1
|
Tripathi RKP. A perspective review on fatty acid amide hydrolase (FAAH) inhibitors as potential therapeutic agents. Eur J Med Chem 2019; 188:111953. [PMID: 31945644 DOI: 10.1016/j.ejmech.2019.111953] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/02/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023]
Abstract
Fatty acid amide hydrolase (FAAH) is an important enzyme creditworthy of hydrolyzing endocannabinoids and related-amidated signalling lipids, discovery of which has pioneered novel arena of pharmacological canvasses to unwrap its curative potency in various diseased circumstances. It presents contemporary basis for understanding molecules regulating and mediating inflammatory reactions, pain, anxiety, depression, and neurodegeneration. FAAH inhibitors form vital approach for discovery of therapeutic agents that are concerned with local elevation of endocannabinoids under certain stimuli, debarring adverse/unwanted secondary effects from global activation of cannabinoid receptors by exogenous cannabimimetics. During past decades, several molecules with excellent potency developed through tailor-made approaches entered into clinical trials, but none could reach market. Hence, hunt for novel, non-toxic and selective FAAH inhibitors are on horizon. This review summarizes present perception on FAAH in conjunction with its structure, mechanism of catalysis and biological functions. It also foregrounds recent development of molecules belonging to diverse chemical classes as potential FAAH inhibitors bobbing up from in-depth chemical, mechanistic and computational studies published since 2015-November 2019, focusing on their potency. This review will assist readers to obtain rationale on FAAH as potential target for addressing various disease conditions, acquiring significant knowledge on recently established inhibitor scaffolds and their development potentials. New technologies including MD-MM simulations and 3D-QSAR studies allow mechanistic characterization of enzyme. Assessment of in-vitro and in-vivo efficacy of existing FAAH inhibitors will facilitate researchers to design novel ligands utilizing modern drug design methods. The discussions will also impose precaution in decision making process, quashing possibility of late stage failure.
Collapse
Affiliation(s)
- Rati Kailash Prasad Tripathi
- Department of Pharmaceutical Science, Sushruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar, Assam, 788011, India; Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
2
|
Meinig JM, Ferrara SJ, Banerji T, Banerji T, Sanford-Crane HS, Bourdette D, Scanlan TS. Structure-Activity Relationships of Central Nervous System Penetration by Fatty Acid Amide Hydrolase (FAAH)-Targeted Thyromimetic Prodrugs. ACS Med Chem Lett 2019; 10:111-116. [PMID: 30655956 DOI: 10.1021/acsmedchemlett.8b00501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 12/04/2018] [Indexed: 02/06/2023] Open
Abstract
Thyroid hormone (TH) action is of clinical interest in treating demyelinating diseases of the central nervous system (CNS). Two amide prodrugs of sobetirome, a potent thyroid hormone agonist, were previously shown to significantly improve CNS selective distribution of the parent drug through hydrolysis in the CNS by fatty acid amide hydrolase (FAAH). This concept is elaborated upon here with a series of 29 amide prodrugs targeting FAAH. We identify that conservative aliphatic modifications such as the N-methyl (4), N-ethyl (5), N-fluoroethyl (15), and N-cyclopropyl (18) substantially favor selective CNS distribution of the parent drug in mice. Additionally, lead compounds exhibit moderate to good rates of hydrolysis at FAAH in vitro suggesting both enzymatic and physicochemical properties are important parameters for optimization. Both 4 and 15 were orally bioavailable while retaining appreciable CNS parent drug delivery following an oral dose. The pharmacokinetic parameters of 4 over 24 h postdose (i.v. and p.o.) were determined.
Collapse
|
3
|
Kiss LE, Beliaev A, Ferreira HS, Rosa CP, Bonifácio MJ, Loureiro AI, Pires NM, Palma PN, Soares-da-Silva P. Discovery of a Potent, Long-Acting, and CNS-Active Inhibitor (BIA 10-2474) of Fatty Acid Amide Hydrolase. ChemMedChem 2018; 13:2177-2188. [PMID: 30113139 PMCID: PMC6582431 DOI: 10.1002/cmdc.201800393] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/27/2018] [Indexed: 11/06/2022]
Abstract
Fatty acid amide hydrolase (FAAH) can be targeted for the treatment of pain associated with various medical conditions. Herein we report the design and synthesis of a novel series of heterocyclic-N-carboxamide FAAH inhibitors that have a good alignment of potency, metabolic stability and selectivity for FAAH over monoacylglycerol lipase (MAGL) and carboxylesterases (CEs). Lead optimization efforts carried out with benzotriazolyl- and imidazolyl-N-carboxamide series led to the discovery of clinical candidate 8 l (3-(1-(cyclohexyl(methyl)carbamoyl)-1H-imidazol-4-yl)pyridine 1-oxide; BIA 10-2474) as a potent and long-acting inhibitor of FAAH. However, during a Phase I clinical trial with compound 8 l, unexpected and unpredictable serious neurological adverse events occurred, affecting five healthy volunteers, including the death of one subject.
