151
|
McReynolds C, Morisseau C, Wagner K, Hammock B. Epoxy Fatty Acids Are Promising Targets for Treatment of Pain, Cardiovascular Disease and Other Indications Characterized by Mitochondrial Dysfunction, Endoplasmic Stress and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1274:71-99. [PMID: 32894508 PMCID: PMC7737916 DOI: 10.1007/978-3-030-50621-6_5] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bioactive lipid mediators resulting from the metabolism of polyunsaturated fatty acids (PUFA) are controlled by many pathways that regulate the levels of these mediators and maintain homeostasis to prevent disease. PUFA metabolism is driven primarily through three pathways. Two pathways, the cyclooxygenase (COX) and lipoxygenase (LO) enzymatic pathways, form metabolites that are mostly inflammatory, while the third route of metabolism results from the oxidation by the cytochrome P450 enzymes to form hydroxylated PUFA and epoxide metabolites. These epoxygenated fatty acids (EpFA) demonstrate largely anti-inflammatory and beneficial properties, in contrast to the other metabolites formed from the degradation of PUFA. Dysregulation of these systems often leads to chronic disease. Pharmaceutical targets of disease focus on preventing the formation of inflammatory metabolites from the COX and LO pathways, while maintaining the EpFA and increasing their concentration in the body is seen as beneficial to treating and preventing disease. The soluble epoxide hydrolase (sEH) is the major route of metabolism of EpFA. Inhibiting its activity increases concentrations of beneficial EpFA, and often disease states correlate to mutations in the sEH enzyme that increase its activity and decrease the concentrations of EpFA in the body. Recent approaches to increasing EpFA include synthetic mimics that replicate biological activity of EpFA while preventing their metabolism, while other approaches focus on developing small molecule inhibitors to the sEH. Increasing EpFA concentrations in the body has demonstrated multiple beneficial effects in treating many diseases, including inflammatory and painful conditions, cardiovascular disease, neurological and disease of the central nervous system. Demonstration of efficacy in so many disease states can be explained by the fundamental mechanism that EpFA have of maintaining healthy microvasculature and preventing mitochondrial and endoplasmic reticulum stress. While there are no FDA approved methods that target the sEH or other enzymes responsible for metabolizing EpFA, current clinical efforts to test for efficacy by increasing EpFA that include inhibiting the sEH or administration of EpFA mimics that block metabolism are in progress.
Collapse
Affiliation(s)
- Cindy McReynolds
- Department of Entomology and Nematology, and U.C. Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
- EicOsis, Davis, CA, USA
| | - Christophe Morisseau
- Department of Entomology and Nematology, and U.C. Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
| | - Karen Wagner
- Department of Entomology and Nematology, and U.C. Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA
- EicOsis, Davis, CA, USA
| | - Bruce Hammock
- Department of Entomology and Nematology, and U.C. Davis Comprehensive Cancer Center, University of California Davis, Davis, CA, USA.
| |
Collapse
|
152
|
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: 64] [Impact Index Per Article: 10.7] [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
|
153
|
Koenders SA, Wijaya LS, Erkelens MN, Bakker AT, van der Noord VE, van Rooden EJ, Burggraaff L, Putter PC, Botter E, Wals K, van den Elst H, den Dulk H, Florea BI, van de Water B, van Westen GJP, Mebius RE, Overkleeft HS, Le Dévédec SE, van der Stelt M. Development of a Retinal-Based Probe for the Profiling of Retinaldehyde Dehydrogenases in Cancer Cells. ACS CENTRAL SCIENCE 2019; 5:1965-1974. [PMID: 31893226 PMCID: PMC6936097 DOI: 10.1021/acscentsci.9b01022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Indexed: 05/13/2023]
Abstract
Retinaldehyde dehydrogenases belong to a superfamily of enzymes that regulate cell differentiation and are responsible for detoxification of anticancer drugs. Chemical tools and methods are of great utility to visualize and quantify aldehyde dehydrogenase (ALDH) activity in health and disease. Here, we present the discovery of a first-in-class chemical probe based on retinal, the endogenous substrate of retinal ALDHs. We unveil the utility of this probe in quantitating ALDH isozyme activity in a panel of cancer cells via both fluorescence and chemical proteomic approaches. We demonstrate that our probe is superior to the widely used ALDEFLUOR assay to explain the ability of breast cancer (stem) cells to produce all-trans retinoic acid. Furthermore, our probe revealed the cellular selectivity profile of an advanced ALDH1A1 inhibitor, thereby prompting us to investigate the nature of its cytotoxicity. Our results showcase the application of substrate-based probes in interrogating pathologically relevant enzyme activities. They also highlight the general power of chemical proteomics in driving the discovery of new biological insights and its utility to guide drug discovery efforts.
Collapse
Affiliation(s)
- Sebastiaan
T. A. Koenders
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
| | - Lukas S. Wijaya
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Martje N. Erkelens
- Department
of Molecular Cell Biology and Immunology, Amsterdam University Medical Centra, Amsterdam 1081 HV, The Netherlands
| | - Alexander T. Bakker
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Vera E. van der Noord
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Eva J. van Rooden
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Lindsey Burggraaff
- Computational
Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic
Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Pasquale C. Putter
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Else Botter
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Kim Wals
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
| | - Hans van den Elst
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Hans den Dulk
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Bogdan I. Florea
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Bob van de Water
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Gerard J. P. van Westen
- Computational
Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic
Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Reina E. Mebius
- Department
of Molecular Cell Biology and Immunology, Amsterdam University Medical Centra, Amsterdam 1081 HV, The Netherlands
| | - Herman S. Overkleeft
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
| | - Sylvia E. Le Dévédec
- Cancer
Therapeutics and Drug Safety, Division of Drug Discovery and Safety,
Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Mario van der Stelt
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden 2300 RA, The Netherlands
- Oncode Institute, Utrecht 3521 AL, The Netherlands
- E-mail:
| |
Collapse
|
154
|
Hardisty JF, Harris SB, Hayes WA, Weber K. Oral repeated-dose toxicity studies of BIA 10-2474 in beagle dogs. Regul Toxicol Pharmacol 2019; 111:104555. [PMID: 31874201 DOI: 10.1016/j.yrtph.2019.104555] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 01/04/2023]
Abstract
BIA 10-2474 is a novel fatty acid amide hydrolase inhibitor developed for the treatment of medical conditions which would benefit from enhanced levels of endogenous anandamide (AEA) such as pain disorders. During a Phase I clinical trial one subject died after receiving BIA 10-2474 and others displayed neurological signs. We describe here the toxicology studies in beagle dogs that supported phase I testing of BIA 10-2474 in humans. A Maximum Tolerated Dose (MTD) study using once-a-day oral (capsule) application of BIA 10-2474 was first conducted to establish suitable dose levels for subsequent studies. Based on these results, 100 mg/kg/day was considered to be the MTD. The 4-week oral (capsule) toxicity study with a 3-week recovery period for BIA 10-2474 was therefore carried out at 20, 50 or 100 mg/kg/day. There were no changes recorded at 50 mg/kg/day and this was considered the oral No Observed Effect Level (NOEL) for four-week once-a-day capsule administration to Beagle dogs. At 100 mg/kg/day, the dose-limiting findings consisted of clinical symptoms including tremor, loss of balance, abnormal gait, decreased motor activity, weakness, vomits, salivation increase and miosis, increased severity of thymic atrophy/involution, and moderate acute, focal/multifocal bronchopneumonia in lungs of three animals. In a 13-week oral (capsule) toxicity study in the Beagle dog with a 6-week recovery period, using the same dose levels, clinical signs were recorded during treatment with BIA 10-274 at 50 and 100 mg/kg/day. The most frequent signs included difficulty breathing, respiratory sounds (with or without auscultation) and cough. Incoordination of the hind limbs with absence of correction reflex were also observed on some occasions. As a result, the 50 and 100 mg/kg/day doses were reduced to 35 and 50 mg/kg/day respectively on day 37. Because of the continued signs, the doses in both groups were further reduced to 20 mg/kg/day from day 77. Under the conditions of this study and given the severe signs recorded in groups treated at 100-50-20 and 50-35-20 mg/kg/day and only very occasional presence of signs in the group treated for the 13-week period at 20 mg/kg/day (abnormal respiratory sounds once in two animals), the dose of 20 mg/kg/day was considered the No Observed Adverse Effect Level (NOAEL).
