51
|
Design, synthesis and biological evaluation of some novel diastereoselective β-lactams bearing 2-mercaptobenzothiazole and benzoquinoline. Med Chem Res 2019. [DOI: 10.1007/s00044-018-02287-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
52
|
Proschak E, Stark H, Merk D. Polypharmacology by Design: A Medicinal Chemist's Perspective on Multitargeting Compounds. J Med Chem 2018; 62:420-444. [PMID: 30035545 DOI: 10.1021/acs.jmedchem.8b00760] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multitargeting compounds comprising activity on more than a single biological target have gained remarkable relevance in drug discovery owing to the complexity of multifactorial diseases such as cancer, inflammation, or the metabolic syndrome. Polypharmacological drug profiles can produce additive or synergistic effects while reducing side effects and significantly contribute to the high therapeutic success of indispensable drugs such as aspirin. While their identification has long been the result of serendipity, medicinal chemistry now tends to design polypharmacology. Modern in vitro pharmacological methods and chemical probes allow a systematic search for rational target combinations and recent innovations in computational technologies, crystallography, or fragment-based design equip multitarget compound development with valuable tools. In this Perspective, we analyze the relevance of multiple ligands in drug discovery and the versatile toolbox to design polypharmacology. We conclude that despite some characteristic challenges remaining unresolved, designed polypharmacology holds enormous potential to secure future therapeutic innovation.
Collapse
Affiliation(s)
- Ewgenij Proschak
- Institute of Pharmaceutical Chemistry , Goethe University Frankfurt , Max-von-Laue-Strasse 9 , D-60438 Frankfurt , Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry , Heinrich Heine University Düsseldorf , Universitaetsstrasse 1 , D-40225 , Duesseldorf , Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry , Goethe University Frankfurt , Max-von-Laue-Strasse 9 , D-60438 Frankfurt , Germany.,Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences , Swiss Federal Institute of Technology (ETH) Zürich , Vladimir-Prelog-Weg 4 , CH-8093 Zürich , Switzerland
| |
Collapse
|
53
|
Potter BVL. SULFATION PATHWAYS: Steroid sulphatase inhibition via aryl sulphamates: clinical progress, mechanism and future prospects. J Mol Endocrinol 2018; 61:T233-T252. [PMID: 29618488 DOI: 10.1530/jme-18-0045] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022]
Abstract
Steroid sulphatase is an emerging drug target for the endocrine therapy of hormone-dependent diseases, catalysing oestrogen sulphate hydrolysis to oestrogen. Drug discovery, developing the core aryl O-sulphamate pharmacophore, has led to steroidal and non-steroidal drugs entering numerous clinical trials, with promising results in oncology and women's health. Steroidal oestrogen sulphamate derivatives were the first irreversible active-site-directed inhibitors and one was developed clinically as an oral oestradiol pro-drug and for endometriosis applications. This review summarizes work leading to the therapeutic concept of sulphatase inhibition, clinical trials executed to date and new insights into the mechanism of inhibition of steroid sulphatase. To date, the non-steroidal sulphatase inhibitor Irosustat has been evaluated clinically in breast cancer, alone and in combination, in endometrial cancer and in prostate cancer. The versatile core pharmacophore both imbues attractive pharmaceutical properties and functions via three distinct mechanisms of action, as a pro-drug, an enzyme active-site-modifying motif, likely through direct sulphamoyl group transfer, and as a structural component augmenting activity, for example by enhancing interactions at the colchicine binding site of tubulin. Preliminary new structural data on the Pseudomonas aeruginosa arylsulphatase enzyme suggest two possible sulphamate-based adducts with the active site formylglycine as candidates for the inhibition end product via sulphamoyl or sulphonylamine transfer, and a speculative choice is suggested. The clinical status of sulphatase inhibition is surveyed and how it might develop in the future. Also discussed are dual-targeting approaches, development of 2-substituted steroidal sulphamates and non-steroidal derivatives as multi-targeting agents for hormone-independent tumours, with other emerging directions.
Collapse
Affiliation(s)
- Barry V L Potter
- Medicinal Chemistry & Drug DiscoveryDepartment of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
54
|
Nandha B, Ramareddy SA, Kuntal H. Synthesis of substituted fluorobenzimidazoles as inhibitors of 5-lipoxygenase and soluble epoxide hydrolase for anti-inflammatory activity. Arch Pharm (Weinheim) 2018; 351:e1800030. [PMID: 29732612 DOI: 10.1002/ardp.201800030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 11/08/2022]
Abstract
A new series of 4-((5-fluoro-6-(substituted)-1H-benzo[d]imidazol-2-ylthio)methyl)-benzoic acids 4a-o and 2-(5-fluoro-6-(substituted)-1H-benzo[d]imidazol-2-ylthio)-2-methylpropanoic acids 8a-e were synthesized, and their inhibitory potencies against soluble epoxide hydrolase (sEH) and 5-lipoxygenase (5-LOX) were investigated. These molecules were designed based on the combination of 5-LOX and sEH pharmacophores, resulting in hybrid analogs with potent sEH and 5-LOX inhibitory activity. Compound 4g showed remarkable activity with IC50 values of less than 1 μM (0.9 μM) against 5-LOX, while compound 4k displayed promising activity against sEH with IC50 ≤ 1 μM (0.7 μM). These compounds were evaluated for their in vivo potential using the carrageenan-induced rat paw edema assay. Based on the obtained results, the structure-activity relationship was established and a correlation between the activities was observed. Compounds 4f, 4g, 4k, 4n, and 8e showed potent anti-inflammatory activity and significant inhibition of edema (64.13, 67.39, 66.30, 65.21, and 58.69%, respectively) at a dose of 100 mg/kg, comparable to the standard drug ibuprofen (70.65%) at 3 h.
Collapse
Affiliation(s)
- B Nandha
- Department of Pharmaceutical Chemistry, Vivekananda College of Pharmacy, Rajiv Gandhi University of Health Sciences, Bangalore, India
| | - Sureshbabu A Ramareddy
- Department of Pharmaceutical Chemistry, KLE University's College of Pharmacy, Bangalore, India
| | - Hazra Kuntal
- Department of Pharmaceutical Chemistry, Bharat Technology, Howrah, Banitabla, India
| |
Collapse
|
55
|
Galdeano C, Coquelle N, Cieslikiewicz-Bouet M, Bartolini M, Pérez B, Clos MV, Silman I, Jean L, Colletier JP, Renard PY, Muñoz-Torrero D. Increasing Polarity in Tacrine and Huprine Derivatives: Potent Anticholinesterase Agents for the Treatment of Myasthenia Gravis. Molecules 2018. [PMID: 29534488 PMCID: PMC6017698 DOI: 10.3390/molecules23030634] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Symptomatic treatment of myasthenia gravis is based on the use of peripherally-acting acetylcholinesterase (AChE) inhibitors that, in some cases, must be discontinued due to the occurrence of a number of side-effects. Thus, new AChE inhibitors are being developed and investigated for their potential use against this disease. Here, we have explored two alternative approaches to get access to peripherally-acting AChE inhibitors as new agents against myasthenia gravis, by structural modification of the brain permeable anti-Alzheimer AChE inhibitors tacrine, 6-chlorotacrine, and huprine Y. Both quaternization upon methylation of the quinoline nitrogen atom, and tethering of a triazole ring, with, in some cases, the additional incorporation of a polyphenol-like moiety, result in more polar compounds with higher inhibitory activity against human AChE (up to 190-fold) and butyrylcholinesterase (up to 40-fold) than pyridostigmine, the standard drug for symptomatic treatment of myasthenia gravis. The novel compounds are furthermore devoid of brain permeability, thereby emerging as interesting leads against myasthenia gravis.
Collapse
Affiliation(s)
- Carles Galdeano
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain.
| | - Nicolas Coquelle
- Institut de Biologie Structurale, Université Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'Énergie Atomique (CEA) (UMR 5075), F-38054 Grenoble, France.
- Large-Scale Structures Group, Institut Laue-Langevin, 71 Avenue des Martyrs, 38000 Grenoble, France.
| | - Monika Cieslikiewicz-Bouet
- Laboratory COBRA (UMR 6014), Normandie Université, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, 76000 Rouen, France.