Collapse
Affiliation(s)
- László E Kiss
- Laboratory of Chemistry, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Alexandre Beliaev
- Laboratory of Chemistry, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Humberto S Ferreira
- Laboratory of Chemistry, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Carla P Rosa
- Laboratory of Chemistry, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Maria João Bonifácio
- Laboratory of Pharmacology, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Ana I Loureiro
- Laboratory of Pharmacology, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Nuno M Pires
- Laboratory of Pharmacology, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - P Nuno Palma
- Laboratory of Pharmacology, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal
| | - Patrício Soares-da-Silva
- Laboratory of Pharmacology, Department of Research and Development, BIAL-Portela & Cª., S.A., À Avenida da Siderurgia Nacional, 4745-457, Coronado (S. Romão and S. Mamede), Portugal.,MedInUp-Center for Drug Discovery and Innovative Medicines, University of Porto, Praça Gomes Teixeira, 4099-002, Porto, Portugal
| |
Collapse
|
4
|
Dato FM, Sheikh M, Uhl RZ, Schüller AW, Steinkrüger M, Koch P, Neudörfl JM, Gütschow M, Goldfuss B, Pietsch M. ω-Phthalimidoalkyl Aryl Ureas as Potent and Selective Inhibitors of Cholesterol Esterase. ChemMedChem 2018; 13:1833-1847. [PMID: 30004170 DOI: 10.1002/cmdc.201800388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Indexed: 11/09/2022]
Abstract
Cholesterol esterase (CEase), a serine hydrolase thought to be involved in atherogenesis and thus coronary heart disease, is considered as a target for inhibitor development. We investigated recombinant human and murine CEases with a new fluorometric assay in a structure-activity relationship study of a small library of ω-phthalimidoalkyl aryl ureas. The urea motif with an attached 3,5-bis(trifluoromethyl)phenyl group and the aromatic character of the ω-phthalimide residue were most important for inhibitory activity. In addition, an alkyl chain composed of three or four methylene groups, connecting the urea and phthalimide moieties, was found to be an optimal spacer for inhibitors. The so-optimized compounds 2 [1-(3,5-bis(trifluoromethyl)phenyl)-3-(3-(1,3-dioxoisoindolin-2-yl)propyl)urea] and 21 [1-(3,5-bis(trifluoromethyl)phenyl)-3-(4-(1,3-dioxoisoindolin-2-yl)butyl)urea] exhibited dissociation constants (Ki ) of 1-19 μm on the two CEases and showed either a competitive (2 on the human enzyme and 21 on the murine enzyme) or a noncompetitive mode of inhibition. Two related serine hydrolases-monoacylglycerol lipase and fatty acid amide hydrolase-were inhibited by ω-phthalimidoalkyl aryl ureas to a lesser extent.
Collapse
Affiliation(s)
- Florian M Dato
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany.,Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Miriam Sheikh
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Rocky Z Uhl
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Alexandra W Schüller
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany.,Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Michaela Steinkrüger
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Peter Koch
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| | - Jörg-Martin Neudörfl
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, An der Immenburg 4, 53121, Bonn, Germany
| | - Bernd Goldfuss
- Institute of Organic Chemistry, Department of Chemistry, University of Cologne, Greinstrasse 4, 50939, Cologne, Germany
| | - Markus Pietsch
- Institute II of Pharmacology, Center of Pharmacology, Medical Faculty, University of Cologne, Gleueler Strasse 24, 50931, Cologne, Germany
| |
Collapse
|
5
|
Boger DL. The Difference a Single Atom Can Make: Synthesis and Design at the Chemistry-Biology Interface. J Org Chem 2017; 82:11961-11980. [PMID: 28945374 PMCID: PMC5712263 DOI: 10.1021/acs.joc.7b02088] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Indexed: 01/24/2023]
Abstract
A Perspective of work in our laboratory on the examination of biologically active compounds, especially natural products, is presented. In the context of individual programs and along with a summary of our work, selected cases are presented that illustrate the impact single atom changes can have on the biological properties of the compounds. The examples were chosen to highlight single heavy atom changes that improve activity, rather than those that involve informative alterations that reduce or abolish activity. The examples were also chosen to illustrate that the impact of such single-atom changes can originate from steric, electronic, conformational, or H-bonding effects, from changes in functional reactivity, from fundamental intermolecular interactions with a biological target, from introduction of a new or altered functionalization site, or from features as simple as improvements in stability or physical properties. Nearly all the examples highlighted represent not only unusual instances of productive deep-seated natural product modifications and were introduced through total synthesis but are also remarkable in that they are derived from only a single heavy atom change in the structure.