Collapse
Affiliation(s)
| | | | - Wallace A Hayes
- University of South Florida College and Michigan State University, USA
| | - Klaus Weber
- AnaPath GmbH, 4625, Oberbuchsiten, Switzerland.
| |
Collapse
|
155
|
The absence of genotoxicity of a novel fatty acid amide hydrolase inhibitor, BIA 10-2474. Regul Toxicol Pharmacol 2019; 111:104556. [PMID: 31866344 DOI: 10.1016/j.yrtph.2019.104556] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
In 2016 one person died and others had neurological sequelae during a clinical trial with BIA 10-2474 (3-(1-(cyclohexyl(methyl)carbamoyl)-lH-imidazol-4-yl)pyridine 1-oxide), a novel fatty acid amide hydrolase (FAAH) inhibitor being developed for the treatment of medical conditions such as pain. Prior to the clinical trial a full battery of regulatory toxicology tests were carried out and this paper describes the genotoxicity/mutagenicity tests undertaken with BIA 10-2474 using the Ames (Salmonella typhimurium) reverse mutation test, the Escherichia coli WP2uvrA forward mutation test, an in vitro chromosome damage assay in human lymphocytes, and an in vivo micronucleus test in mice. All tests were conducted with and without a rat liver S9 metabolic activation system. None of the test results were judged to be positive with regards to the mutagenicity/genotoxicity of BIA 10-2474 making it unlikely that any such effect was involved in the toxicity observed in the clinic.
Collapse
|
156
|
Zhang X, Wang Q, Li Y, Ruan C, Wang S, Hu L, Ye M. Solvent-Induced Protein Precipitation for Drug Target Discovery on the Proteomic Scale. Anal Chem 2019; 92:1363-1371. [PMID: 31794197 DOI: 10.1021/acs.analchem.9b04531] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
High-throughput drug discovery is highly dependent on the targets available to accelerate the process of candidates screening. Traditional chemical proteomics approaches for the screening of drug targets usually require the immobilization/modification of the drug molecules to pull down the interacting proteins. Recently, energetics-based proteomics methods provide an alternative way to study drug-protein interaction by using complex cell lysate directly without any modification of the drugs. In this study, we developed a novel energetics-based proteomics strategy, the solvent-induced protein precipitation (SIP) approach, to profile the interaction of drugs with their target proteins by using quantitative proteomics. The method is easy to use for any laboratory with the common chemical reagents of acetone, ethanol, and acetic acid. The SIP approach was able to identify the well-known protein targets of methotrexate, SNS-032, and a pan-kinase inhibitor of staurosporine in cell lysate. We further applied this approach to discover the off-targets of geldanamycin. Three known protein targets of the HSP90 family were successfully identified, and several potential off-targets including NADH dehydrogenase subunits NDUFV1 and NDUFAB1 were identified for the first time, and the NDUFV1 was validated by using Western blotting. In addition, this approach was capable of evaluating the affinity of the drug-target interaction. The data collectively proved that our approach provides a powerful platform for drug target discovery.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, National Engineering Laboratory of AIDS Vaccine, School of Life Sciences , Jilin University , Changchun 130012 , China.,CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Qi Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Yanan Li
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Chengfei Ruan
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Shuyue Wang
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, National Engineering Laboratory of AIDS Vaccine, School of Life Sciences , Jilin University , Changchun 130012 , China.,CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| | - Lianghai Hu
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, National Engineering Laboratory of AIDS Vaccine, School of Life Sciences , Jilin University , Changchun 130012 , China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, National Chromatographic R&A Center , Dalian Institute of Chemical Physics, Chinese Academy of Sciences , Dalian 116023 , China
| |
Collapse
|
157
|
Bozkurt TE. Endocannabinoid System in the Airways. Molecules 2019; 24:E4626. [PMID: 31861200 PMCID: PMC6943521 DOI: 10.3390/molecules24244626] [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: 11/13/2019] [Revised: 12/12/2019] [Accepted: 12/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cannabinoids and the mammalian endocannabinoid system is an important research area of interest and attracted many researchers because of their widespread biological effects. The significant immune-modulatory role of cannabinoids has suggested their therapeutic use in several inflammatory conditions. Airways are prone to environmental irritants and stimulants, and increased inflammation is an important process in most of the respiratory diseases. Therefore, the main strategies for treating airway diseases are suppression of inflammation and producing bronchodilation. The ability of cannabinoids to induce bronchodilation and modify inflammation indicates their importance for airway physiology and pathologies. In this review, the contribution of cannabinoids and the endocannabinoid system in the airways are discussed, and the existing data for their therapeutic use in airway diseases are presented.
Collapse
Affiliation(s)
- Turgut Emrah Bozkurt
- Department of Pharmacology, Faculty of Pharmacy, Hacettepe University, Ankara 06100, Turkey
| |
Collapse
|
158
|
van Esbroeck ACM, Kantae V, Di X, van der Wel T, den Dulk H, Stevens AF, Singh S, Bakker AT, Florea BI, Stella N, Overkleeft HS, Hankemeier T, van der Stelt M. Identification of α,β-Hydrolase Domain Containing Protein 6 as a Diacylglycerol Lipase in Neuro-2a Cells. Front Mol Neurosci 2019; 12:286. [PMID: 31849602 PMCID: PMC6901982 DOI: 10.3389/fnmol.2019.00286] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022] Open
Abstract
The endocannabinoid 2-arachidonoylglycerol (2-AG) is involved in neuronal differentiation. This study aimed to identify the biosynthetic enzymes responsible for 2-AG production during retinoic acid (RA)-induced neurite outgrowth of Neuro-2a cells. First, we confirmed that RA stimulation of Neuro-2a cells increases 2-AG production and neurite outgrowth. The diacylglycerol lipase (DAGL) inhibitor DH376 blocked 2-AG production and reduced neuronal differentiation. Surprisingly, CRISPR/Cas9-mediated knockdown of DAGLα and DAGLβ in Neuro-2a cells did not reduce 2-AG levels, suggesting another enzyme capable of producing 2-AG in this cell line. Chemical proteomics revealed DAGLβ and α,β-hydrolase domain containing protein (ABHD6) as the only targets of DH376 in Neuro-2a cells. Biochemical, genetic and lipidomic studies demonstrated that ABHD6 possesses DAGL activity in conjunction with its previously reported monoacylglycerol lipase activity. RA treatment of Neuro-2a cells increased by three-fold the amount of active ABHD6. Our study shows that ABHD6 exhibits significant DAG lipase activity in Neuro-2a cells in addition to its known MAG lipase activity and suggest it is involved in neuronal differentiation.
Collapse
Affiliation(s)
- Annelot C M van Esbroeck
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Vasudev Kantae
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands.,Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Xinyu Di
- Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Tom van der Wel
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Hans den Dulk
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Anna F Stevens
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Simar Singh
- Department of Pharmacology, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Alexander T Bakker
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Bogdan I Florea
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Nephi Stella
- Department of Pharmacology, University of Washington, Seattle, WA, United States.,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
| | - Herman S Overkleeft
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Thomas Hankemeier
- Department of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| |
Collapse
|
159
|
Harris SB, Hardisty JF, Hayes AW, Weber K. Developmental and reproductive toxicity studies of BIA 10-2474. Regul Toxicol Pharmacol 2019; 111:104543. [PMID: 31759137 DOI: 10.1016/j.yrtph.2019.104543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/22/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
A series of regulatory studies were carried out to investigate the effects of the FAAH inhibitor BIA 10-2474 on fertility, embryo-fetal toxicity and pre- and post-natal development in rats and rabbits. Despite some reductions in sperm count in rats from 50 mg/kg, there were no major changes in male fertility up to 100 mg/kg. In female rats administered up to GD6, there were increases in pre-implantation loss at 50 and 100 mg/kg but neither post-implantation loss nor early embryonic development was affected. In contrast, when administered to female rats during pregnancy (GD6-GD17), BIA 10-2474 at 75 mg/kg/day reduced food consumption resulting in weight loss, increased post-implantation loss and reduced mean fetal body weight. In rabbits, the same maternal toxicity was seen but there were no effects in this species on post-implantation loss or fetal body weights. There were no teratological effects clearly due to BIA 10-2474 and developmental milestones and behavior of offspring were not affected. When administered during pregnancy and lactation (GD6-PND20), some post-implantation loss was seen from 20 mg/kg/day, but developmental milestones and behavior of the offspring were not affected, although males tended to have lower body weight. Based on these data the NOAEL for parental fertility was established as 50 mg/kg/day, the maternal NOAEL during pregnancy was 25 mg/kg/day in rats and developmental NOAEL was 25 and 75 mg/kg/day in rats and rabbits, respectively. When administered during post-natal development to rats the maternal NOAEL was 6 mg/kg/day. The parental reproductive NOAEL, the NOAEL for viability and growth of the F1 offspring, the F1 parental NOAEL and the F1 reproductive NOAEL were all considered to be 20 mg/kg/day.