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy.
| | - Belén Pérez
- Department of Pharmacology, Therapeutics and Toxicology, Neuroscience Institute, Autonomous University of Barcelona, E-08193 Barcelona, Spain.
| | - M Victòria Clos
- Department of Pharmacology, Therapeutics and Toxicology, Neuroscience Institute, Autonomous University of Barcelona, E-08193 Barcelona, Spain.
| | - Israel Silman
- Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Ludovic Jean
- Laboratory COBRA (UMR 6014), Normandie Université, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, 76000 Rouen, France.
| | - Jacques-Philippe Colletier
- Institut de Biologie Structurale, Université Grenoble Alpes, Centre National de la Recherche Scientifique (CNRS)-Commissariat à l'Énergie Atomique (CEA) (UMR 5075), F-38054 Grenoble, France.
| | - Pierre-Yves Renard
- Laboratory COBRA (UMR 6014), Normandie Université, UNIROUEN, Institut National des Sciences Appliquées (INSA) Rouen, CNRS, 76000 Rouen, France.
| | - Diego Muñoz-Torrero
- Laboratory of Pharmaceutical Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, and Institute of Biomedicine (IBUB), University of Barcelona, Av. Joan XXIII 27-31, E-08028 Barcelona, Spain.
| |
Collapse
|
56
|
Ramsay RR, Popovic-Nikolic MR, Nikolic K, Uliassi E, Bolognesi ML. A perspective on multi-target drug discovery and design for complex diseases. Clin Transl Med 2018; 7:3. [PMID: 29340951 PMCID: PMC5770353 DOI: 10.1186/s40169-017-0181-2] [Citation(s) in RCA: 417] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/30/2017] [Indexed: 12/11/2022] Open
Abstract
Diseases of infection, of neurodegeneration (such as Alzheimer’s and Parkinson’s diseases), and of malignancy (cancers) have complex and varied causative factors. Modern drug discovery has the power to identify potential modulators for multiple targets from millions of compounds. Computational approaches allow the determination of the association of each compound with its target before chemical synthesis and biological testing is done. These approaches depend on the prior identification of clinically and biologically validated targets. This Perspective will focus on the molecular and computational approaches that underpin drug design by medicinal chemists to promote understanding and collaboration with clinical scientists.
Collapse
Affiliation(s)
- Rona R Ramsay
- Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, KY16 9ST, UK.
| | - Marija R Popovic-Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000, Belgrade, Serbia
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000, Belgrade, Serbia
| | - Elisa Uliassi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Bologna University, Via Belmeloro 6, 40126, Bologna, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-Bologna University, Via Belmeloro 6, 40126, Bologna, Italy
| |
Collapse
|
57
|
Catto M, Trisciuzzi D, Alberga D, Mangiatordi GF, Nicolotti O. Multitarget Drug Design for Neurodegenerative Diseases. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/7653_2018_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
58
|
Kumar A, Tiwari A, Sharma A. Changing Paradigm from one Target one Ligand Towards Multi-target Directed Ligand Design for Key Drug Targets of Alzheimer Disease: An Important Role of In Silico Methods in Multi-target Directed Ligands Design. Curr Neuropharmacol 2018; 16:726-739. [PMID: 29542413 PMCID: PMC6080096 DOI: 10.2174/1570159x16666180315141643] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 04/01/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
Alzheimer disease (AD) is now considered as a multifactorial neurodegenerative disorder and rapidly increasing to an alarming situation and causing higher death rate. One target one ligand hypothesis does not provide complete solution of AD due to multifactorial nature of the disease and one target one drug fails to provide better treatment against AD. Moreover, currently available treatments are limited and most of the upcoming treatments under clinical trials are based on modulating single target. So, the current AD drug discovery research is shifting towards a new approach for a better solution that simultaneously modulates more than one targets in the neurodegenerative cascade. This can be achieved by network pharmacology, multi-modal therapies, multifaceted, and/or the more recently proposed term "multi-targeted designed drugs". Drug discovery project is a tedious, costly and long-term project. Moreover, multi-target AD drug discovery added extra challenges such as the good binding affinity of ligands for multiple targets, optimal ADME/T properties, no/less off-target side effect and crossing of the blood-brain barrier. These hurdles may be addressed by insilico methods for an efficient solution in less time and cost as computational methods successfully applied to single target drug discovery project. Here, we are summarizing some of the most prominent and computationally explored single targets against AD and further, we discussed a successful example of dual or multiple inhibitors for same targets. Moreover, we focused on ligand and structure-based computational approach to design MTDL against AD. However, it is not an easy task to balance dual activity in a single molecule but computational approach such as virtual screening docking, QSAR, simulation and free energy is useful in future MTDLs drug discovery alone or in combination with a fragment-based method. However, rational and logical implementations of computational drug designing methods are capable of assisting AD drug discovery and play an important role in optimizing multi-target drug discovery.
Collapse
Affiliation(s)
- Akhil Kumar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| | - Ashish Tiwari
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow-226015, (U.P.), India
| |
Collapse
|
59
|
Chen H, Bauer U, Engkvist O. Merged Multiple Ligands. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527674381.ch9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Hongming Chen
- Discovery Sciences, Innovative Medicines and Early Development; AstraZeneca; Pepparedsleden 1 431 83 Mölndal Sweden
| | - Udo Bauer
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development; AstraZeneca; Pepparedsleden 1 431 83 Mölndal Sweden
| | - Ola Engkvist
- Discovery Sciences, Innovative Medicines and Early Development; AstraZeneca; Pepparedsleden 1 431 83 Mölndal Sweden
| |
Collapse
|
60
|
Jamieson KL, Endo T, Darwesh AM, Samokhvalov V, Seubert JM. Cytochrome P450-derived eicosanoids and heart function. Pharmacol Ther 2017; 179:47-83. [DOI: 10.1016/j.pharmthera.2017.05.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
61
|
Toviwek B, Suphakun P, Choowongkomon K, Hannongbua S, Gleeson MP. Synthesis and evaluation of the NSCLC anti-cancer activity and physical properties of 4-aryl- N -phenylpyrimidin-2-amines. Bioorg Med Chem Lett 2017; 27:4749-4754. [DOI: 10.1016/j.bmcl.2017.08.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 08/19/2017] [Accepted: 08/25/2017] [Indexed: 10/18/2022]
|
62
|
Sanni DM, Fatoki TH, Kolawole AO, Akinmoladun AC. Xeronine structure and function: computational comparative mastery of its mystery. In Silico Pharmacol 2017; 5:8. [PMID: 28955650 DOI: 10.1007/s40203-017-0028-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Morinda citrifolia (Noni) fruit has a long history of dietary use in tropical regions of the world. Pharmacological properties that have been attributed to the fruit include anti-inflammatory, anti-cancer, and antioxidant properties. Xeronine, a small alkaloid which has been patented (US4543212) is one of the bioactive compounds of Noni fruit, which is believed to be capable of modifying the molecular structure of specific inactive proteins thereby regulating proper folding to active enzymes. Despite reports of the potential of Xeronine as therapeutic agent, its presence is controversial and its structure has not been explored. In this study, standard chemoinformatics tools and servers such as ChemSketch, ChemMine, Swisstargetprediction, SwissADME and Swisssimilarity have been employed to predict its possible structure. In addition, synthetic xeronine structures based on the known bioactive components of Noni fruit were designed. Results showed that the hypothetical structure of xeronine provided by the patent inventor is a mystery based on its <5% probable protein targets and no similarity match to the US Food and Drug Administration (FDA) approved drugs and experimental compounds by in silico evaluation. By constrast, final designed xeronine structure possess all the features that were described in the patent document, and has >40% probable protein targets related to neurodegenerative diseases such as Alzheimer's disease (AD), which possibly justifies the key function stated in the patent.