Collapse
Affiliation(s)
- Dale L. Boger
- Department of Chemistry and
The Skaggs Research Institute, The Scripps
Research Institute, 10550
North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
6
|
Zahov S, Garzinsky D, Hanekamp W, Lehr M. 1-Heteroarylpropan-2-ones as inhibitors of fatty acid amide hydrolase: Studies on structure-activity relationships and metabolic stability. Bioorg Med Chem 2016; 25:825-837. [PMID: 27989417 DOI: 10.1016/j.bmc.2016.11.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 11/09/2016] [Accepted: 11/13/2016] [Indexed: 10/20/2022]
Abstract
The serine hydrolase fatty acid amide hydrolase (FAAH) catalyzes the degradation of the endocannabinoid anandamide, which possesses analgesic and anti-inflammatory effects. A new series of 1-heteroarylpropan-2-ones was synthesized and evaluated for FAAH inhibition. Structure-activity relationship studies revealed that 1H-benzotriazol-1-yl, 1H-7-azabenzotriazol-1-yl, 1H-tetrazol-1-yl and 2H-tetrazol-2-yl substituents have the highest impact on inhibitory potency. Furthermore, attempts were made to increase the limited metabolic stability of the ketone functionality of these compounds towards metabolic reduction by introduction of shielding alkyl substituents in proximity of this serine reactive group.
Collapse
Affiliation(s)
- Stefan Zahov
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstrasse 48, D-48149 Münster, Germany
| | - David Garzinsky
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstrasse 48, D-48149 Münster, Germany
| | - Walburga Hanekamp
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstrasse 48, D-48149 Münster, Germany
| | - Matthias Lehr
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstrasse 48, D-48149 Münster, Germany.
| |
Collapse
|
7
|
Beliaev A, Ferreira HS, Learmonth DA, Bonifácio MJ, Torrão L, Pires NM, Soares-da-Silva P, Kiss LE. Synthesis and structure–activity relationships of ionizable 1,3,4-oxadiazol-2(3H)-ones as peripherally selective FAAH inhibitors with improved aqueous solubility. PURE APPL CHEM 2016. [DOI: 10.1515/pac-2016-0104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractNovel 5-(2,4-difluorophenoxy)-3-aryl-1,3,4-oxadiazol-2(3H)-ones were prepared and in vivo SAR are discussed. Ionisable substituents on the N-phenyl ring provided compounds with significantly improved aqueous solubility. In addition, these analogues retained equivalent or improved potency against FAAH enzyme compared to the parent phenols 2–3. FAAH inhibition by the 2-(piperazin-1-yl)ethyl and 3-(piperazin-1-yl)propyl derivatives 24 and 30 was confined to the periphery in mice (30 mg/kg), whereas hepatic FAAH activity was inhibited by over 90%.
Collapse
Affiliation(s)
- Alexandre Beliaev
- 1Laboratory of Chemistry, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| | - Humberto S. Ferreira
- 1Laboratory of Chemistry, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| | - David A. Learmonth
- 2Stemmatters, Biotecnologia e Medicina Regenerativa S.A., 4805-017 Guimarães, Portugal
| | - Maria João Bonifácio
- 3Laboratory of Pharmacology, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| | - Leonel Torrão
- 3Laboratory of Pharmacology, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| | - Nuno M. Pires
- 3Laboratory of Pharmacology, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| | | | - László E. Kiss
- 1Laboratory of Chemistry, Department of Research and Development, BIAL – Portela and Cª., S.A., Coronado (S. Romão e S. Mamede), Portugal
| |
Collapse
|
8
|
Terwege T, Hanekamp W, Garzinsky D, König S, Koch O, Lehr M. ω-Imidazolyl- and ω-Tetrazolylalkylcarbamates as Inhibitors of Fatty Acid Amide Hydrolase: Biological Activity and in vitro Metabolic Stability. ChemMedChem 2016; 11:429-43. [DOI: 10.1002/cmdc.201500445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/07/2015] [Indexed: 12/20/2022]
Affiliation(s)
- Tobias Terwege
- Institute of Pharmaceutical and Medicinal Chemistry; University of Münster; Corrensstrasse 48 48149 Münster Germany
| | - Walburga Hanekamp
- Institute of Pharmaceutical and Medicinal Chemistry; University of Münster; Corrensstrasse 48 48149 Münster Germany
| | - David Garzinsky
- Institute of Pharmaceutical and Medicinal Chemistry; University of Münster; Corrensstrasse 48 48149 Münster Germany
| | - Simone König
- Core Unit Proteomics; Interdisciplinary Center for Clinical Research (IZKF); University of Münster; Röntgenstrasse 21 48149 Münster Germany
| | - Oliver Koch
- Faculty of Chemistry and Chemical Biology; TU Dortmund University; Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Matthias Lehr
- Institute of Pharmaceutical and Medicinal Chemistry; University of Münster; Corrensstrasse 48 48149 Münster Germany
| |
Collapse
|
9
|
Micoli A, De Simone A, Russo D, Ottonello G, Colombano G, Ruda GF, Bandiera T, Cavalli A, Bottegoni G. Aryl and heteroaryl N-[4-[4-(2,3-substituted-phenyl)piperazine-1-yl]alkyl]carbamates with improved physico-chemical properties as dual modulators of dopamine D3 receptor and fatty acid amide hydrolase. MEDCHEMCOMM 2016. [DOI: 10.1039/c5md00590f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Improving the physico-chemical profile of a MTDL series.