Collapse
Affiliation(s)
| | - Jerry F Hardisty
- Experimental Pathology Laboratories, Inc, PO Box 12766, Research Triangle Park, NC, USA
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA.
| | - Klaus Weber
- AnaPath GmbH, 4625, Oberbuchsiten, Switzerland
| |
Collapse
|
160
|
Machine learning for target discovery in drug development. Curr Opin Chem Biol 2019; 56:16-22. [PMID: 31734566 DOI: 10.1016/j.cbpa.2019.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 12/15/2022]
Abstract
The discovery of macromolecular targets for bioactive agents is currently a bottleneck for the informed design of chemical probes and drug leads. Typically, activity profiling against genetically manipulated cell lines or chemical proteomics is pursued to shed light on their biology and deconvolute drug-target networks. By taking advantage of the ever-growing wealth of publicly available bioactivity data, learning algorithms now provide an attractive means to generate statistically motivated research hypotheses and thereby prioritize biochemical screens. Here, we highlight recent successes in machine intelligence for target identification and discuss challenges and opportunities for drug discovery.
Collapse
|
161
|
Cramer S, Johnson J, Ngo T, El‐Alfy AT, Stec J. Modulation of the Endocannabinoid System via Inhibition of Fatty Acid Amide Hydrolase (FAAH) by Novel Urea and Carbamate Derivatives. ChemistrySelect 2019. [DOI: 10.1002/slct.201903375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Sarah Cramer
- Chicago State UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 9501 S. King Drive, Chicago Illinois 60628 United States
| | - Jacklyn Johnson
- Chicago State UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 9501 S. King Drive, Chicago Illinois 60628 United States
| | - Thanh Ngo
- Chicago State UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 9501 S. King Drive, Chicago Illinois 60628 United States
| | - Abir T. El‐Alfy
- Chicago State UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 9501 S. King Drive, Chicago Illinois 60628 United States
- Medical College of Wisconsin School of PharmacyDepartment of Biopharmaceutical Sciences 8701 Watertown Plank Rd, Milwaukee Wisconsin 53226 United States
| | - Jozef Stec
- Chicago State UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 9501 S. King Drive, Chicago Illinois 60628 United States
- Marshall B. Ketchum UniversityCollege of PharmacyDepartment of Pharmaceutical Sciences 2575 Yorba Linda Blvd., Fullerton California 82831 United States
| |
Collapse
|
162
|
Baggelaar MP, den Dulk H, Florea BI, Fazio D, Bernabò N, Raspa M, Janssen APA, Scavizzi F, Barboni B, Overkleeft HS, Maccarrone M, van der Stelt M. ABHD2 Inhibitor Identified by Activity-Based Protein Profiling Reduces Acrosome Reaction. ACS Chem Biol 2019; 14:2295-2304. [PMID: 31525885 PMCID: PMC6878212 DOI: 10.1021/acschembio.9b00640] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
ABHD2 is a serine hydrolase that belongs to the subgroup of the α,β-hydrolase fold-containing proteins, which is involved in virus propagation, immune response, and fertilization. Chemical tools to selectively modulate the activity of ABHD2 in an acute setting are highly desired to investigate its biological role, but are currently lacking. Here, we report a library-versus-library screening using activity-based protein profiling (ABPP) to evaluate in parallel the selectivity and activity of a focused lipase inhibitor library against ABHD2 and a panel of closely related ABHD proteins. This screen resulted in the rapid identification of novel inhibitors for ABHD2. The selectivity of the inhibitor was further investigated in native mouse testis proteome by competitive ABPP, revealing a highly restricted off-target profile. The progesterone-induced acrosome reaction was reduced in a dose-dependent manner by the newly identified inhibitor, which provides further support for the key-role of ABHD2 in the P4-stimulated acrosome reaction. On this basis, the ABHD2 inhibitor is an excellent starting point for further optimization of ABHD2 inhibitors that can modulate sperm fertility and may lead to novel contraceptives.
Collapse
Affiliation(s)
| | | | | | - Domenico Fazio
- Unit of Basic and Applied Biosciences, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Nicola Bernabò
- Unit of Basic and Applied Biosciences, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | - Marcello Raspa
- National Research Council (IBCN), CNR-Campus International Development (EMMA INFRAFRONTIER-IMPC), Via E. Ramarini 32, 00015 Monterotondo Scalo, Italy
| | | | - Ferdinando Scavizzi
- National Research Council (IBCN), CNR-Campus International Development (EMMA INFRAFRONTIER-IMPC), Via E. Ramarini 32, 00015 Monterotondo Scalo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, University of Teramo, Via Balzarini 1, 64100 Teramo, Italy
| | | | - Mauro Maccarrone
- Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
- European Centre for Brain Research/IRCCS Santa Lucia Foundation, via del Fosso del Fiorano 65, 00143 Rome, Italy
| | | |
Collapse
|
163
|
Greco R, Demartini C, Zanaboni AM, Tumelero E, Reggiani A, Misto A, Piomelli D, Tassorelli C. FAAH inhibition as a preventive treatment for migraine: A pre-clinical study. Neurobiol Dis 2019; 134:104624. [PMID: 31629892 DOI: 10.1016/j.nbd.2019.104624] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/02/2019] [Accepted: 09/23/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Fatty-acid amide hydrolase (FAAH) is an intracellular serine hydrolase that catalyzes the cleavage of endogenous fatty-acid amides, including the endocannabinoid anandamide (AEA). We previously reported that the peripherally restricted FAAH inhibitor URB937, which selectively increases AEA levels outside the central nervous system, reduces hyperalgesia and c-Fos expression in the trigeminal nucleus caudalis (TNC) and the locus coeruleus in an animal model of migraine based on nitroglycerin (NTG) administration. AIM To further investigate the relevance of FAAH inhibition in the NTG animal model of migraine by testing the effects of the globally active FAAH inhibitor URB597. METHODS Our experimental approach involved mapping neuronal c-Fos protein expression, measurement of AEA levels in brain areas and in trigeminal ganglia, evaluation of pain-related behavior and quantification of molecular mediators in rats that received URB597 (2 mg/kg i.p.) either before or after NTG administration (10 mg/kg, i.p.). RESULTS Pre-treatment with URB597 significantly reduced c-Fos immunoreactivity in the TNC and inhibited NTG-induced hyperalgesia in the orofacial formalin test. This behavioral response was associated with a decrease in neuronal nitric oxide synthase, calcitonin gene-related peptide and cytokine gene expression levels in central and peripheral structures. Administration of URB597 after NTG had no such effect. CONCLUSIONS The findings suggest that global FAAH inhibition may offer a therapeutic approach to the prevention, but not the abortive treatment, of migraine attacks. Further studies are needed to elucidate the exact cellular and molecular mechanisms underlying the protective effects of FAAH inhibition.
Collapse
Affiliation(s)
- Rosaria Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation Pavia, Italy.
| | - Chiara Demartini
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation Pavia, Italy
| | - Anna Maria Zanaboni
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Italy
| | - Elena Tumelero
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation Pavia, Italy
| | - Angelo Reggiani
- Dept. of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessandra Misto
- Dept. of Drug Discovery and Development, Istituto Italiano di Tecnologia, Genova, Italy
| | - Daniele Piomelli
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Cristina Tassorelli
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation Pavia, Italy; Department of Brain and Behavioral Sciences, University of Pavia, Italy
| |
Collapse
|
164
|
|
165
|
Abstract
Substance use disorder (SUD) is a major public health crisis worldwide, and effective treatment options are limited. During the past 2 decades, researchers have investigated the impact of a variety of pharmacological approaches to treat SUD, one of which is the use of medical cannabis or cannabinoids. Significant progress was made with the discovery of rimonabant, a selective CB1 receptor (CB1R) antagonist (also an inverse agonist), as a promising therapeutic for SUDs and obesity. However, serious adverse effects such as depression and suicidality led to the withdrawal of rimonabant (and almost all other CB1R antagonists/inverse agonists) from clinical trials worldwide in 2008. Since then, much research interest has shifted to other cannabinoid-based strategies, such as peripheral CB1R antagonists/inverse agonists, neutral CB1R antagonists, allosteric CB1R modulators, CB2R agonists, fatty acid amide hydrolase (FAAH) inhibitors, monoacylglycerol lipase (MAGL) inhibitors, fatty acid binding protein (FABP) inhibitors, or nonaddictive phytocannabinoids with CB1R or CB2R-binding profiles, as new therapeutics for SUDs. In this article, we first review recent progress in research regarding the endocannabinoid systems, cannabis reward versus aversion, and the underlying receptor mechanisms. We then review recent progress in cannabinoid-based medication development for the treatment of SUDs. As evidence continues to accumulate, neutral CB1R antagonists (such as AM4113), CB2R agonists (JWH133, Xie2-64), and nonselective phytocannabinoids (cannabidiol, β-caryophyllene, ∆9-tetrahydrocannabivarin) have shown great therapeutic potential for SUDs, as shown in experimental animals. Several cannabinoid-based medications (e.g., dronabinol, nabilone, PF-04457845) that entered clinical trials have shown promising results in reducing withdrawal symptoms in cannabis and opioid users.