Collapse
Affiliation(s)
- David Morakinyo Sanni
- Enzyme Biotechnology and Bioinformatics Unit, Department of Biochemistry, Federal University of Technology, PMB 704 Akure, Nigeria
| | - Toluwase Hezekiah Fatoki
- Enzyme Biotechnology and Bioinformatics Unit, Department of Biochemistry, Federal University of Technology, PMB 704 Akure, Nigeria
| | - Ayodele Oluseyi Kolawole
- Enzyme Biotechnology and Bioinformatics Unit, Department of Biochemistry, Federal University of Technology, PMB 704 Akure, Nigeria
| | - Afolabi Clement Akinmoladun
- Biochemical Pharmacology and Toxicology Unit, Department of Biochemistry, Federal University of Technology, PMB 704 Akure, Nigeria
| |
Collapse
|
63
|
Lagorce D, Douguet D, Miteva MA, Villoutreix BO. Computational analysis of calculated physicochemical and ADMET properties of protein-protein interaction inhibitors. Sci Rep 2017; 7:46277. [PMID: 28397808 PMCID: PMC5387685 DOI: 10.1038/srep46277] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/13/2017] [Indexed: 12/18/2022] Open
Abstract
The modulation of PPIs by low molecular weight chemical compounds, particularly by orally bioavailable molecules, would be very valuable in numerous disease indications. However, it is known that PPI inhibitors (iPPIs) tend to have properties that are linked to poor Absorption, Distribution, Metabolism, Excretion and Toxicity (ADMET) and in some cases to poor clinical outcomes. Previously reported in silico analyses of iPPIs have essentially focused on physicochemical properties but several other ADMET parameters would be important to assess. In order to gain new insights into the ADMET properties of iPPIs, computations were carried out on eight datasets collected from several databases. These datasets involve compounds targeting enzymes, GPCRs, ion channels, nuclear receptors, allosteric modulators, oral marketed drugs, oral natural product-derived marketed drugs and iPPIs. Several trends are reported that should assist the design and optimization of future PPI inhibitors, either for drug discovery endeavors or for chemical biology projects.
Collapse
Affiliation(s)
- David Lagorce
- INSERM, U973, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Dominique Douguet
- CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Maria A. Miteva
- INSERM, U973, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | |
Collapse
|
64
|
Proschak E, Heitel P, Kalinowsky L, Merk D. Opportunities and Challenges for Fatty Acid Mimetics in Drug Discovery. J Med Chem 2017; 60:5235-5266. [PMID: 28252961 DOI: 10.1021/acs.jmedchem.6b01287] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fatty acids beyond their role as an endogenous energy source and storage are increasingly considered as signaling molecules regulating various physiological effects in metabolism and inflammation. Accordingly, the molecular targets involved in formation and physiological activities of fatty acids hold significant therapeutic potential. A number of these fatty acid targets are addressed by some of the oldest and most widely used drugs such as cyclooxygenase inhibiting NSAIDs, whereas others remain unexploited. Compounds orthosterically binding to proteins that endogenously bind fatty acids are considered as fatty acid mimetics. On the basis of their structural resemblance, fatty acid mimetics constitute a family of bioactive compounds showing specific binding thermodynamics and following similar pharmacokinetic mechanisms. This perspective systematically evaluates targets for fatty acid mimetics, investigates their common structural characteristics, and highlights demands in their discovery and design. In summary, fatty acid mimetics share particularly favorable characteristics justifying the conclusion that their therapeutic potential vastly outweighs the challenges in their design.
Collapse
Affiliation(s)
- Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Lena Kalinowsky
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| |
Collapse
|
65
|
Chaudhari R, Tan Z, Huang B, Zhang S. Computational polypharmacology: a new paradigm for drug discovery. Expert Opin Drug Discov 2017; 12:279-291. [PMID: 28067061 PMCID: PMC7241838 DOI: 10.1080/17460441.2017.1280024] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Over the past couple of years, the cost of drug development has sharply increased along with the high rate of clinical trial failures. Such increase in expenses is partially due to the inability of the "one drug - one target" approach to predict drug side effects and toxicities. To tackle this issue, an alternative approach, known as polypharmacology, is being adopted to study small molecule interactions with multiple targets. Apart from developing more potent and effective drugs, this approach allows for studies of off-target activities and the facilitation of drug repositioning. Although exhaustive polypharmacology studies in-vitro or in-vivo are not practical, computational methods of predicting unknown targets or side effects are being developed. Areas covered: This article describes various computational approaches that have been developed to study polypharmacology profiles of small molecules. It also provides a brief description of the algorithms used in these state-of-the-art methods. Expert opinion: Recent success in computational prediction of multi-targeting drugs has established polypharmacology as a promising alternative approach to tackle some of the daunting complications in drug discovery. This will not only help discover more effective agents, but also present tremendous opportunities to study novel target pharmacology and facilitate drug repositioning efforts in the pharmaceutical industry.
Collapse
Affiliation(s)
- Rajan Chaudhari
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Zhi Tan
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| | - Beibei Huang
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Shuxing Zhang
- Integrated Molecular Discovery Laboratory, Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030
| |
Collapse
|
66
|
Wang Y, Wang H, Chen HZ. AChE Inhibition-based Multi-target-directed Ligands, a Novel Pharmacological Approach for the Symptomatic and Disease-modifying Therapy of Alzheimer's Disease. Curr Neuropharmacol 2016; 14:364-75. [PMID: 26786145 PMCID: PMC4876592 DOI: 10.2174/1570159x14666160119094820] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 10/31/2015] [Accepted: 11/12/2015] [Indexed: 11/26/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia in elder people, characterised by a progressive decline in memory as a result of an impairment of cholinergic neurotransmission. To date acetylcholinesterase inhibitors (AChEIs) have become the most prescribed drugs for the symptomatic treatment of mild to moderate AD. However, the traditional “one molecule-one target” paradigm is not sufficient and appropriate to yield the desired therapeutic efficacy since multiple factors, such as amyloid-β (Aβ) deposits, neuroinflammation, oxidative stress, and decreased levels of acetylcholine (ACh) have been thought to play significant roles in the AD pathogenesis. New generation of multi-target drugs is earnestly demanded not only for ameliorating symptoms but also for modifying the disease. Herein, we delineated the catalytic and non-catalytic functions of AChE, and summarized the works of our group and others in research and development of novel AChEI-based multi-target-directed ligands (MTDLs), such as dual binding site AChEIs and multi-target AChEIs inhibiting Aβ aggregation, regulating Aβ procession, antagonizing platelet-activating factor (PAF) receptor, scavenging oxygen radical, chelating metal ions, inhibiting monoamine oxidase B (MAO-B), blocking N-methyl-D-aspartic acid (NMDA) receptor and others.
Collapse
Affiliation(s)
| | - Hao Wang
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China.
| | - Hong-zhuan Chen
- Department of Pharmacology, Institute of Medical Sciences, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, PR China.
| |
Collapse
|
67
|
Meirer K, Glatzel D, Kretschmer S, Wittmann SK, Hartmann M, Blöcher R, Angioni C, Geisslinger G, Steinhilber D, Hofmann B, Fürst R, Proschak E. Design, Synthesis and Cellular Characterization of a Dual Inhibitor of 5-Lipoxygenase and Soluble Epoxide Hydrolase. Molecules 2016; 22:molecules22010045. [PMID: 28036068 PMCID: PMC6155600 DOI: 10.3390/molecules22010045] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/15/2016] [Accepted: 12/23/2016] [Indexed: 01/13/2023] Open
Abstract
The arachidonic acid cascade is a key player in inflammation, and numerous well-established drugs interfere with this pathway. Previous studies have suggested that simultaneous inhibition of 5-lipoxygenase (5-LO) and soluble epoxide hydrolase (sEH) results in synergistic anti-inflammatory effects. In this study, a novel prototype of a dual 5-LO/sEH inhibitor KM55 was rationally designed and synthesized. KM55 was evaluated in enzyme activity assays with recombinant enzymes. Furthermore, activity of KM55 in human whole blood and endothelial cells was investigated. KM55 potently inhibited both enzymes in vitro and attenuated the formation of leukotrienes in human whole blood. KM55 was also tested in a cell function-based assay. The compound significantly inhibited the LPS-induced adhesion of leukocytes to endothelial cells by blocking leukocyte activation.