Collapse
Affiliation(s)
- A. Micoli
- CompuNet
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
| | - A. De Simone
- School of Chemistry
- The University of Edinburgh
- Edinburgh
- UK
| | - D. Russo
- PharmaChemistry
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
| | - G. Ottonello
- PharmaChemistry
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
| | - G. Colombano
- Cancer Research UK Cancer Therapeutics Unit
- The Institute of Cancer Research
- Sutton
- UK
| | - G. F. Ruda
- Structural Genomics Consortium (SGC)
- University of Oxford
- Oxford OX3 7DQ
- UK
- Target Discovery Institute (TDI)
| | - T. Bandiera
- PharmaChemistry
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
| | - A. Cavalli
- CompuNet
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
- FaBit
| | - G. Bottegoni
- CompuNet
- Istituto Italiano di Tecnologia
- 16163 Genova
- Italy
- BiKi Technologies S.r.l
| |
Collapse
|
10
|
Abstract
INTRODUCTION Benzisoxazoles represent a class of heterocyclic compounds of great importance for the preparation of biologically active compounds. Benzisoxazoles are an important structure and some benzisoxazole-based medicines have been approved for human clinical use, including atypical antipsychotics (risperidone, paliperidone and iloperidone) and an anticonvulsant (zonisamide). AREAS COVERED This review puts emphasis on the recent progress in therapeutically attractive benzisoxazole derivatives especially 1,2-benzisoxazoles, which were published in the patent literature between 2009 and 2014. As for the class of medicines, the main focus is on atypical antipsychotics and potential therapeutic treatments for other CNS disorders. This review also covers the examples of benzisoxazole-based kinase inhibitors. Moreover, novel benzisoxazoles with significant therapeutic interest are also mentioned. EXPERT OPINION More recent examples of structural modification of existing drugs led to the discovery of some promising benzisoxazoles for antipsychotic use. The design of multi-target ligands is important for the manipulation of pharmacological properties and safety profiles for the use of antipsychotics. Benzisoxazoles have been widely used as pharmacophores in the search for novel drug candidates in a variety of therapeutic area. It is fair to assume that the wide and frequent use of benzisoxazoles in drug discovery and development will continue into the future.
Collapse
Affiliation(s)
- Yoshikazu Uto
- Daiichi Sankyo Co., Ltd, Venture Science Laboratories , 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710 , Japan
| |
Collapse
|
11
|
De Simone A, Ruda GF, Albani C, Tarozzo G, Bandiera T, Piomelli D, Cavalli A, Bottegoni G. Applying a multitarget rational drug design strategy: the first set of modulators with potent and balanced activity toward dopamine D3 receptor and fatty acid amide hydrolase. Chem Commun (Camb) 2014; 50:4904-7. [PMID: 24691497 PMCID: PMC4038168 DOI: 10.1039/c4cc00967c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Combining computer-assisted drug design and synthetic efforts, we generated compounds with potent and balanced activities toward both D3 dopamine receptor and fatty acid amide hydrolase (FAAH) enzyme. By concurrently modulating these targets, our compounds hold great potential toward exerting a disease-modifying effect on nicotine addiction and other forms of compulsive behavior.