Collapse
Affiliation(s)
- Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medication Discoveries Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| |
Collapse
|
166
|
Bedse G, Centanni SW, Winder DG, Patel S. Endocannabinoid Signaling in the Central Amygdala and Bed Nucleus of the Stria Terminalis: Implications for the Pathophysiology and Treatment of Alcohol Use Disorder. Alcohol Clin Exp Res 2019; 43:2014-2027. [PMID: 31373708 PMCID: PMC6779484 DOI: 10.1111/acer.14159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 12/23/2022]
Abstract
High rates of relapse are a chronic and debilitating obstacle to effective treatment of alcohol use disorder (AUD); however, no effective treatments are available to treat symptoms induced by protracted abstinence. In the first part of this 2-part review series, we examine the literature supporting the effects of alcohol exposure within the extended amygdala (EA) neural circuitry. In Part 2, we focus on a potential way to combat negative affect associated with AUD, by exploring the therapeutic potential of the endogenous cannabinoid (eCB) system. The eCB system is a potent modulator of neural activity in the brain, and its ability to mitigate stress and negative affect has long been an area of interest for developing novel therapeutics. This review details the recent advances in our understanding of eCB signaling in 2 key regions of the EA, the central nucleus of the amygdala and the bed nucleus of the stria terminalis (BNST), and their role in regulating negative affect. Despite an established role for EA eCB signaling in reducing negative affect, few studies have examined the potential for eCB-based therapies to treat AUD-associated negative affect. In this review, we present an overview of studies focusing on eCB signaling in EA and cannabinoid modulation on EA synaptic activity. We further discuss studies suggesting dysregulation of eCB signaling in models of AUD and propose that pharmacological augmentation of eCB could be a novel approach to treat aspects of AUD. Lastly, future directions are proposed to advance our understanding of the relationship between AUD-associated negative affect and the EA eCB system that could yield new pharmacotherapies targeting negative affective symptoms associated with AUD.
Collapse
Affiliation(s)
- Gaurav Bedse
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Samuel W. Centanni
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| | - Sachin Patel
- Vanderbilt Center for Addiction Research, Nashville, TN, USA
- Molecular Physiology & Biophysics, the, Nashville, TN, USA
- Vanderbilt Brain Institute, Nashville, TN, USA
- Vanderbilt J.F. Kennedy Center for Research on Human Development, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Nashville, TN, USA
| |
Collapse
|
167
|
Wu MM, Zhang X, Asher MJ, Thayer SA. Druggable targets of the endocannabinoid system: Implications for the treatment of HIV-associated neurocognitive disorder. Brain Res 2019; 1724:146467. [PMID: 31539547 DOI: 10.1016/j.brainres.2019.146467] [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: 05/06/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/24/2022]
Abstract
HIV-associated neurocognitive disorder (HAND) affects nearly half of all HIV-infected individuals. Synaptodendritic damage correlates with neurocognitive decline in HAND, and many studies have demonstrated that HIV-induced neuronal injury results from excitotoxic and inflammatory mechanisms. The endocannabinoid (eCB) system provides on-demand protection against excitotoxicity and neuroinflammation. Here, we discuss evidence of the neuroprotective and anti-inflammatory properties of the eCB system from in vitro and in vivo studies. We examine the pharmacology of the eCB system and evaluate the therapeutic potential of drugs that modulate eCB signaling to treat HAND. Finally, we provide perspective on the need for additional studies to clarify the role of the eCB system in HIV neurotoxicity and speculate that strategies that enhance eCB signaling might slow cognitive decline in HAND.
Collapse
Affiliation(s)
- Mariah M Wu
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | - Xinwen Zhang
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | - Melissa J Asher
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| | - Stanley A Thayer
- Graduate Program in Neuroscience, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
168
|
Giacovazzo G, Bisogno T, Piscitelli F, Verde R, Oddi S, Maccarrone M, Coccurello R. Different Routes to Inhibit Fatty Acid Amide Hydrolase: Do All Roads Lead to the Same Place? Int J Mol Sci 2019; 20:ijms20184503. [PMID: 31514437 PMCID: PMC6771131 DOI: 10.3390/ijms20184503] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 11/25/2022] Open
Abstract
There is robust evidence indicating that enhancing the endocannabinoid (eCB) tone has therapeutic potential in several brain disorders. The inhibition of eCBs degradation by fatty acid amide hydrolase (FAAH) blockade, is the best-known option to increase N-acyl-ethanolamines-(NAEs)-mediated signaling. Here, we investigated the hypothesis that intranasal delivery is an effective route for different FAAH inhibitors, such as URB597 and PF-04457845. URB597 and PF-04457845 were subchronically administered in C57BL/6 male mice every other day for 20 days for overall 10 drug treatment, and compared for their ability to inhibit FAAH activity by the way of three different routes of administration: intranasal (i.n.), intraperitoneal (i.p.) and oral (p.o.). Lastly, we compared the efficacy of the three routes in terms of URB597-induced increase of NAEs levels in liver and in different brain areas. Results: We show that PF-04457845 potently inhibits FAAH regardless the route selected, and that URB597 was less effective in the brain after p.o. administration while reached similar effects by i.n. and i.p. routes. Intranasal URB597 delivery always increased NAEs levels in brain areas, whereas a parallel increase was not observed in the liver. By showing the efficacy of intranasal FAAH inhibition, we provide evidence that nose-to-brain delivery is a suitable alternative to enhance brain eCB tone for the treatment of neurodegenerative disorders and improve patients’ compliance.
Collapse
Affiliation(s)
- Giacomo Giacovazzo
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Tiziana Bisogno
- Endocannabinoid Research Group, Institute of Translational Pharmacology, CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Via C. Flegrei 34, 80078 Pozzuoli, Italy
| | - Roberta Verde
- Endocannabinoid Research Group, Institute of Biomolecular Chemistry, CNR, Via C. Flegrei 34, 80078 Pozzuoli, Italy
| | - Sergio Oddi
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via del Fosso di Fiorano 64, 00143 Rome, Italy
- Faculty of Veterinary Medicine, University of Teramo, via R. Balzarini 1, 64100 Teramo, Italy
| | - Mauro Maccarrone
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
- Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy.
| | - Roberto Coccurello
- Fondazione Santa Lucia IRCCS, Preclinical Neuroscience, Via del Fosso di Fiorano 64, 00143 Rome, Italy.
- Institute for Complex Systems (ISC), C.N.R., Via dei Taurini 19, 00185 Rome, Italy.
| |
Collapse
|
169
|
Janssen APA, van Hengst JMA, Béquignon OJM, Deng H, van Westen GJP, van der Stelt M. Structure Kinetics Relationships and Molecular Dynamics Show Crucial Role for Heterocycle Leaving Group in Irreversible Diacylglycerol Lipase Inhibitors. J Med Chem 2019; 62:7910-7922. [PMID: 31437392 PMCID: PMC6745892 DOI: 10.1021/acs.jmedchem.9b00686] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
![]()
Drug discovery programs of covalent
irreversible, mechanism-based
enzyme inhibitors often focus on optimization of potency as determined
by IC50-values in biochemical assays. These assays do not
allow the characterization of the binding activity (Ki) and reactivity (kinact)
as individual kinetic parameters of the covalent inhibitors. Here,
we report the development of a kinetic substrate assay to study the
influence of the acidity (pKa) of heterocyclic
leaving group of triazole urea derivatives as diacylglycerol lipase
(DAGL)-α inhibitors. Surprisingly, we found that the reactivity
of the inhibitors did not correlate with the pKa of the leaving group, whereas the position of the nitrogen
atoms in the heterocyclic core determined to a large extent the binding
activity of the inhibitor. This finding was confirmed and clarified
by molecular dynamics simulations on the covalently bound Michaelis–Menten
complex. A deeper understanding of the binding properties of covalent
serine hydrolase inhibitors is expected to aid in the discovery and
development of more selective covalent inhibitors.
