Mach RH, Rendina LM, Kassiou M. A systematic exploration of the effects of flexibility and basicity on sigma (σ) receptor binding in a series of substituted diamines. Org Biomol Chem 2016; 14:9388-9405. [PMID: 27714195 DOI: 10.1039/c6ob00615a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The sigma-1 receptor (S1R) has attracted a great deal of attention as a prospective drug target due to its involvement in numerous neurological disorders and, more recently, for its therapeutic potential in neuropathic pain. As there was no crystal structure of this membrane-bound protein reported until 2016, ligand generation was driven by pharmacophore refinements to the general model suggested by Glennon and co-workers. The generalised S1R pharmacophore comprises a central region where a basic amino group is preferred, flanked by two hydrophobic groups. Guided by this pharmacophore, S1R ligands containing piperazines, piperazinones, and ethylenediamines have been developed. In the current work, we systematically deconstructed the piperazine core of a prototypic piperazine S1R ligand (vide infra) developed in our laboratories. Although we did not improve the affinity at the S1R compared to the lead, we identified several features important for affinity and selectivity. These included at least one basic nitrogen atom, conformational flexibility and, for S1R, a secondary or tertiary amine group proximal to the anisole. Furthermore, S2R selectivity can be tailored with functional group modifications of the N-atom proximal to the anisole.
Collapse
Affiliation(s)
- Trent Conroy
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Madhura Manohar
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Yu Gong
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Shane M Wilkinson
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Michael Webster
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Brian P Lieberman
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Samuel D Banister
- Department of Radiation Oncology, Stanford University School of Medicine, CA 94305, USA
| | - Tristan A Reekie
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Robert H Mach
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Louis M Rendina
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Michael Kassiou
- School of Chemistry, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
6
|
Kiss LE, Soares-da-Silva P. Medicinal chemistry of catechol O-methyltransferase (COMT) inhibitors and their therapeutic utility. J Med Chem 2014; 57:8692-717. [PMID: 25080080 DOI: 10.1021/jm500572b] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Catechol O-methyltransferase (COMT) is the enzyme responsible for the O-methylation of endogenous neurotransmitters and of xenobiotic substances and hormones incorporating catecholic structures. COMT is a druggable biological target for the treatment of various central and peripheral nervous system disorders, including Parkinson's disease, depression, schizophrenia, and other dopamine deficiency-related diseases. The purpose of this perspective is fourfold: (i) to summarize the physiological role of COMT inhibitors in central and peripheral nervous system disorders; (ii) to provide the history and perspective of the medicinal chemistry behind the discovery and development of COMT inhibitors; (iii) to discuss how the physicochemical properties of recognized COMT inhibitors are understood to exert influence over their pharmacological properties; and (iv) to evaluate the clinical benefits of the most relevant COMT inhibitors.
Collapse
Affiliation(s)
- László E Kiss
- Department of Research & Development, BIAL - Portela & Ca, S.A. , À Avenida da Siderurgia Nacional, 4745-457 S. Mamede do Coronado, Portugal
| | | |
Collapse
|
7
|
Kamal A, Reddy CR, Vishnuvardhan M, Mahesh R, Lakshma Nayak V, Prabhakar S, Reddy CS. Synthesis and biological evaluation of cinnamido linked benzophenone hybrids as tubulin polymerization inhibitors and apoptosis inducing agents. Bioorg Med Chem Lett 2014; 24:2309-14. [DOI: 10.1016/j.bmcl.2014.03.076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/13/2014] [Accepted: 03/25/2014] [Indexed: 12/19/2022]
|
8
|
Jatana N, Sharma A, Latha N. Pharmacophore modeling and virtual screening studies to design potential COMT inhibitors as new leads. J Mol Graph Model 2012; 39:145-64. [PMID: 23280413 DOI: 10.1016/j.jmgm.2012.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/15/2012] [Accepted: 10/08/2012] [Indexed: 10/27/2022]
Abstract
Catechol-O-methyltransferase (COMT) catalyzes the methylation of catecholamines, including neurotransmitters like dopamine, epinephrine and norepinephrine, leading to their degradation. COMT has been a subject of study for its implications in numerous neurological disorders like Parkinson's disease (PD), schizophrenia, and depression. The COMT gene is associated with many allelic variants, the Val108Met polymorphism being the most clinically significant. Availability of crystal structure of both 108V and 108M forms of human soluble-COMT (S-COMT) facilitated us to use structure-based virtual screening approach to obtain new hits by screening a library of CNS permeable compounds from ZINC database. In this study, E-pharmacophore was also used to generate pharmacophore models based on a series of known COMT inhibitors. A five-point pharmacophore model consisting of one hydrogen-bond acceptor (A), two hydrogen bond donors (D), and two aromatic rings (R) was generated for both the polymorphic forms of COMT. These models were then used for filtering ZINC-CNS permeable library to obtain new hits. Physicochemical properties were also calculated for all the hits obtained from both the approaches for favorable ADME properties. These identified hits maybe of interest for further structural optimization and biological evaluation assays.