Collapse
Affiliation(s)
- Karin Meirer
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Daniel Glatzel
- Institute of Pharmaceutical Biology, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Simon Kretschmer
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Sandra K Wittmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Markus Hartmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - René Blöcher
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Carlo Angioni
- Institute of Clinical Pharmacology, Goethe-University of Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe-University of Frankfurt, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Bettina Hofmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Max-von-Laue Str. 9, D-60438 Frankfurt am Main, Germany.
| |
Collapse
|
68
|
Prati F, Cavalli A, Bolognesi ML. Navigating the Chemical Space of Multitarget-Directed Ligands: From Hybrids to Fragments in Alzheimer's Disease. Molecules 2016; 21:466. [PMID: 27070562 PMCID: PMC6273289 DOI: 10.3390/molecules21040466] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 04/03/2016] [Accepted: 04/05/2016] [Indexed: 12/15/2022] Open
Abstract
Multitarget drug discovery is one of the hottest topics and most active fields in the search for new molecules against Alzheimer’s disease (AD). Over the last 20 years, many promising multitarget-directed ligands (MTDLs) have been identified and developed at a pre-clinical level. However, how to design them in a rational way remains the most fundamental challenge of medicinal chemists. This is related to the foundational question of achieving an optimized activity towards multiple targets of interest, while preserving drug-like properties. In this respect, large hybrid molecules and small fragments are poles apart. In this review article, our aim is to appraise what we have accomplished in the development of both hybrid- and fragment-like molecules directed to diverse AD targets (i.e., acetylcholinesterase, NMDA receptors, metal chelation, BACE-1 and GSK-3β). In addition, we attempt to highlight what are the persistent needs that deserve to be improved and cared for, with the ultimate goal of moving an MTDL to AD clinical studies.
Collapse
Affiliation(s)
- Federica Prati
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy.
- Sir James Black Centre, College of Life Sciences, University of Dundee, Dundee DD1 5EH, UK.
| | - Andrea Cavalli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy.
- Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy.
| |
Collapse
|
69
|
Abboussi O, Said N, Fifel K, Lakehayli S, Tazi A, El Ganouni S. Behavioral effects of D3 receptor inhibition and 5-HT4 receptor activation on animals undergoing chronic cannabinoid exposure during adolescence. Metab Brain Dis 2016; 31:321-7. [PMID: 26497809 DOI: 10.1007/s11011-015-9753-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/21/2015] [Indexed: 01/26/2023]
Abstract
Chronic exposure to cannabinoids during adolescence results in long-lasting behavioral deficits that match some symptomatologic aspects of schizophrenia. The aim of this study was to investigate the reversibility of the emotional and the cognitive effects of chronic exposure to cannabinoids during adolescence, via subsequent modulation of the serotoninergic 5-HT4 and dopaminergic D3 receptors. RS67333 as a 5-HT4 agonist and U-99194A as a D3 antagonist were administered separately at 1 mg/kg and 20 mg/kg, and in combination at 0.5 mg/kg and 10 mg/kg to adult animals undergoing chronic treatment with the synthetic cannabinoid receptor agonist WIN55,212-2 (1 mg/kg) during adolescence. Animals were tested for anxiety-like behavior and episodic-like memory in the open field and novel object recognition tests respectively 30 minutes after the last drug administration. Chronic WIN55,212-2 treated animals exhibited a lasting disruption of episodic memory and increased anxiety levels. The effect on episodic-like memory were partially restored by acute administration of RS67333 and U-99194A and completely by administration of both drugs in combination at lower doses. However, only RS67333 (20 mg/kg) improved the anxiogenic-like effect of WIN55,212-2. These findings give further support that chronic exposure to cannabinoids during adolescence may be used as an animal model for schizophrenia, and highlight D3 and 5-HT4 receptors as potential targets for an enhanced treatment of the cognitive aspect of this disease.
Collapse
Affiliation(s)
- Oualid Abboussi
- Laboratory of Biochemistry and Neurosciences, Faculty of Sciences and Technics, Hassan 1er University, B.P. 577, Route of Casablanca, Settat, Morocco.
| | - Nadia Said
- Department of Pharmacology, Faculty of Medicine, Hassan II University, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - Karim Fifel
- Laboratory of Neurophysiology, Department of Molecular Cell Biology, Leiden University, Medical Center, PO Box 9600, Mailbox S5-P, 2300 RC, Leiden, The Netherlands
| | - Sara Lakehayli
- Department of Pharmacology, Faculty of Medicine, Hassan II University, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - Abdelouahhab Tazi
- Department of Pharmacology, Faculty of Medicine, Hassan II University, 19 Rue Tarik Bnou Ziad, Casablanca, Morocco
| | - Soumaya El Ganouni
- Laboratory of Biochemistry and Neurosciences, Faculty of Sciences and Technics, Hassan 1er University, B.P. 577, Route of Casablanca, Settat, Morocco
| |
Collapse
|
70
|
Jourdan JP, Since M, El Kihel L, Lecoutey C, Corvaisier S, Legay R, Sopkova-de Oliveira Santos J, Cresteil T, Malzert-Fréon A, Rochais C, Dallemagne P. Novel benzylidenephenylpyrrolizinones with pleiotropic activities potentially useful in Alzheimer's disease treatment. Eur J Med Chem 2016; 114:365-79. [PMID: 27046230 DOI: 10.1016/j.ejmech.2016.03.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/21/2022]
Abstract
This work describes the synthesis and the biological evaluation of novel benzylidenephenylpyrrolizinones as potential antioxidant, metal chelating or amyloid β (βA) aggregation inhibitors. Some derivatives exhibited interesting results in regard to several of the performed evaluations and appear as valuable Multi-Target Directed Ligands with potential therapeutic interest in Alzheimer's disease. Among them, compound 29 particularly appears as a valuable radical and NO scavenger, a Cu(II) and Fe(II) chelating agent and exhibits moderate βA aggregation inhibition properties. These activities, associated to a good predictive bioavailability and a lack of cytotoxicity, design it as a promising hit for further in vivo investigation.
Collapse
Affiliation(s)
- Jean-Pierre Jourdan
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Marc Since
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Laïla El Kihel
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Cédric Lecoutey
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Sophie Corvaisier
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Rémi Legay
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | | | - Thierry Cresteil
- CIBLOT, IPSIT - IFR14, 5 rue Jean Baptiste Clément, 92290 Chatenay-Malabry, France
| | - Aurélie Malzert-Fréon
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Christophe Rochais
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France.
| | - Patrick Dallemagne
- Normandie Université, UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France.
| |
Collapse
|
71
|
Khan SN, Khan AU. Breaking the Spell: Combating Multidrug Resistant 'Superbugs'. Front Microbiol 2016; 7:174. [PMID: 26925046 PMCID: PMC4757689 DOI: 10.3389/fmicb.2016.00174] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/01/2016] [Indexed: 12/15/2022] Open
Abstract
Multidrug-resistant (MDR) bacteria have become a severe threat to community wellbeing. Conventional antibiotics are getting progressively more ineffective as a consequence of resistance, making it imperative to realize improved antimicrobial options. In this review we emphasized the microorganisms primarily reported of being resistance, referred as ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumanii, Pseudomonas aeruginosa, and Enterobacteriaceae) accentuating their capacity to "escape" from routine antimicrobial regimes. The upcoming antimicrobial agents showing great potential and can serve as alternative therapeutic options are discussed. We also provided succinct overview of two evolving technologies; specifically network pharmacology and functional genomics profiling. Furthermore, In vivo imaging techniques can provide novel targets and a real time tool for potential lead molecule assessment. The employment of such approaches at prelude of a drug development process, will enables more informed decisions on candidate drug selection and will maximize or predict therapeutic potential before clinical testing.
Collapse
Affiliation(s)
| | - Asad U. Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim UniversityAligarh, India
| |
Collapse
|
72
|
Efavirenz a nonnucleoside reverse transcriptase inhibitor of first-generation: Approaches based on its medicinal chemistry. Eur J Med Chem 2016; 108:455-465. [DOI: 10.1016/j.ejmech.2015.11.025] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/12/2015] [Accepted: 11/17/2015] [Indexed: 11/21/2022]
|
73
|
Lavecchia A, Cerchia C. In silico methods to address polypharmacology: current status, applications and future perspectives. Drug Discov Today 2015; 21:288-98. [PMID: 26743596 DOI: 10.1016/j.drudis.2015.12.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/20/2015] [Accepted: 12/21/2015] [Indexed: 12/15/2022]
Abstract
Polypharmacology, a new paradigm in drug discovery that focuses on multi-target drugs (MTDs), has potential application for drug repurposing, the process of finding new uses for existing approved drugs, prediction of off-target toxicities and rational design of MTDs. In this scenario, computational strategies have demonstrated great potential in predicting polypharmacology and in facilitating drug repurposing. Here, we provide a comprehensive overview of various computational approaches that enable the prediction and analysis of in vitro and in vivo drug-response phenotypes and outline their potential for drug discovery. We give an outlook on the latest advances in rational design of MTDs and discuss possible future directions of algorithm development in this field.