Collapse
Affiliation(s)
- Antonius P A Janssen
- Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2300RA Leiden , The Netherlands
| | - Jacob M A van Hengst
- Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2300RA Leiden , The Netherlands
| | - Olivier J M Béquignon
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Hui Deng
- Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2300RA Leiden , The Netherlands
| | - Gerard J P van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research , Leiden University , P.O. Box 9502, 2300 RA Leiden , The Netherlands
| | - Mario van der Stelt
- Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2300RA Leiden , The Netherlands
| |
Collapse
|
170
|
Sartori SB, Singewald N. Novel pharmacological targets in drug development for the treatment of anxiety and anxiety-related disorders. Pharmacol Ther 2019; 204:107402. [PMID: 31470029 DOI: 10.1016/j.pharmthera.2019.107402] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 08/19/2019] [Indexed: 12/24/2022]
Abstract
Current medication for anxiety disorders is suboptimal in terms of efficiency and tolerability, highlighting the need for improved drug treatments. In this review an overview of drugs being studied in different phases of clinical trials for their potential in the treatment of fear-, anxiety- and trauma-related disorders is presented. One strategy followed in drug development is refining and improving compounds interacting with existing anxiolytic drug targets, such as serotonergic and prototypical GABAergic benzodiazepines. A more innovative approach involves the search for compounds with novel mechanisms of anxiolytic action using the growing knowledge base concerning the relevant neurocircuitries and neurobiological mechanisms underlying pathological fear and anxiety. The target systems evaluated in clinical trials include glutamate, endocannabinoid and neuropeptide systems, as well as ion channels and targets derived from phytochemicals. Examples of promising novel candidates currently in clinical development for generalised anxiety disorder, social anxiety disorder, panic disorder, obsessive compulsive disorder or post-traumatic stress disorder include ketamine, riluzole, xenon with one common pharmacological action of modulation of glutamatergic neurotransmission, as well as the neurosteroid aloradine. Finally, compounds such as D-cycloserine, MDMA, L-DOPA and cannabinoids have shown efficacy in enhancing fear-extinction learning in humans. They are thus investigated in clinical trials as an augmentative strategy for speeding up and enhancing the long-term effectiveness of exposure-based psychotherapy, which could render chronic anxiolytic drug treatment dispensable for many patients. These efforts are indicative of a rekindled interest and renewed optimism in the anxiety drug discovery field, after decades of relative stagnation.
Collapse
Affiliation(s)
- Simone B Sartori
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria
| | - Nicolas Singewald
- Institute of Pharmacy, Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck (CMBI), Leopold Franzens University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
171
|
Brunetti L, Loiodice F, Piemontese L, Tortorella P, Laghezza A. New Approaches to Cancer Therapy: Combining Fatty Acid Amide Hydrolase (FAAH) Inhibition with Peroxisome Proliferator-Activated Receptors (PPARs) Activation. J Med Chem 2019; 62:10995-11003. [PMID: 31407888 DOI: 10.1021/acs.jmedchem.9b00885] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Over the course of the past decade, peroxisome proliferator-activated receptors (PPARs) have been identified as part of the cannabinoid signaling system: both phytocannabinoids and endocannabinoids are capable of binding and activating these nuclear receptors. Fatty acid amide hydrolase (FAAH) hydrolyzes the endocannabinoid anandamide and other N-acylethanolamines. These substances have been shown to have numerous anticancer effects, and indeed the inhibition of FAAH has multiple beneficial effects that are mediated by PPARα subtype and by PPARγ subtype, especially antiproliferation and activation of apoptosis. The substrates of FAAH are also PPAR agonists, which explains the PPAR-mediated effects of FAAH inhibitors. Much like cannabinoid ligands and FAAH inhibitors, PPARγ agonists show antiproliferative effects on cancer cells, suggesting that additive or synergistic effects may be achieved through the positive modulation of both signaling systems. In this Miniperspective, we discuss the development of novel FAAH inhibitors able to directly act as PPAR agonists and their promising utilization as leads for the discovery of highly effective anticancer compounds.
Collapse
Affiliation(s)
- Leonardo Brunetti
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Luca Piemontese
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Paolo Tortorella
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze del Farmaco , Università degli Studi di Bari "Aldo Moro" , via Orabona 4 , 70125 Bari , Italy
| |
Collapse
|
172
|
Bell J. Aspirin killed the cat: animal research models do not always apply to humans. Expert Opin Drug Metab Toxicol 2019; 15:683-685. [DOI: 10.1080/17425255.2019.1652596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Jennifer Bell
- Independent biotechnology industry professional, Dundalk, Republic of Ireland
| |
Collapse
|
173
|
De Gregorio D, Manchia M, Carpiniello B, Valtorta F, Nobile M, Gobbi G, Comai S. Role of palmitoylethanolamide (PEA) in depression: Translational evidence: Special Section on "Translational and Neuroscience Studies in Affective Disorders". Section Editor, Maria Nobile MD, PhD. This Section of JAD focuses on the relevance of translational and neuroscience studies in providing a better understanding of the neural basis of affective disorders. The main aim is to briefly summaries relevant research findings in clinical neuroscience with particular regards to specific innovative topics in mood and anxiety disorders. J Affect Disord 2019; 255:S0165-0327(18)31599-4. [PMID: 30391203 DOI: 10.1016/j.jad.2018.10.117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/18/2018] [Accepted: 10/24/2018] [Indexed: 01/23/2023]
Abstract
BACKGROUND Antidepressants have a low rate of response paired with a delayed onset of action. Translational studies are thus seeking for novel targets for antidepressant drug development. Preclinical evidence has demonstrated that the endocannabinoid system plays an important role in mood and stress response, even if drugs targeting this system have not yet become available for clinical use. The dietary supplement N-Palmitoylethanolamide (PEA) is a fatty acid amide belonging to the endocannabinoid system with potential antidepressant properties. METHODS We performed a bibliographic search to review current knowledge on the potential antidepressant effects of PEA and its underlying mechanism of action. RESULTS PEA targets not only the peroxisome proliferator-activated receptor-alpha (PPAR-α), but also the endocannabinoid system, binding the G-protein-coupled receptor 55, a non-CB1/CB2 cannabinoid receptor, and also the CB1/CB2 receptors, although with a weak affinity. Preclinical studies have shown antidepressant activity of PEA in animal paradigms of depression and of depression associated with neuropathic pain and traumatic brain injury. In a translational perspective, PEA is increased in stress conditions, and a randomized, double-blind study in depressed patients indicated a fast-antidepressant action of PEA when associated with citalopram. LIMITATIONS There are still limited preclinical and clinical studies investigating the effect of PEA upon the endocannabinoid system and its potential as antidepressant. CONCLUSIONS PEA has potential antidepressant effects alone or in combinations with other classes of antidepressants. Future studies in depressed patients are needed to confirm the mood-modulating properties of PEA and its role as a biomarker of depression.
Collapse
Affiliation(s)
- Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Mirko Manchia
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | - Bernardo Carpiniello
- Section of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Flavia Valtorta
- San Raffaele Scientific Institute and Vita Salute University, Via Olgettina 58, Milano 20132, Italy
| | - Maria Nobile
- Scientific Institute, IRCCS E. Medea, Child Psychopathology Unit, Bosisio Parini, Lecco, Italy
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University Health Center, McGill University, Montreal, QC, Canada; San Raffaele Scientific Institute and Vita Salute University, Via Olgettina 58, Milano 20132, Italy.
| |
Collapse
|
174
|
Ney LJ, Matthews A, Bruno R, Felmingham KL. Cannabinoid interventions for PTSD: Where to next? Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:124-140. [PMID: 30946942 DOI: 10.1016/j.pnpbp.2019.03.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/20/2019] [Accepted: 03/29/2019] [Indexed: 01/18/2023]
Abstract
Cannabinoids are a promising method for pharmacological treatment of post-traumatic stress disorder (PTSD). Despite considerable research devoted to the effect of cannabinoid modulation on PTSD symptomology, there is not a currently agreed way by which the cannabinoid system should be targeted in humans. In this review, we present an overview of recent research identifying neurological pathways by which different cannabinoid-based treatments may exert their effects on PTSD symptomology. We evaluate the strengths and weaknesses of each of these different approaches, including recent challenges presented to favourable options such as fatty acid amide hydrolase (FAAH) inhibitors. This article makes the strengths and challenges of different potential cannabinoid treatments accessible to psychological researchers interested in cannabinoid therapeutics and aims to aid selection of appropriate tools for future clinical trials.