Collapse
Affiliation(s)
- Alessio De Simone
- Dept. of Drug Discovery and Development, Istituto Italiano di Tecnologia, via Morego n.30, 16163 Genova, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Otrubova K, Cravatt BF, Boger DL. Design, synthesis, and characterization of α-ketoheterocycles that additionally target the cytosolic port Cys269 of fatty acid amide hydrolase. J Med Chem 2014; 57:1079-89. [PMID: 24456116 PMCID: PMC3940414 DOI: 10.1021/jm401820q] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
A series
of α-ketooxazoles incorporating electrophiles at
the C5 position of the pyridyl ring of 2 (OL-135) and
related compounds were prepared and examined as inhibitors of fatty
acid amide hydrolase (FAAH) that additionally target the cytosolic
port Cys269. From this series, a subset of the candidate inhibitors
exhibited time-dependent FAAH inhibition and noncompetitive irreversible
inactivation of the enzyme, consistent with the targeted Cys269 covalent
alkylation or addition, and maintained or enhanced the intrinsic selectivity
for FAAH versus other serine hydrolases. A preliminary in vivo assessment
demonstrates that these inhibitors raise endogenous brain levels of
anandamide and other FAAH substrates upon intraperitoneal (i.p.) administration
to mice, with peak levels achieved within 1.5–3 h, and that
the elevations of the signaling lipids were maintained >6 h, indicating
that the inhibitors effectively reach and remain active in the brain,
inhibiting FAAH for a sustained period.
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry, ‡Chemical Physiology, and §The Skaggs Institute for Chemical Biology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | | | |
Collapse
|
13
|
Lodola A, Rivara S, Mor M. Insights in the Mechanism of Action and Inhibition of N-Acylethanolamine Acid Amidase by Means of Computational Methods. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2014; 96:219-34. [DOI: 10.1016/bs.apcsb.2014.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Terwege T, Dahlhaus H, Hanekamp W, Lehr M. ω-Heteroarylalkylcarbamates as inhibitors of fatty acid amide hydrolase (FAAH). MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00181h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of heteroaryl-substituted alkylcarbamates have been synthesized and evaluated for their inhibitory potency against fatty acid amide hydrolase (FAAH).
Collapse
Affiliation(s)
- Tobias Terwege
- Institute of Pharmaceutical and Medicinal Chemistry
- University of Münster
- Corrensstrasse 48
- D-48149 Münster, Germany
| | - Helmut Dahlhaus
- Institute of Pharmaceutical and Medicinal Chemistry
- University of Münster
- Corrensstrasse 48
- D-48149 Münster, Germany
| | - Walburga Hanekamp
- Institute of Pharmaceutical and Medicinal Chemistry
- University of Münster
- Corrensstrasse 48
- D-48149 Münster, Germany
| | - Matthias Lehr
- Institute of Pharmaceutical and Medicinal Chemistry
- University of Münster
- Corrensstrasse 48
- D-48149 Münster, Germany
| |
Collapse
|
15
|
Lambruschini C, Veronesi M, Romeo E, Garau G, Bandiera T, Piomelli D, Scarpelli R, Dalvit C. Development of fragment-based n-FABS NMR screening applied to the membrane enzyme FAAH. Chembiochem 2013; 14:1611-9. [PMID: 23918626 DOI: 10.1002/cbic.201300347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Indexed: 12/26/2022]
Abstract
Despite the recognized importance of membrane proteins as pharmaceutical targets, the reliable identification of fragment hits that are able to bind these proteins is still a major challenge. Among different ¹⁹F NMR spectroscopic methods, n-fluorine atoms for biochemical screening (n-FABS) is a highly sensitive technique that has been used efficiently for fragment screening, but its application for membrane enzymes has not been reported yet. Herein, we present the first successful application of n-FABS to the discovery of novel fragment hits, targeting the membrane-bound enzyme fatty acid amide hydrolase (FAAH), using a library of fluorinated fragments generated based on the different local environment of fluorine concept. The use of the recombinant fusion protein MBP-FAAH and the design of compound 11 as a suitable novel fluorinated substrate analogue allowed n-FABS screening to be efficiently performed using a very small amount of enzyme. Notably, we have identified 19 novel fragment hits that inhibit FAAH with a median effective concentration (IC₅₀) in the low mM-μM range. To the best of our knowledge, these results represent the first application of a ¹⁹F NMR fragment-based functional assay to a membrane protein.