Collapse
Affiliation(s)
- Antonio Lavecchia
- Department of Pharmacy, Drug Discovery Laboratory, University of Napoli Federico II, via D. Montesano 49, I-80131 Napoli, Italy.
| | - Carmen Cerchia
- Department of Pharmacy, Drug Discovery Laboratory, University of Napoli Federico II, via D. Montesano 49, I-80131 Napoli, Italy
| |
Collapse
|
74
|
Thomas MP, Potter BVL. Discovery and Development of the Aryl O-Sulfamate Pharmacophore for Oncology and Women's Health. J Med Chem 2015; 58:7634-58. [PMID: 25992880 PMCID: PMC5159624 DOI: 10.1021/acs.jmedchem.5b00386] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In 1994, following work from this laboratory, it was reported that estrone-3-O-sulfamate irreversibly inhibits a new potential hormone-dependent cancer target steroid sulfatase (STS). Subsequent drug discovery projects were initiated to develop the core aryl O-sulfamate pharmacophore that, over some 20 years, have led to steroidal and nonsteroidal drugs in numerous preclinical and clinical trials, with promising results in oncology and women's health, including endometriosis. Drugs have been designed to inhibit STS, e.g., Irosustat, as innovative dual-targeting aromatase-steroid sulfatase inhibitors (DASIs) and as multitargeting agents for hormone-independent tumors, such as the steroidal STX140 and nonsteroidal counterparts, acting inter alia through microtubule disruption. The aryl sulfamate pharmacophore is highly versatile, operating via three distinct mechanisms of action, and imbues attractive pharmaceutical properties. This Perspective gives a personal view of the work leading both to the therapeutic concepts and these drugs, their current status, and how they might develop in the future.
Collapse
Affiliation(s)
- Mark P. Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath, BA2 7AY, United Kingdom
| | - Barry V. L. Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
75
|
Thomas MP, Potter BVL. Estrogen O-sulfamates and their analogues: Clinical steroid sulfatase inhibitors with broad potential. J Steroid Biochem Mol Biol 2015; 153:160-9. [PMID: 25843211 DOI: 10.1016/j.jsbmb.2015.03.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 03/31/2015] [Indexed: 01/20/2023]
Abstract
Estrogen sulfamate derivatives were the first irreversible active-site-directed inhibitors of steroid sulfatase (STS), an emerging drug target for endocrine therapy of hormone dependent diseases that catalyzes inter alia the hydrolysis of estrone sulfate to estrone. In recent years this has stimulated clinical investigation of the estradiol derivative both as an oral prodrug and its currently ongoing exploration in endometriosis. 2-Substituted steroid sulfamate derivatives show considerable potential as multi-targeting agents for hormone-independent disease, but are also potent STS inhibitors. The steroidal template has spawned nonsteroidal STS inhibitors one of which, Irosustat, has been evaluated clinically in breast cancer, endometrial cancer and prostate cancer and there is potential for innovative dual-targeting approaches. This review surveys the role of estrogen sulfamates, their analogues and current status.
Collapse
Affiliation(s)
- Mark P Thomas
- Wolfson Laboratory of Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Barry V L Potter
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom.
| |
Collapse
|
76
|
Ross AG, Benton BM, Chin D, De Pascale G, Fuller J, Leeds JA, Reck F, Richie DL, Vo J, LaMarche MJ. Synthesis of ciprofloxacin dimers for evaluation of bacterial permeability in atypical chemical space. Bioorg Med Chem Lett 2015; 25:3468-75. [PMID: 26189081 DOI: 10.1016/j.bmcl.2015.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/30/2015] [Accepted: 07/02/2015] [Indexed: 11/19/2022]
Abstract
We describe the synthesis and evaluation of a library of variably-linked ciprofloxacin dimers. These structures unify and expand on the use of fluoroquinolones as probes throughout the antibiotic literature. A dimeric analog (19) showed enhanced inhibition of its intracellular target (DNA gyrase), and translation to antibacterial activity in whole cells was demonstrated. Overall, cell permeation was governed by physicochemical properties and bacterial type. A principal component analysis demonstrated that the dimers occupy a unique and privileged region of chemical space most similar to the macrolide class of antibiotics.
Collapse
Affiliation(s)
- Audrey G Ross
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Bret M Benton
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Donovan Chin
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - John Fuller
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Jennifer A Leeds
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Folkert Reck
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Daryl L Richie
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Jason Vo
- Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | | |
Collapse
|
77
|
Rochais C, Lecoutey C, Gaven F, Giannoni P, Hamidouche K, Hedou D, Dubost E, Genest D, Yahiaoui S, Freret T, Bouet V, Dauphin F, Sopkova de Oliveira Santos J, Ballandonne C, Corvaisier S, Malzert-Fréon A, Legay R, Boulouard M, Claeysen S, Dallemagne P. Novel multitarget-directed ligands (MTDLs) with acetylcholinesterase (AChE) inhibitory and serotonergic subtype 4 receptor (5-HT4R) agonist activities as potential agents against Alzheimer's disease: the design of donecopride. J Med Chem 2015; 58:3172-87. [PMID: 25793650 DOI: 10.1021/acs.jmedchem.5b00115] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
In this work, we describe the synthesis and in vitro evaluation of a novel series of multitarget-directed ligands (MTDL) displaying both nanomolar dual-binding site (DBS) acetylcholinesterase inhibitory effects and partial 5-HT4R agonist activity, among which donecopride was selected for further in vivo evaluations in mice. The latter displayed procognitive and antiamnesic effects and enhanced sAPPα release, accounting for a potential symptomatic and disease-modifying therapeutic benefit in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Christophe Rochais
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Cédric Lecoutey
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Florence Gaven
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Patrizia Giannoni
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Katia Hamidouche
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Damien Hedou
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Emmanuelle Dubost
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - David Genest
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Samir Yahiaoui
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Thomas Freret
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Valentine Bouet
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - François Dauphin
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | | | - Céline Ballandonne
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Sophie Corvaisier
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France.,⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Aurélie Malzert-Fréon
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Remi Legay
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| | - Michel Boulouard
- ⊥UNICAEN, GMPc5 (Groupe Mémoire et Plasticité comportementale), F-14032 Caen, France
| | - Sylvie Claeysen
- ‡CNRS, UMR-5203, Institut de Génomique Fonctionnelle, F-34000 Montpellier, France.,§Inserm, U1191, F-34000 Montpellier, France.,∥Université de Montpellier, UMR-5203, F-34000 Montpellier, France
| | - Patrick Dallemagne
- †UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), F-14032 Caen, France
| |
Collapse
|
78
|
Sasso O, Migliore M, Habrant D, Armirotti A, Albani C, Summa M, Moreno-Sanz G, Scarpelli R, Piomelli D. Multitarget fatty acid amide hydrolase/cyclooxygenase blockade suppresses intestinal inflammation and protects against nonsteroidal anti-inflammatory drug-dependent gastrointestinal damage. FASEB J 2015; 29:2616-27. [PMID: 25757568 DOI: 10.1096/fj.15-270637] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 02/19/2015] [Indexed: 01/02/2023]
Abstract
The ability of nonsteroidal anti-inflammatory drugs (NSAIDs) to inhibit cyclooxygenase (Cox)-1 and Cox-2 underlies the therapeutic efficacy of these drugs, as well as their propensity to damage the gastrointestinal (GI) epithelium. This toxic action greatly limits the use of NSAIDs in inflammatory bowel disease (IBD) and other chronic pathologies. Fatty acid amide hydrolase (FAAH) degrades the endocannabinoid anandamide, which attenuates inflammation and promotes GI healing. Here, we describe the first class of systemically active agents that simultaneously inhibit FAAH, Cox-1, and Cox-2 with high potency and selectivity. The class prototype 4: (ARN2508) is potent at inhibiting FAAH, Cox-1, and Cox-2 (median inhibitory concentration: FAAH, 0.031 ± 0.002 µM; Cox-1, 0.012 ± 0.002 µM; and Cox-2, 0.43 ± 0.025 µM) but does not significantly interact with a panel of >100 off targets. After oral administration in mice, ARN2508 engages its intended targets and exerts profound therapeutic effects in models of intestinal inflammation. Unlike NSAIDs, ARN2508 causes no gastric damage and indeed protects the GI from NSAID-induced damage through a mechanism that requires FAAH inhibition. Multitarget FAAH/Cox blockade may provide a transformative approach to IBD and other pathologies in which FAAH and Cox are overactive.