Collapse
Affiliation(s)
- Luke J Ney
- School of Psychology, University of Tasmania, Australia.
| | | | | | - Kim L Felmingham
- School of Psychological Sciences, University of Melbourne, Australia
| |
Collapse
|
175
|
Impaired anandamide/palmitoylethanolamide signaling in hippocampal glutamatergic neurons alters synaptic plasticity, learning, and emotional responses. Neuropsychopharmacology 2019; 44:1377-1388. [PMID: 30532004 PMCID: PMC6784910 DOI: 10.1038/s41386-018-0274-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 12/28/2022]
Abstract
Endocannabinoid signaling via anandamide (AEA) is implicated in a variety of neuronal functions and considered a promising therapeutic target for numerous emotion-related disorders. The major AEA degrading enzyme is fatty acid amide hydrolase (FAAH). Genetic deletion and pharmacological inhibition of FAAH reduce anxiety and improve emotional responses and memory in rodents and humans. Complementarily, the mechanisms and impact of decreased AEA signaling remain to be delineated in detail. In the present study, using the Cre/loxP system combined with an adeno-associated virus (AAV)-mediated delivery system, FAAH was selectively overexpressed in hippocampal CA1-CA3 glutamatergic neurons of adult mice. This approach led to specific FAAH overexpression at the postsynaptic site of CA1-CA3 neurons, to increased FAAH enzymatic activity, and, in consequence, to decreased hippocampal levels of AEA and palmitoylethanolamide (PEA), but the levels of the second major endocannabinoid 2-arachidonoyl glycerol (2-AG) and of oleoylethanolamide (OEA) were unchanged. Electrophysiological recordings revealed an enhancement of both excitatory and inhibitory synaptic activity and of long-term potentiation (LTP). In contrast, excitatory and inhibitory long-term depression (LTD) and short-term synaptic plasticity, apparent as depolarization-induced suppression of excitation (DSE) and inhibition (DSI), remained unaltered. These changes in hippocampal synaptic activity were associated with an increase in anxiety-like behavior, and a deficit in object recognition memory and in extinction of aversive memory. This study indicates that AEA is not involved in hippocampal short-term plasticity, or eLTD and iLTD, but modulates glutamatergic transmission most likely via presynaptic sites, and that disturbances in this process impair learning and emotional responses.
Collapse
|
176
|
Backus KM, Cao J, Maddox SM. Opportunities and challenges for the development of covalent chemical immunomodulators. Bioorg Med Chem 2019; 27:3421-3439. [PMID: 31204229 DOI: 10.1016/j.bmc.2019.05.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023]
Abstract
Compounds that react irreversibly with cysteines have reemerged as potent and selective tools for altering protein function, serving as chemical probes and even clinically approved drugs. The exquisite sensitivity of human immune cell signaling pathways to oxidative stress indicates the likely, yet still underexploited, general utility of covalent probes for selective chemical immunomodulation. Here, we provide an overview of immunomodulatory cysteines, including identification of electrophilic compounds available to label these residues. We focus our discussion on three protein classes essential for cell signaling, which span the 'druggability' spectrum from amenable to chemical probes (kinases), somewhat druggable (proteases), to inaccessible (phosphatases). Using existing inhibitors as a guide, we identify general strategies to guide the development of covalent probes for selected undruggable classes of proteins and propose the application of such compounds to alter immune cell functions.
Collapse
Affiliation(s)
- Keriann M Backus
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA.
| | - Jian Cao
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA
| | - Sean M Maddox
- Departments of Biological Chemistry and Chemistry and Biochemistry, University of California Los Angeles, USA
| |
Collapse
|
177
|
Response of safety pharmacologists to challenges arising from the rapidly evolving changes in the pharmaceutical industry. J Pharmacol Toxicol Methods 2019; 98:106593. [PMID: 31158459 DOI: 10.1016/j.vascn.2019.106593] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/14/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023]
Abstract
This commentary highlights and expands upon the thoughts conveyed in the lecture by Dr. Alan S. Bass, recipient of the 2017 Distinguished Service Award from the Safety Pharmacology Society, given on 27 September 2017 in Berlin, Germany. The lecture discussed the societal, scientific, technological, regulatory and economic events that dramatically impacted the pharmaceutical industry and ultimately led to significant changes in the strategic operations and practices of safety pharmacology. It focused on the emerging challenges and opportunities, and considered the lessons learned from drug failures and the influences of world events, including the financial crisis that ultimately led to a collapse of the world economies from which we are now recovering. Events such as these, which continue to today, challenge the assumptions that form the foundation of our discipline and dramatically affect the way that safety pharmacology is practiced. These include the latest scientific and technological developments contributing to the design and advancement of safe medicines. More broadly, they reflect the philosophical mission of safety pharmacology and the roles and responsibilities served by safety pharmacologists. As the discipline of Safety Pharmacology continues to evolve, develop and mature, the reader is invited to reflect on past experiences as a framework towards a vision of the future of the field.
Collapse
|
178
|
Tassorelli C, Greco R, Silberstein SD. The endocannabinoid system in migraine: from bench to pharmacy and back. Curr Opin Neurol 2019; 32:405-412. [DOI: 10.1097/wco.0000000000000688] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
179
|
Deplano A, Cipriano M, Moraca F, Novellino E, Catalanotti B, Fowler CJ, Onnis V. Benzylamides and piperazinoarylamides of ibuprofen as fatty acid amide hydrolase inhibitors. J Enzyme Inhib Med Chem 2019; 34:562-576. [PMID: 30688118 PMCID: PMC6352954 DOI: 10.1080/14756366.2018.1532418] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fatty Acid Amide Hydrolase (FAAH) is a serine hydrolase that plays a key role in controlling endogenous levels of endocannabinoids. FAAH inhibition is considered a powerful approach to enhance the endocannabinoid signalling, and therefore it has been largely studied as a potential target for the treatment of neurological disorders such as anxiety or depression, or of inflammatory processes. We present two novel series of amide derivatives of ibuprofen designed as analogues of our reference FAAH inhibitor Ibu-AM5 to further explore its structure-activity relationships. In the new amides, the 2-methylpyridine moiety of Ibu-AM5 was substituted by benzylamino and piperazinoaryl moieties. The obtained benzylamides and piperazinoarylamides showed FAAH inhibition ranging from the low to high micromolar potency. The binding of the new amides in the active site of FAAH, estimated using the induced fit protocol, indicated arylpiperazinoamides binding the ACB channel and the cytosolic port, and benzylamides binding the ACB channel.
Collapse
Affiliation(s)
- Alessandro Deplano
- a Department of Life and Environmental Sciences - Unit of Pharmaceutical, Pharmacological and Nutraceutical Sciences , University of Cagliari , Cagliari , Italy
| | - Mariateresa Cipriano
- b Department of Pharmacology and Clinical Neuroscience , Umeå University , Umeå , Sweden
| | - Federica Moraca
- c Department of Chemical Sciences , University of Napoli Federico II , Napoli , Italy
| | - Ettore Novellino
- d Department of Pharmacy , University of Napoli Federico II , Napoli , Italy
| | - Bruno Catalanotti
- d Department of Pharmacy , University of Napoli Federico II , Napoli , Italy
| | - Christopher J Fowler
- b Department of Pharmacology and Clinical Neuroscience , Umeå University , Umeå , Sweden
| | - Valentina Onnis
- a Department of Life and Environmental Sciences - Unit of Pharmaceutical, Pharmacological and Nutraceutical Sciences , University of Cagliari , Cagliari , Italy
| |
Collapse
|
180
|
Abstract
Novel pharmacological treatments are needed for Tourette syndrome. Our goal was to examine the current evidence base and biological rationale for the use of cannabis-derived medications or medications that act on the cannabinoid system in Tourette syndrome. We conducted a comprehensive literature search of PubMed for randomized controlled trials or clinical trials of cannabis-derived medications in Tourette syndrome. Data regarding the population, intervention, safety profile, and outcomes for each trial were extracted and reported and the evidence supporting use of individual cannabis-derived medications was critiqued. There is a strong biological rationale regarding how cannabis-derived medications could affect tic severity. Anecdotal case reports and series have noted that many patients report that their tics improve after using cannabis. However, only two small randomized, placebo-controlled trials of Δ9-tetrahydrocannabinol have been published; these suggested possible benefits of cannabis-derived agents for the treatment of tics. Trials examining other agents active on the cannabinoid system for tic disorders are currently ongoing. Cannabinoid-based treatments are a promising avenue of new research for medications that may help the Tourette syndrome population. However, given the limited research available, the overall efficacy and safety of cannabinoid-based treatments is largely unknown. Further trials are needed to examine dosing, active ingredients, and optimal mode of administration of cannabis-derived compounds, assuming initial trials suggest efficacy. Clinical use for refractory patients should at the very least be restricted to adult populations, given the uncertain efficacy and risk of developmental adverse effects that cannabinoids may have in children. Even in adult populations, cannabis-derived medications are associated with significant issues such as the effects they have on driving safety and the fact that they cause positive urine drug screens that can affect employment.