Collapse
Affiliation(s)
- Nidhi Jatana
- Bioinformatics Infrastructure Facility, Sri Venkateswara College (University of Delhi), Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | | | | |
Collapse
|
9
|
Liu QC, Guo TT, Fan Z, Li D, Li WH. First total synthesis of two new heterocyclic compounds: Bretschneiderazines A and B. CHINESE CHEM LETT 2011. [DOI: 10.1016/j.cclet.2010.11.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Schobert R, Biersack B, Dietrich A, Effenberger K, Knauer S, Mueller T. 4-(3-Halo/amino-4,5-dimethoxyphenyl)-5-aryloxazoles and -N-methylimidazoles That Are Cytotoxic against Combretastatin A Resistant Tumor Cells and Vascular Disrupting in a Cisplatin Resistant Germ Cell Tumor Model. J Med Chem 2010; 53:6595-602. [DOI: 10.1021/jm100345r] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Rainer Schobert
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, D-95440 Bayreuth, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, D-95440 Bayreuth, Germany
| | - Andrea Dietrich
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle, Germany
| | - Katharina Effenberger
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, D-95440 Bayreuth, Germany
| | - Sebastian Knauer
- Organic Chemistry Laboratory, University Bayreuth, Universitätsstrasse 30, D-95440 Bayreuth, Germany
| | - Thomas Mueller
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle, Germany
| |
Collapse
|
11
|
Aparicio F, Vicente F, Sánchez L. Amplification of chirality in N,N′-1,2-ethanediylbisbenzamides: from planar sheets to twisted ribbons. Chem Commun (Camb) 2010; 46:8356-8. [DOI: 10.1039/c0cc02726j] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
12
|
Nissinen E, Männistö PT. Biochemistry and Pharmacology of Catechol-O-Methyltransferase Inhibitors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 95:73-118. [DOI: 10.1016/b978-0-12-381326-8.00005-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Learmonth DA, Kiss LE, Soares-da-Silva P. The Chemistry of Catechol-O-Methyltransferase Inhibitors. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2010; 95:119-62. [DOI: 10.1016/b978-0-12-381326-8.00006-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14
|
Bonifácio MJ, Palma PN, Almeida L, Soares‐da‐Silva P. Catechol-O-methyltransferase and its inhibitors in Parkinson's disease. CNS DRUG REVIEWS 2007; 13:352-79. [PMID: 17894650 PMCID: PMC6494163 DOI: 10.1111/j.1527-3458.2007.00020.x] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is a neurological disorder characterized by the degeneration of dopaminergic neurons, with consequent reduction in striatal dopamine levels leading to characteristic motor symptoms. The most effective treatment for this disease continues to be the dopamine replacement therapy with levodopa together with an inhibitor of aromatic amino acid decarboxylase (AADC). The efficacy of this therapy, however, decreases with time and most patients develop fluctuating responses and dyskinesias. The last decade showed that the use of catechol-O-methyltransferase inhibitors as adjuvants to the levodopa/AADC inhibitor therapy, significantly improves the clinical benefits of this therapy. The purpose of this article is to review the current knowledge on the enzyme catechol-O-methyltransferase (COMT) and the role of COMT inhibitors in PD as a new therapeutic approach to PD involving conversion of levodopa to dopamine at the target region in the brain and facilitation of the continuous action of this amine at the receptor sites. A historical overview of the discovery and development of COMT inhibitors is presented with a special emphasis on nebicapone, presently under clinical development, as well as entacapone and tolcapone, which are already approved as adjuncts in the therapy of PD. This article reviews human pharmacokinetic and pharmacodynamic properties of these drugs as well as their clinical efficacy and safety.
Collapse
Affiliation(s)
- Maria João Bonifácio
- Department of Research and Development, BIAL (Portela & Co S.A.), S Mamede do Coronado, Portugal
| | - P. Nuno Palma
- Department of Research and Development, BIAL (Portela & Co S.A.), S Mamede do Coronado, Portugal
| | - Luís Almeida
- Department of Research and Development, BIAL (Portela & Co S.A.), S Mamede do Coronado, Portugal
- Institute of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal
| | - Patrício Soares‐da‐Silva
- Department of Research and Development, BIAL (Portela & Co S.A.), S Mamede do Coronado, Portugal
- Institute of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal
| |
Collapse
|