Collapse
Affiliation(s)
- Oscar Sasso
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Marco Migliore
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Damien Habrant
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Andrea Armirotti
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Clara Albani
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Maria Summa
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Guillermo Moreno-Sanz
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Rita Scarpelli
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| | - Daniele Piomelli
- *Department of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genoa, Italy; and Departments of Anatomy and Neurobiology and Pharmacology and Biological Chemistry, University of California-Irvine, Irvine, California, USA
| |
Collapse
|
79
|
Design of donecopride, a dual serotonin subtype 4 receptor agonist/acetylcholinesterase inhibitor with potential interest for Alzheimer's disease treatment. Proc Natl Acad Sci U S A 2014; 111:E3825-30. [PMID: 25157130 DOI: 10.1073/pnas.1410315111] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
RS67333 is a partial serotonin subtype 4 receptor (5-HT4R) agonist that has been widely studied for its procognitive effect. More recently, it has been shown that its ability to promote the nonamyloidogenic cleavage of the precursor of the neurotoxic amyloid-β peptide leads to the secretion of the neurotrophic protein sAPPα. This effect has generated great interest in RS67333 as a potential treatment for Alzheimer's disease (AD). We show herein that RS67333 is also a submicromolar acetylcholinesterase (AChE) inhibitor and therefore, could contribute, through this effect, to the restoration of the cholinergic neurotransmission that becomes altered in AD. We planned to pharmacomodulate RS67333 to enhance its AChE inhibitory activity to take advantage of this pleiotropic pharmacological profile in the design of a novel multitarget-directed ligand that is able to exert not only a symptomatic but also, a disease-modifying effect against AD. These efforts allowed us to select donecopride as a valuable dual (h)5-HT4R partial agonist (Ki = 10.4 nM; 48.3% of control agonist response)/(h)AChEI (IC50 = 16 nM) that further promotes sAPPα release (EC50 = 11.3 nM). Donecopride, as a druggable lead, was assessed for its in vivo procognitive effects (0.1, 0.3, 1, and 3 mg/kg) with an improvement of memory performances observed at 0.3 and 1 mg/kg on the object recognition test. On the basis of these in vitro and in vivo activities, donecopride seems to be a promising drug candidate for AD treatment.
Collapse
|
80
|
On 'polypharmacy' and multi-target agents, complementary strategies for improving the treatment of depression: a comparative appraisal. Int J Neuropsychopharmacol 2014; 17:1009-37. [PMID: 23719026 DOI: 10.1017/s1461145712001496] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Major depression is a heterogeneous disorder, both in terms of symptoms, ranging from anhedonia to cognitive impairment, and in terms of pathogenesis, with many interacting genetic, epigenetic, developmental and environmental causes. Accordingly, it seems unlikely that depressive states could be fully controlled by a drug possessing one discrete mechanism of action and, in the wake of disappointing results with several classes of highly selective agent, multi-modal treatment concepts are attracting attention. As concerns pharmacotherapy, there are essentially two core strategies. First, multi-target antidepressants that act via two or more complementary mechanisms and, second, polypharmacy, which refers to co-administration of two distinct drugs, usually in separate pills. Both multi-target agents and polypharmacy ideally couple a therapeutically unexploited action to a clinically established mechanism in order to enhance efficacy, moderate side-effects, accelerate onset of action and treat a broader range of symptoms. The melatonin MT1/MT2 agonist and 5-HT(2C) antagonist, agomelatine, which is effective in the short- and long-term treatment of depression, exemplifies the former approach, while evidence-based polypharmacy is illustrated by the adjunctive use of second-generation antipsychotics with serotonin reuptake inhibitors for treatment of resistant depression. Histone acetylation and methylation, ghrelin signalling, inflammatory modulators, metabotropic glutamate-7 receptors and trace amine-associated-1 receptors comprise attractive substrates for new multi-target and polypharmaceutical strategies. The present article outlines the rationale underpinning multi-modal approaches for treating depression, and critically compares and contrasts the pros and cons of established and potentially novel multi-target vs. polypharmaceutical treatments. On balance, the former appear the most promising for the elaboration, development and clinical implementation of innovative concepts for the more effective management of depression.
Collapse
|
81
|
Multitarget ligands and theranostics: sharpening the medicinal chemistry sword against prion diseases. Future Med Chem 2014; 6:1017-29. [DOI: 10.4155/fmc.14.56] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Prion diseases (PrDs) are fatal neurodegenerative disorders, for which no effective therapeutic and diagnostic tools exist. The main pathogenic event has been identified as the misfolding of a disease-associated prion protein. Nevertheless, pathogenesis seems to involve an intricate array of concomitant processes. Thus, it may be unlikely that drugs acting on single targets can effectively control PrDs. In addition, diagnosis occurs late in the disease process, by which point it is difficult to determine a successful therapeutic intervention. In this context, multitarget ligands (MTLs) and theranostic ligands (TLs) emerge for their potential to effectively cure and diagnose PrDs. In this review, we discuss the medicinal chemistry challenges of identifying novel MTLs and TLs against PrDs, and envision their impact on prion drug discovery.
Collapse
|
82
|
Filimonov DA, Lagunin AA, Gloriozova TA, Rudik AV, Druzhilovskii DS, Pogodin PV, Poroikov VV. Prediction of the Biological Activity Spectra of Organic Compounds Using the Pass Online Web Resource. Chem Heterocycl Compd (N Y) 2014. [DOI: 10.1007/s10593-014-1496-1] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
83
|
Abstract
The productivity challenge facing the pharmaceutical industry is well documented. Strategies to improve productivity have mainly focused on enhancing efficiency, such as the application of Lean Six Sigma process improvement methods and the introduction of modeling and simulation in place of ‘wet’ experiments. While these strategies have their benefits, the real challenge is to improve effectiveness by reducing clinical failure rates. We advocate redesigning the screening cascade to identify and optimize novel compounds with improved efficacy against disease, not just with improved potency against the target. There should be greater use of disease-relevant phenotypic screens in conjunction with target-based assays to drive medicinal chemistry optimization. An opportunistic approach to polypharmacology is recommended. There should also be more emphasis on optimization of the molecular mechanism of action incorporating understanding of binding kinetics, consideration of covalent drug strategies and targeting allosteric modulators.
Collapse
|
84
|
Raj R, Sharma V, Hopper MJ, Patel N, Hall D, Wrischnik LA, Land KM, Kumar V. Synthesis and preliminary in vitro activity of mono- and bis-1 H-1,2,3-triazole-tethered β-lactam-isatin conjugates against the human protozoal pathogen Trichomonas vaginalis. Med Chem Res 2014; 23:3671-3680. [PMID: 32214766 PMCID: PMC7080013 DOI: 10.1007/s00044-014-0956-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 02/12/2014] [Indexed: 02/03/2023]
Abstract
In this study, we describe the synthesis of mono- and bis-1H-1,2,3-triazole-tethered β-lactam-isatin conjugates using copper-catalysed azide-alkyne cycloaddition reaction between mono- and di-propargylated azetidin-2-ones and N-alkylazido isatins. The synthesized conjugates were evaluated for their preliminary in vitro analysis against Trichomonas vaginalis at 50 μM. The efficacy of synthesized hybrids was observed to depend on the substituent at N-1 position of β-lactam ring, as well as the presence of single/double 1H-1,2,3-triazole linker. Among the synthesized conjugates, the presence of a p-tolyl substituent at N-1 of β-lactam ring was preferred for good activity profiles while the increase in spacer length did not influence the efficacy of the compounds. Compounds with high levels of potency were further analysed to determine their IC50 values, as well as cytotoxicity profiles against mammalian cells. The most active compound in the synthesized conjugates displayed an IC50 value of 10.49 μM against cultured G3 strain of T. vaginalis and was non-toxic to cultured mammalian HeLa cells at the same concentration.