Collapse
Affiliation(s)
- Bekir B Artukoglu
- Yale University, Yale Child Study Center, PO Box 207900, New Haven, CT, 06520, USA.
| | - Michael H Bloch
- Department of Psychiatry, Yale University, Yale Child Study Center, New Haven, CT, USA
| |
Collapse
|
181
|
|
182
|
Vasudevan A, Argiriadi MA, Baranczak A, Friedman MM, Gavrilyuk J, Hobson AD, Hulce JJ, Osman S, Wilson NS. Covalent binders in drug discovery. PROGRESS IN MEDICINAL CHEMISTRY 2019; 58:1-62. [PMID: 30879472 DOI: 10.1016/bs.pmch.2018.12.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Covalent modulation of protein function can have multiple utilities including therapeutics, and probes to interrogate biology. While this field is still viewed with scepticism due to the potential for (idiosyncratic) toxicities, significant strides have been made in terms of understanding how to tune electrophilicity to selectively target specific residues. Progress has also been made in harnessing the potential of covalent binders to uncover novel biology and to provide an enhanced utility as payloads for Antibody Drug Conjugates. This perspective covers the tenets and applications of covalent binders.
Collapse
Affiliation(s)
| | | | | | | | - Julia Gavrilyuk
- AbbVie Stemcentrx, LLC, South San Francisco, CA, United States
| | | | | | - Sami Osman
- AbbVie Bioresearch Center, Worcester, MA, United States
| | | |
Collapse
|
183
|
Otrubova K, Chatterjee S, Ghimire S, Cravatt BF, Boger DL. N-Acyl pyrazoles: Effective and tunable inhibitors of serine hydrolases. Bioorg Med Chem 2019; 27:1693-1703. [PMID: 30879861 DOI: 10.1016/j.bmc.2019.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/08/2019] [Accepted: 03/08/2019] [Indexed: 11/24/2022]
Abstract
A series of N-acyl pyrazoles was examined as candidate serine hydrolase inhibitors in which the active site acylating reactivity and the leaving group ability of the pyrazole could be tuned not only through the nature of the acyl group (reactivity: amide > carbamate > urea), but also through pyrazole C4 substitution with electron-withdrawing or electron-donating substituents. Their impact on enzyme inhibitory activity displayed pronounced effects with the activity improving substantially as one alters both the nature of the reacting carbonyl group (urea > carbamate > amide) and the pyrazole C4 substituent (CN > H > Me). It was further demonstrated that the acyl chain of the N-acyl pyrazole ureas can be used to tailor the potency and selectivity of the inhibitor class to a targeted serine hydrolase. Thus, elaboration of the acyl chain of pyrazole-based ureas provided remarkably potent, irreversible inhibitors of fatty acid amide hydrolase (FAAH, apparent Ki = 100-200 pM), dual inhibitors of FAAH and monoacylglycerol hydrolase (MGLL), or selective inhibitors of MGLL (IC50 = 10-20 nM) while simultaneously minimizing off-target activity (e.g., ABHD6 and KIAA1363).
Collapse
Affiliation(s)
- Katerina Otrubova
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shreyosree Chatterjee
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Srijana Ghimire
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Benjamin F Cravatt
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Dale L Boger
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
184
|
Tóth KF, Ádám D, Bíró T, Oláh A. Cannabinoid Signaling in the Skin: Therapeutic Potential of the "C(ut)annabinoid" System. Molecules 2019; 24:E918. [PMID: 30845666 PMCID: PMC6429381 DOI: 10.3390/molecules24050918] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
The endocannabinoid system (ECS) has lately been proven to be an important, multifaceted homeostatic regulator, which influences a wide-variety of physiological processes all over the body. Its members, the endocannabinoids (eCBs; e.g., anandamide), the eCB-responsive receptors (e.g., CB₁, CB₂), as well as the complex enzyme and transporter apparatus involved in the metabolism of the ligands were shown to be expressed in several tissues, including the skin. Although the best studied functions over the ECS are related to the central nervous system and to immune processes, experimental efforts over the last two decades have unambiguously confirmed that cutaneous cannabinoid ("c[ut]annabinoid") signaling is deeply involved in the maintenance of skin homeostasis, barrier formation and regeneration, and its dysregulation was implicated to contribute to several highly prevalent diseases and disorders, e.g., atopic dermatitis, psoriasis, scleroderma, acne, hair growth and pigmentation disorders, keratin diseases, various tumors, and itch. The current review aims to give an overview of the available skin-relevant endo- and phytocannabinoid literature with a special emphasis on the putative translational potential, and to highlight promising future research directions as well as existing challenges.
Collapse
Affiliation(s)
- Kinga Fanni Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Dorottya Ádám
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
- HCEMM Nonprofit Ltd., 6720 Szeged, Hungary.
| | - Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
185
|
Bailey J, Balls M. Recent efforts to elucidate the scientific validity of animal-based drug tests by the pharmaceutical industry, pro-testing lobby groups, and animal welfare organisations. BMC Med Ethics 2019; 20:16. [PMID: 30823899 PMCID: PMC6397470 DOI: 10.1186/s12910-019-0352-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 02/19/2019] [Indexed: 01/09/2023] Open
Abstract
Background Even after several decades of human drug development, there remains an absence of published, substantial, comprehensive data to validate the use of animals in preclinical drug testing, and to point to their predictive nature with regard to human safety/toxicity and efficacy. Two recent papers, authored by pharmaceutical industry scientists, added to the few substantive publications that exist. In this brief article, we discuss both these papers, as well as our own series of three papers on the subject, and also various views and criticisms of lobby groups that advocate the animal testing of new drugs. Main text We argue that there still remains no published evidence to support the current regulatory paradigm of animal testing in supporting safe entry to clinical trials. In fact, the data in these recent studies, as well as in our own studies, support the contention that tests on rodents, dogs and monkeys provide next to no evidential weight to the probability of there being a lack of human toxicity, when there is no apparent toxicity in the animals. Conclusion Based on these data, and in particular on this finding, it must be concluded that animal drug tests are therefore not fit for their stated purpose. At the very least, it is now incumbent on—and we very much encourage—the pharmaceutical industry and its regulators to commission, conduct and/or facilitate further independent studies involving the use of substantial proprietary data.
Collapse
Affiliation(s)
- Jarrod Bailey
- Cruelty Free International, 16a Crane Grove, London, N7 8NN, UK.
| | - Michael Balls
- Faculty of Medicine and Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| |
Collapse
|
186
|
Sydow D, Burggraaff L, Szengel A, van Vlijmen HWT, IJzerman AP, van Westen GJP, Volkamer A. Advances and Challenges in Computational Target Prediction. J Chem Inf Model 2019; 59:1728-1742. [DOI: 10.1021/acs.jcim.8b00832] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Dominique Sydow
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Lindsey Burggraaff
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Angelika Szengel
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Herman W. T. van Vlijmen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
- Computational Chemistry, Janssen Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Adriaan P. IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Gerard J. P. van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | - Andrea Volkamer
- In silico Toxicology, Institute of Physiology, Charité − Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
187
|
Huang Z, Ogasawara D, Seneviratne UI, Cognetta AB, am Ende CW, Nason DM, Lapham K, Litchfield J, Johnson DS, Cravatt BF. Global Portrait of Protein Targets of Metabolites of the Neurotoxic Compound BIA 10-2474. ACS Chem Biol 2019; 14:192-197. [PMID: 30702848 DOI: 10.1021/acschembio.8b01097] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Clinical investigation of the fatty acid amide hydrolase (FAAH) inhibitor BIA 10-2474 resulted in serious adverse neurological events. Structurally unrelated FAAH inhibitors tested in humans have not presented safety concerns, suggesting that BIA 10-2474 has off-target activities. A recent activity-based protein profiling (ABPP) study revealed that BIA 10-2474 and one of its major metabolites inhibit multiple members of the serine hydrolase class to which FAAH belongs. Here, we extend these studies by performing a proteome-wide analysis of covalent targets of BIA 10-2474 metabolites. Using alkynylated probes for click chemistry-ABPP in human cells, we show that des-methylated metabolites of BIA 10-2474 covalently modify the conserved catalytic cysteine in aldehyde dehydrogenases, including ALDH2, which has been implicated in protecting the brain from oxidative stress-related damage. These findings indicate that BIA 10-2474 and its metabolites have the potential to inhibit multiple mechanistically distinct enzyme classes involved in nervous system function.