Collapse
Affiliation(s)
- Chiara Lambruschini
- Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova (Italy)
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Otrubova K, Brown M, McCormick MS, Han GW, O’Neal ST, Cravatt BF, Stevens RC, Lichtman AH, Boger DL. Rational design of fatty acid amide hydrolase inhibitors that act by covalently bonding to two active site residues. J Am Chem Soc 2013; 135:6289-99. [PMID: 23581831 PMCID: PMC3678763 DOI: 10.1021/ja4014997] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The design and characterization of α-ketoheterocycle fatty acid amide hydrolase (FAAH) inhibitors are disclosed that additionally and irreversibly target a cysteine (Cys269) found in the enzyme cytosolic port while maintaining the reversible covalent Ser241 attachment responsible for their rapid and initially reversible enzyme inhibition. Two α-ketooxazoles (3 and 4) containing strategically placed electrophiles at the C5 position of the pyridyl substituent of 2 (OL-135) were prepared and examined as inhibitors of FAAH. Consistent with the observed time-dependent noncompetitive inhibition, the cocrystal X-ray structure of 3 bound to a humanized variant of rat FAAH revealed that 3 was not only covalently bound to the active site catalytic nucleophile Ser241 as a deprotonated hemiketal, but also to Cys269 through the pyridyl C5-substituent, thus providing an inhibitor with dual covalent attachment in the enzyme active site. In vivo characterization of the prototypical inhibitors in mice demonstrates that they raise endogenous brain levels of FAAH substrates to a greater extent and for a much longer duration (>6 h) than the reversible inhibitor 2, indicating that the inhibitors accumulate and persist in the brain to completely inhibit FAAH for a prolonged period. Consistent with this behavior and the targeted irreversible enzyme inhibition, 3 reversed cold allodynia in the chronic constriction injury model of neuropathic pain in mice for a sustained period (>6 h) beyond that observed with the reversible inhibitor 2, providing effects that were unchanged over the 1-6 h time course monitored.
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Monica Brown
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Michael S. McCormick
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Gye W. Han
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Scott T. O’Neal
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Raymond C. Stevens
- Department of Molecular Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Aron H. Lichtman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| |
Collapse
|
17
|
Otrubova K, Boger DL. α-Ketoheterocycle-based Inhibitors of Fatty Acid Amide Hydrolase (FAAH). ACS Chem Neurosci 2012; 3:340-348. [PMID: 22639704 PMCID: PMC3359644 DOI: 10.1021/cn2001206] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 12/19/2011] [Indexed: 12/20/2022] Open
Abstract
A summary of the initial discovery and characterization of the enzyme fatty acid amide hydrolase (FAAH), and the subsequent advancement of an important class of competitive, reversible, potent and selective inhibitors is presented. Initially explored using substrate-inspired inhibitors bearing electrophilic carbonyls, the examination of α-ketoheterocyle-based inhibitors of FAAH with the benefit of a unique activity-based protein-profiling (ABPP)-based proteome-wide selectivity assay, a powerful in vivo biomarker-based in vivo screen, and subsequent retrospective X-ray co-crystal structures with the enzyme, is summarized. These efforts defined the impact of the central activating heterocycle and its key substituents, provided key simplifications in the C2 acyl side chain and clear interpretations for the unique role and subsequent optimization of the central activating heterocycle, and established the basis for the recent further conformational constraints in the C2 acyl side chain, providing potent, long-acting, orally-active FAAH inhibitors.
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry and the Skaggs
Institute for
Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| | - Dale L. Boger
- Department of Chemistry and the Skaggs
Institute for
Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United
States
| |
Collapse
|
18
|
Discovery of inhibitors and substrates of brassinin hydrolase: Probing selectivity with dithiocarbamate bioisosteres. Bioorg Med Chem 2012; 20:225-33. [DOI: 10.1016/j.bmc.2011.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 11/03/2011] [Accepted: 11/05/2011] [Indexed: 11/19/2022]
|
19
|
Fatty acid amide hydrolase inhibitors. 3: Tetra-substituted azetidine ureas with in vivo activity. Bioorg Med Chem Lett 2012; 22:901-6. [DOI: 10.1016/j.bmcl.2011.12.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 12/05/2011] [Accepted: 12/06/2011] [Indexed: 12/22/2022]
|
20
|
Bowman AL, Makriyannis A. Approximating protein flexibility through dynamic pharmacophore models: application to fatty acid amide hydrolase (FAAH). J Chem Inf Model 2011; 51:3247-53. [PMID: 22098169 DOI: 10.1021/ci200371z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A structure-based drug discovery method is described that incorporates target flexibility through the use of an ensemble of protein conformations. The approach was applied to fatty acid amide hydrolase (FAAH), a key deactivating enzyme in the endocannabinoid system. The resultant dynamic pharmacophore models are rapidly able to identify known FAAH inhibitors over drug-like decoys. Different sources of FAAH conformational ensembles were explored, with both snapshots from molecular dynamics simulations and a group of X-ray structures performing well. Results were compared to those from docking and pharmacophore models generated from a single X-ray structure. Increasing conformational sampling consistently improved the pharmacophore models, emphasizing the importance of incorporating target flexibility in structure-based drug design.