Collapse
Affiliation(s)
- Raghu Raj
- Department of Chemistry, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Vaishali Sharma
- Department of Chemistry, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| | - Melissa J. Hopper
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211 USA
| | - Neal Patel
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211 USA
| | - Dominique Hall
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211 USA
| | - Lisa A. Wrischnik
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211 USA
| | - Kirkwood M. Land
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211 USA
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, 143005 Punjab India
| |
Collapse
|
85
|
Raj R, Gut J, Rosenthal PJ, Kumar V. 1H-1,2,3-Triazole-tethered isatin-7-chloroquinoline and 3-hydroxy-indole-7-chloroquinoline conjugates: synthesis and antimalarial evaluation. Bioorg Med Chem Lett 2014; 24:756-9. [PMID: 24424135 DOI: 10.1016/j.bmcl.2013.12.109] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 12/23/2013] [Accepted: 12/25/2013] [Indexed: 01/13/2023]
Abstract
A series of 1H-1,2,3-triazole-tethered isatin-7-chloroquinoline and 3-hydroxy-indole-7-chloroquinoline conjugates have been synthesized and evaluated for their antimalarial activity against chloroquine-resistant W2 strain of Plasmodium falciparum. The most potent of the test compound with an optimum combination of 3-hydroxy-indole ring and a n-butyl linker displayed an IC50 value of 69 nM.
Collapse
Affiliation(s)
- Raghu Raj
- Department of Chemistry, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Jiri Gut
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar 143005, Punjab, India.
| |
Collapse
|
86
|
Heteromeric dopamine receptor signaling complexes: emerging neurobiology and disease relevance. Neuropsychopharmacology 2014; 39:156-68. [PMID: 23774533 PMCID: PMC3857642 DOI: 10.1038/npp.2013.148] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Revised: 04/29/2013] [Accepted: 05/10/2013] [Indexed: 12/13/2022]
Abstract
The pharmacological modification of dopamine transmission has long been employed as a therapeutic tool in the treatment of many mental health disorders. However, as many of the pharmacotherapies today are not without significant side effects, or they alleviate only a particular subset of symptoms, the identification of novel therapeutic targets is imperative. In light of these challenges, the recognition that dopamine receptors can form heteromers has significantly expanded the range of physiologically relevant signaling complexes as well as potential drug targets. Furthermore, as the physiology and disease relevance of these receptor heteromers is further understood, their ability to exhibit pharmacological and functional properties distinct from their constituent receptors, or modulate the function of endogenous homomeric receptor complexes, may allow for the development of alternate therapeutic strategies and provide new avenues for drug design. In this review, we describe the emerging neurobiology of the known dopamine receptor heteromers, their physiological relevance in brain, and discuss the potential role of these receptor complexes in neuropsychiatric disease. We highlight their value as targets for future drug development and discuss innovative research strategies designed to selectively target these dopamine receptor heteromers in the search for novel and clinically efficacious pharmacotherapies.
Collapse
|
87
|
Proschak E. Reconsidering the drug discovery pipeline for designed multitarget drugs. Drug Discov Today 2013; 18:1129-30. [DOI: 10.1016/j.drudis.2013.08.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 10/26/2022]
|
88
|
Liggi S, Drakakis G, Hendry AE, Hanson KM, Brewerton SC, Wheeler GN, Bodkin MJ, Evans DA, Bender A. Extensions to In Silico Bioactivity Predictions Using Pathway Annotations and Differential Pharmacology Analysis: Application toXenopus laevisPhenotypic Readouts. Mol Inform 2013; 32:1009-24. [DOI: 10.1002/minf.201300102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/06/2013] [Indexed: 12/20/2022]
|
89
|
Lee JA, Berg EL. Neoclassic drug discovery: the case for lead generation using phenotypic and functional approaches. ACTA ACUST UNITED AC 2013; 18:1143-55. [PMID: 24080259 DOI: 10.1177/1087057113506118] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Innovation and new molecular entity production by the pharmaceutical industry has been below expectations. Surprisingly, more first-in-class small-molecule drugs approved by the U.S. Food and Drug Administration (FDA) between 1999 and 2008 were identified by functional phenotypic lead generation strategies reminiscent of pre-genomics pharmacology than contemporary molecular targeted strategies that encompass the vast majority of lead generation efforts. This observation, in conjunction with the difficulty in validating molecular targets for drug discovery, has diminished the impact of the "genomics revolution" and has led to a growing grassroots movement and now broader trend in pharma to reconsider the use of modern physiology-based or phenotypic drug discovery (PDD) strategies. This "From the Guest Editors" column provides an introduction and overview of the two-part special issues of Journal of Biomolecular Screening on PDD. Terminology and the business case for use of PDD are defined. Key issues such as assay performance, chemical optimization, target identification, and challenges to the organization and implementation of PDD are discussed. Possible solutions for these challenges and a new neoclassic vision for PDD that combines phenotypic and functional approaches with technology innovations resulting from the genomics-driven era of target-based drug discovery (TDD) are also described. Finally, an overview of the manuscripts in this special edition is provided.
Collapse
Affiliation(s)
- Jonathan A Lee
- 1Quantitative and Structural Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | | |
Collapse
|
90
|
Chromiński M, Banach Ł, Karczewski M, ó Proinsias K, Sharina I, Gryko D, Martin E. Synthesis and evaluation of bifunctional sGC regulators: optimization of a connecting linker. J Med Chem 2013; 56:7260-77. [PMID: 23961771 DOI: 10.1021/jm400715h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hybrid molecules composed of PpIX and cobyrinic acid derivatives conjugated through linkers of varying length and composition were prepared via 1,3-dipolar cycloaddition (CuAAC) or amidation/esteryfication reactions. They were tested for activation of soluble guanylyl cyclase (sGC), a key enzyme in the NO/cGMP signaling pathway, by an in vitro GTP→cGMP conversion assay. Using purified heme-deficient sGC and truncated sGC variants lacking a heme-binding domain, we demonstrated that such hybrid molecules may activate sGC by targeting heme-binding and/or catalytic domain. While all conjugates activated sGC, only selected compounds served as bifunctional regulators and were capable of simultaneous targeting both heme and catalytic domains of sGC. The length and type of a linker connecting both components had a profound effect on the extent of sGC activation, indicating that the linker's type is crucial for their binding affinities with regulatory and catalytic domains. Only hybrids with the conjugated linker of 13-16 atom length synergistically target both domains and displayed the lowest EC50 and highest activating potency. Compounds with shorter connecting linkers were much less potent and were no more active than the cobyrinic acid component alone. The most active conjugate, which showed a 60-fold activation of sGC, was compound 11, in which PpIX and cobyrinic acid components are separated by 11 atoms chain with the triazole moiety in between.
Collapse
Affiliation(s)
- Mikołaj Chromiński
- Institute of Organic Chemistry, Polish Academy of Sciences , Kasprzaka 44/52, 01-224 Warsaw, Poland
| | | | | | | | | | | | | |
Collapse
|
91
|
McLoughlin D, Bertelli F, Williams C. The A, B, Cs of G-protein-coupled receptor pharmacology in assay development for HTS. Expert Opin Drug Discov 2013; 2:603-19. [PMID: 23488953 DOI: 10.1517/17460441.2.5.603] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
G-protein-coupled receptors represent one of the most important areas of research in the pharmaceutical industry, being one of the largest druggable gene families. Recognising this fact, manufacturers have developed a huge variety of homogeneous assay technologies that facilitate the quantification of receptor ligand binding events and their downstream signalling cascades. However, while early emphasis was placed on the most sensitive, high-throughput and cost-effective screening technologies to enable identification of the most lead matter for further development, in recent years emphasis has shifted to a focus on maximising the identification of compounds that are new and developing assays that are more biologically/pharmacologically relevant. Therefore, this review provides an overview of the binding and functional techniques available for high-throughput screening, with particular attention on how assay application and configuration can be maximised to ensure their successful identification of relevant chemical matter and thereby optimising project success.
Collapse
Affiliation(s)
- Dj McLoughlin
- HTS CoE, Pfizer Global Research and Development, Ramsgate Road, Sandwich, Kent, CT13 9NJ, UK +44(0)1304644616 ; +44(0)1304655592 ;
| | | | | |
Collapse
|
92
|
Chen Y, Sun J, Huang Z, Liao H, Peng S, Lehmann J, Zhang Y. Design, synthesis and evaluation of tacrine-flurbiprofen-nitrate trihybrids as novel anti-Alzheimer's disease agents. Bioorg Med Chem 2013; 21:2462-70. [PMID: 23541836 DOI: 10.1016/j.bmc.2013.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 11/18/2022]
Abstract
To search for multifunctional anti-Alzheimer's disease (AD) agents with good safety, the previously synthesized tacrine-flurbiprofen hybrids 1a and 1b were modified into tacrine-flurbiprofen-nitrate trihybrids 3a-h. These compounds displayed comparable or higher cholinesterase inhibitory activity relative to the bivalent hybrids. Compound 3a was the most potent, which released moderate NO, exerted blood vessel relaxative activity, and showed significant Aβ inhibitory effects whereas tacrine and flurbiprofen did not exhibit any Aβ inhibitory activity at the same dose. In addition, 3a was active in improving memory impairment in vivo. More importantly, the hepatotoxicity study showed that 3a was much safer than tacrine, suggesting it might be a promising anti-AD agent for further investigation.
Collapse
Affiliation(s)
- Yao Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | | | | | |
Collapse
|
93
|
Chromiński M, Proinsias KÓ, Martin E, Gryko D. Protoporphyrin IX/Cobyrinate Derived Hybrids - Novel Activators of Soluble Guanylyl Cyclase. European J Org Chem 2013; 2013:1530-1537. [PMID: 23519483 PMCID: PMC3600653 DOI: 10.1002/ejoc.201201303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Indexed: 11/10/2022]
Abstract
A new cobyrinate/protoporphyrin IX molecular hybrids were prepared via CuAAC reaction. The synthesis involved selective preparation of cobyrinate and PpIX derived building blocks possessing respectively terminal alkyne and azide moieties followed by the CuOAc catalyzed cycloaddition reaction. Synthesized molecules activated soluble guanylyl cyclase showing strong linker length/activation dependence.
Collapse
Affiliation(s)
- Mikołaj Chromiński
- Institute of Organic Chemistry Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Keith ó Proinsias
- Institute of Organic Chemistry Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Emil Martin
- Department of Internal Medicine, Division of Cardiology, University of Texas Health Science Center in Houston, 1941 East Road, The University of Texas, Houston, Texas USA, 77054
| | - Dorota Gryko
- Institute of Organic Chemistry Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
94
|
Gattrell W, Johnstone C, Patel S, Smith CS, Scheel A, Schindler M. Designed multiple ligands in metabolic disease research: from concept to platform. Drug Discov Today 2013; 18:692-6. [PMID: 23454344 DOI: 10.1016/j.drudis.2013.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/31/2013] [Accepted: 02/14/2013] [Indexed: 10/27/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a multifactorial disease, and drug monotherapy typically results in unsatisfactory treatment outcomes for patients. Even when used in combination, existing therapies lack efficacy in the long term. Designed multiple ligands (DMLs) are compounds developed to modulate multiple targets relevant to a disease. DMLs offer the potential to yield greater efficacy over monotherapies, either by modulating different biological pathways, or by boosting a single one. However, examples of DMLs progressing into clinical trials, or onto the market are rare; DML drug discovery is challenging, and perceived by some to be almost impossible. Nevertheless, with the judicious selection of biological targets, both from a biological and chemical perspective, it is possible to develop drug-like DMLs.
Collapse
Affiliation(s)
- W Gattrell
- Research Evaluation Unit, Oxford PharmaGenesis, Oxford, UK.
| | | | | | | | | | | |
Collapse
|
95
|
East SP, Silver LL. Multitarget ligands in antibacterial research: progress and opportunities. Expert Opin Drug Discov 2012; 8:143-56. [PMID: 23252414 DOI: 10.1517/17460441.2013.743991] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Resistance to current antibacterial therapies is an inevitability that represents a significant global health concern. Bacteria have the capacity to render all current drug treatments ineffective, which places a demand on the drug discovery community to constantly develop new antibacterial agents. Compounds that inhibit multiple biological targets, often referred to as multitarget ligands, are an inviting prospect in antibacterial research because, although they will not solve the issue of resistance, they might help to delay the onset. AREAS COVERED This review covers some of the recent progress in identifying new ligands that deliberately interact with more than one essential biological target in bacteria. The two principal areas covered are inhibitors of DNA replication and cell wall biosynthesis. EXPERT OPINION Antibacterial programs for the design of multitarget ligands present an important opportunity for production of antibacterial agents. Their longevity, due to slow development of resistance, is comparable to that seen with other successful agents - but is much improved over single-targeted agents for which resistance can appear in vitro overnight. The preclinical development of these agents will have to overcome the standard problems of antibacterial discovery. Such problems include optimization of characteristics favoring cell entry and particularly the demonstration of selectivity of inhibition of the desired multiple targets without inhibition of other bacterial or any mammalian functions.
Collapse
Affiliation(s)
- Stephen P East
- Evotec (UK) Ltd., 114 Milton Park, Abingdon, Oxfordshire OX14 4SA, UK.
| | | |
Collapse
|
96
|
Geldenhuys WJ, Van der Schyf CJ. Designing drugs with multi-target activity: the next step in the treatment of neurodegenerative disorders. Expert Opin Drug Discov 2012; 8:115-29. [DOI: 10.1517/17460441.2013.744746] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Werner J Geldenhuys
- Northeast Ohio Medical University, College of Pharmacy, Neurotherapeutics Emphasis Group, Department of Pharmaceutical Sciences,
Rootstown, 4209 State Route 44, P.O. Box 95, OH 44272, USA ;
| | - Cornelis J Van der Schyf
- Northeast Ohio Medical University, College of Pharmacy, Neurotherapeutics Emphasis Group, Department of Pharmaceutical Sciences,
Rootstown, 4209 State Route 44, P.O. Box 95, OH 44272, USA ;
| |
Collapse
|
97
|
Hanson RN, Hua E, Labaree D, Hochberg RB, Proffitt K, Essigmann JM, Croy RG. Convergent synthesis of a steroidal antiestrogen-mitomycin C hybrid using "click" chemistry. Org Biomol Chem 2012; 10:8501-8. [PMID: 23007562 DOI: 10.1039/c2ob25902h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A convergent synthesis of a novel estrogen receptor-targeted drug hybrid was developed based on structures of the potent anti-proliferative mitomycin C and the steroidal anti-estrogen RU 39411. The steroidal antiestrogen was prepared with an azido-triethylene glycoloxy linker while the mitomycin C derivative (porfirimycin) incorporated a complementary 7-N-terminal alkyne. The two components were ligated using the Huisgen [3 + 2] cycloaddition ("click") reaction. Preliminary biological assays demonstrated that the final hybrid compound retained both potent anti-estrogenic and anti-proliferative activities.
Collapse
Affiliation(s)
- Robert N Hanson
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA.
| | | | | | | | | | | | | |
Collapse
|
98
|
Liu KC, Fang JM, Jan JT, Cheng TJR, Wang SY, Yang ST, Cheng YSE, Wong CH. Enhanced anti-influenza agents conjugated with anti-inflammatory activity. J Med Chem 2012; 55:8493-501. [PMID: 22963087 DOI: 10.1021/jm3009844] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Influenza therapy with a single targeted compound is often limited in efficacy due to the rapidly developed drug resistance. Moreover, the uncontrolled virus-induced cytokines could cause the high mortality of human infected by H5N1 avian influenza virus. In this study, we explored the novel dual-targeted bifunctional anti-influenza drugs formed by conjugation with anti-inflammatory agents. In particular, the caffeic acid (CA)-bearing zanamivir (ZA) conjugates ZA-7-CA (1) and ZA-7-CA-amide (7) showed simultaneous inhibition of influenza virus neuraminidase and suppression of pro-inflammatory cytokines. These ZA conjugates provided remarkable protection of cells and mice against influenza infections. Intranasal administration of low dosage (<1.2 μmol/kg/day) of ZA conjugates exhibited much greater effect than the combination therapy with ZA and the anti-inflammatory agents in protection of the lethally infected mice by H1N1 or H5N1 influenza viruses.
Collapse
Affiliation(s)
- Kung-Cheng Liu
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Ajmani S, Kulkarni SA. Application of GQSAR for Scaffold Hopping and Lead Optimization in Multitarget Inhibitors. Mol Inform 2012; 31:473-90. [DOI: 10.1002/minf.201100160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 04/27/2012] [Indexed: 11/09/2022]
|
100
|
Richard Morphy J. The Challenges of Multi-Target Lead Optimization. DESIGNING MULTI-TARGET DRUGS 2012. [DOI: 10.1039/9781849734912-00141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
During multi-target lead optimization, medicinal chemists must address three principal challenges. First, the various desired activities must be adequately balanced and the activity profile of any active metabolites must also be considered. Second, undesired activities associated with side effects need to be removed. Third, a pharmacokinetic profile appropriate for the desired route of administration, usually oral, must be obtained. Given the extent of these challenges, the need for a high quality lead compound is paramount.
Collapse
|