Collapse
Affiliation(s)
- Anna L Bowman
- Center for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
21
|
Rusch M, Zahov S, Vetter IR, Lehr M, Hedberg C. Design, synthesis and evaluation of polar head group containing 2-keto-oxazole inhibitors of FAAH. Bioorg Med Chem 2011; 20:1100-12. [PMID: 22196515 DOI: 10.1016/j.bmc.2011.11.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/08/2011] [Accepted: 11/11/2011] [Indexed: 11/30/2022]
Abstract
2-α-Keto oxazoles containing polar head groups in their C5-side chains were designed as fatty acid amide hydrolase (FAAH) inhibitors. Variation in the spacer length resulted in submicromolar α-keto-oxazole FAAH inhibitor (IC(50)=436 nM) presenting electrostatic stabilizing interactions between its polar head group contained in the C5-side chain and the hydrophilic pocket of the enzyme.
Collapse
Affiliation(s)
- Marion Rusch
- Department of Chemical Biology, Max-Planck Institute of Molecular Physiology, Otto-Hahn Strasse 11, D-44227 Dortmund, Germany
| | | | | | | | | |
Collapse
|
22
|
Feledziak M, Muccioli GG, Lambert DM, Marchand-Brynaert J. SAR and LC/MS Studies of β-Lactamic Inhibitors of Human Fatty Acid Amide Hydrolase (hFAAH): Evidence of a Nonhydrolytic Process. J Med Chem 2011; 54:6812-23. [DOI: 10.1021/jm200723m] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marion Feledziak
- Laboratoire de Chimie Organique et Médicinale, Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, Bâtiment Lavoisier, Place Louis Pasteur L4.01.02, B-1348 Louvain-La-Neuve, Belgium
- Unité de Chimie Pharmaceutique et de Radiopharmacie, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier 73.40, B-1200 Bruxelles, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Laboratory, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier 72, B1.72.01, B-1200 Bruxelles, Belgium
| | - Didier M. Lambert
- Unité de Chimie Pharmaceutique et de Radiopharmacie, Louvain Drug Research Institute, Université catholique de Louvain, Avenue E. Mounier 73.40, B-1200 Bruxelles, Belgium
| | - Jacqueline Marchand-Brynaert
- Laboratoire de Chimie Organique et Médicinale, Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, Bâtiment Lavoisier, Place Louis Pasteur L4.01.02, B-1348 Louvain-La-Neuve, Belgium
| |
Collapse
|
23
|
Otrubova K, Ezzili C, Boger DL. The discovery and development of inhibitors of fatty acid amide hydrolase (FAAH). Bioorg Med Chem Lett 2011; 21:4674-85. [PMID: 21764305 PMCID: PMC3146581 DOI: 10.1016/j.bmcl.2011.06.096] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2011] [Revised: 06/18/2011] [Accepted: 06/20/2011] [Indexed: 11/16/2022]
Abstract
A summary of the discovery and advancement of inhibitors of fatty acid amide hydrolase (FAAH) is presented.
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey, Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
24
|
Vacondio F, Silva C, Lodola A, Carmi C, Rivara S, Duranti A, Tontini A, Sanchini S, Clapper JR, Piomelli D, Tarzia G, Mor M. Biphenyl-3-yl alkylcarbamates as fatty acid amide hydrolase (FAAH) inhibitors: steric effects of N-alkyl chain on rat plasma and liver stability. Eur J Med Chem 2011; 46:4466-73. [PMID: 21820769 DOI: 10.1016/j.ejmech.2011.07.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 07/12/2011] [Accepted: 07/12/2011] [Indexed: 12/27/2022]
Abstract
Secondary alkylcarbamic acid biphenyl-3-yl esters are a class of Fatty Acid Amide Hydrolase (FAAH) inhibitors, which include the reference compounds URB597 and URB694. Given the intrinsic reactivity of the carbamate group, the in vivo potency of these molecules in rats is strongly affected by their hydrolysis in plasma or hepatic metabolism. In the present study, in vitro chemical and metabolic stability assays (rat plasma and rat liver S(9) fraction) were used to investigate the structure-property relationships (SPRs) for a focused series of title compounds, where lipophilicity and steric hindrance of the carbamate N-substituent had been modulated. The resulting degradation rates indicate that a secondary or tertiary alkyl group at the carbamate nitrogen atom increases hydrolytic stability towards rat plasma esterases. The calculated solvent accessible surface area (SASA) of the carbamate fragment was employed to describe the differences observed in rate constants of hydrolysis in rat plasma (log k(plasma)), suggesting that stability in plasma increases if the substituent exerts a shielding effect on the carbamate carbonyl. Stability in rat liver S(9) fraction is increased when a tertiary carbon is bound to the carbamate nitrogen atom, while other steric effects showed complex relationships with degradation rates. The SPRs here described may be applied at the pharmacokinetic optimization of other classes of carbamate FAAH inhibitors.
Collapse
Affiliation(s)
- Federica Vacondio
- Dipartimento Farmaceutico, Università degli Studi di Parma, Viale GP Usberti 27/A, I-43124 Parma, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Mileni M, Garfunkle J, Ezzili C, Cravatt BF, Stevens RC, Boger DL. Fluoride-mediated capture of a noncovalent bound state of a reversible covalent enzyme inhibitor: X-ray crystallographic analysis of an exceptionally potent α-ketoheterocycle inhibitor of fatty acid amide hydrolase. J Am Chem Soc 2011; 133:4092-100. [PMID: 21355555 PMCID: PMC3060301 DOI: 10.1021/ja110877y] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two cocrystal X-ray structures of the exceptionally potent α-ketoheterocycle inhibitor 1 (K(i) = 290 pM) bound to a humanized variant of rat fatty acid amide hydrolase (FAAH) are disclosed, representing noncovalently and covalently bound states of the same inhibitor with the enzyme. Key to securing the structure of the noncovalently bound state of the inhibitor was the inclusion of fluoride ion in the crystallization conditions that is proposed to bind the oxyanion hole precluding inhibitor covalent adduct formation with stabilization of the tetrahedral hemiketal. This permitted the opportunity to detect important noncovalent interactions stabilizing the binding of the inhibitor within the FAAH active site independent of the covalent reaction. Remarkably, noncovalently bound 1 in the presence of fluoride appears to capture the active site in the same "in action" state with the three catalytic residues Ser241-Ser217-Lys142 occupying essentially identical positions observed in the covalently bound structure of 1, suggesting that this technique of introducing fluoride may have important applications in structural studies beyond inhibiting substrate or inhibitor oxyanion hole binding. Key insights to emerge from the studies include the observations that noncovalently bound 1 binds in its ketone (not gem diol) form, that the terminal phenyl group in the acyl side chain of the inhibitor serves as the key anchoring interaction overriding the intricate polar interactions in the cytosolic port, and that the role of the central activating heterocycle is dominated by its intrinsic electron-withdrawing properties. These two structures are also briefly compared with five X-ray structures of α-ketoheterocycle-based inhibitors bound to FAAH recently disclosed.
Collapse
Affiliation(s)
- Mauro Mileni
- Department of Molecular Biology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Joie Garfunkle
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Cyrine Ezzili
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Benjamin F. Cravatt
- Department of Chemical Physiology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Raymond C. Stevens
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- Department of Molecular Biology, The Scripps Research
Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| | - Dale L. Boger
- Department of Chemistry, The Scripps Research Institute,
10550 North Torrey Pines Road, La Jolla, California 92037
- The Skaggs Institute for Chemical Biology, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037
| |
Collapse
|
26
|
Zahov S, Drews A, Hess M, Schulze Elfringhoff A, Lehr M. 1‐(3‐Biaryloxy‐2‐oxopropyl)indole‐5‐carboxylic Acids and Related Compounds as Dual Inhibitors of Human Cytosolic Phospholipase A
2
α and Fatty Acid Amide Hydrolase. ChemMedChem 2011; 6:544-9. [DOI: 10.1002/cmdc.201000473] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 11/26/2010] [Indexed: 12/20/2022]
Affiliation(s)
- Stefan Zahov
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Hittorfstrasse 58‐62, 48149 Münster (Germany), Fax: (+49) 251‐83‐32144
| | - Andreas Drews
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Hittorfstrasse 58‐62, 48149 Münster (Germany), Fax: (+49) 251‐83‐32144
| | - Mark Hess
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Hittorfstrasse 58‐62, 48149 Münster (Germany), Fax: (+49) 251‐83‐32144
| | - Alwine Schulze Elfringhoff
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Hittorfstrasse 58‐62, 48149 Münster (Germany), Fax: (+49) 251‐83‐32144
| | - Matthias Lehr
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Hittorfstrasse 58‐62, 48149 Münster (Germany), Fax: (+49) 251‐83‐32144
| |
Collapse
|
27
|
Kiss LE, Ferreira HS, Beliaev A, Torrão L, Bonifácio MJ, Learmonth DA. Design, synthesis, and structure–activity relationships of 1,3,4-oxadiazol-2(3H)-ones as novel FAAH inhibitors. MEDCHEMCOMM 2011. [DOI: 10.1039/c1md00136a] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|