Collapse
Affiliation(s)
- Zhen Huang
- Medicine Design, Chemical Biology, Pfizer Worldwide Research and Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Daisuke Ogasawara
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Uthpala I. Seneviratne
- Medicine Design, Chemical Biology, Pfizer Worldwide Research and Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Armand B. Cognetta
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Christopher W. am Ende
- Medicine Design, Medicinal Chemistry, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Deane M. Nason
- Medicine Design, Medicinal Chemistry, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kimberly Lapham
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - John Litchfield
- Medicine Design, Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Douglas S. Johnson
- Medicine Design, Chemical Biology, Pfizer Worldwide Research and Development, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Benjamin F. Cravatt
- Department of Chemistry, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
188
|
Zhou J, Mock ED, Martella A, Kantae V, Di X, Burggraaff L, Baggelaar MP, Al-Ayed K, Bakker A, Florea BI, Grimm SH, den Dulk H, Li CT, Mulder L, Overkleeft HS, Hankemeier T, van Westen GJP, van der Stelt M. Activity-Based Protein Profiling Identifies α-Ketoamides as Inhibitors for Phospholipase A2 Group XVI. ACS Chem Biol 2019; 14:164-169. [PMID: 30620559 PMCID: PMC6379856 DOI: 10.1021/acschembio.8b00969] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023]
Abstract
Phospholipase A2, group XVI (PLA2G16) is a thiol hydrolase from the HRASLS family that regulates lipolysis in adipose tissue and has been identified as a host factor enabling the cellular entry of picornaviruses. Chemical tools are essential to visualize and control PLA2G16 activity, but they have not been reported to date. Here, we show that MB064, which is a fluorescent lipase probe, also labels recombinant and endogenously expressed PLA2G16. Competitive activity-based protein profiling (ABPP) using MB064 enabled the discovery of α-ketoamides as the first selective PLA2G16 inhibitors. LEI110 was identified as a potent PLA2G16 inhibitor ( Ki = 20 nM) that reduces cellular arachidonic acid levels and oleic acid-induced lipolysis in human HepG2 cells. Gel-based ABPP and chemical proteomics showed that LEI110 is a selective pan-inhibitor of the HRASLS family of thiol hydrolases (i.e., PLA2G16, HRASLS2, RARRES3 and iNAT). Molecular dynamic simulations of LEI110 in the reported crystal structure of PLA2G16 provided insight in the potential ligand-protein interactions to explain its binding mode. In conclusion, we have developed the first selective inhibitor that can be used to study the cellular role of PLA2G16.
Collapse
Affiliation(s)
- Juan Zhou
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Elliot D. Mock
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Andrea Martella
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Vasudev Kantae
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Department
of Analytical BioSciences and Metabolomics, Leiden Academic Centre
for Drug Research, Leiden University, Leiden, The Netherlands
| | - Xinyu Di
- Department
of Analytical BioSciences and Metabolomics, Leiden Academic Centre
for Drug Research, Leiden University, Leiden, The Netherlands
| | - Lindsey Burggraaff
- Department
of Computational Drug Discovery, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Marc P. Baggelaar
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Karol Al-Ayed
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Bakker
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Bogdan I. Florea
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Sebastian H. Grimm
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Hans den Dulk
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Chun T. Li
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Laura Mulder
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Herman S. Overkleeft
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Thomas Hankemeier
- Department
of Analytical BioSciences and Metabolomics, Leiden Academic Centre
for Drug Research, Leiden University, Leiden, The Netherlands
| | - Gerard J. P. van Westen
- Department
of Computational Drug Discovery, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mario van der Stelt
- Department
of Molecular Physiology, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| |
Collapse
|
189
|
Brain permeant and impermeant inhibitors of fatty-acid amide hydrolase suppress the development and maintenance of paclitaxel-induced neuropathic pain without producing tolerance or physical dependence in vivo and synergize with paclitaxel to reduce tumor cell line viability in vitro. Pharmacol Res 2019; 142:267-282. [PMID: 30739035 DOI: 10.1016/j.phrs.2019.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/22/2018] [Accepted: 02/01/2019] [Indexed: 12/19/2022]
Abstract
Activation of cannabinoid CB1 receptors suppresses pathological pain but also produces unwanted side effects, including tolerance and physical dependence. Inhibition of fatty-acid amide hydrolase (FAAH), the major enzyme catalyzing the degradation of anandamide (AEA), an endocannabinoid, and other fatty-acid amides, suppresses pain without unwanted side effects typical of direct CB1 agonists. However, FAAH inhibitors have failed to show efficacy in several clinical trials suggesting that the right partnership of FAAH inhibition and pathology has yet to be identified. We compared efficacy of chronic treatments with a centrally penetrant FAAH inhibitor (URB597), a peripherally restricted FAAH inhibitor (URB937) and an orthosteric pan-cannabinoid agonist (WIN55,212-2) in suppressing neuropathic pain induced by the chemotherapeutic agent paclitaxel. Each FAAH inhibitor suppressed the development of paclitaxel-induced neuropathic pain and reduced the maintenance of already established allodynia with sustained efficacy. Tolerance developed to the anti-allodynic efficacy of WIN55,212-2, but not to that of URB597 or URB937, in each dosing paradigm. Challenge with the CB1 antagonist rimonabant precipitated CB1-dependent withdrawal in paclitaxel-treated mice receiving WIN55,212-2 but not URB597 or URB937. When dosing with either URB597 or URB937 was restricted to the development of neuropathy, paclitaxel-induced allodynia emerged following termination of drug delivery. These observations suggest that both FAAH inhibitors were anti-allodynic rather than curative. Moreover, neither URB597 nor URB937 impeded the ability of paclitaxel to reduce breast (4T1) or ovarian (HeyA8) tumor cell line viability. In fact, URB597 and URB937 alone reduced 4T1 tumor cell line viability, albeit with low potency, and the dose matrix of each combination with paclitaxel was synergistic in reducing 4T1 and HeyA8 tumor cell line viability according to Bliss, Highest Single Agent (HSA) and Loewe additivity models. Both FAAH inhibitors synergized with paclitaxel to reduce 4T1 and HeyA8 tumor cell line viability without reducing viability of non-tumor HEK293 cells. Neither FAAH inhibitor reduced viability of non-tumor HEK293 cells in either the presence or absence of paclitaxel, suggesting that nonspecific cytotoxic effects were not produced by the same treatments. Our results suggest that FAAH inhibitors reduce paclitaxel-induced allodynia without the occurrence of CB1-dependence in vivo and may, in fact, enhance the anti-tumor actions of paclitaxel in vitro.
Collapse
|
190
|
Kuo CL, Kallemeijn WW, Lelieveld LT, Mirzaian M, Zoutendijk I, Vardi A, Futerman AH, Meijer AH, Spaink HP, Overkleeft HS, Aerts JMFG, Artola M. In vivo inactivation of glycosidases by conduritol B epoxide and cyclophellitol as revealed by activity-based protein profiling. FEBS J 2019; 286:584-600. [PMID: 30600575 PMCID: PMC6850446 DOI: 10.1111/febs.14744] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/16/2018] [Accepted: 01/01/2019] [Indexed: 01/18/2023]
Abstract
Glucocerebrosidase (GBA) is a lysosomal β‐glucosidase‐degrading glucosylceramide. Its deficiency causes Gaucher disease (GD), a common lysosomal storage disorder. Carrying a genetic abnormality in GBA constitutes at present the largest genetic risk factor for Parkinson's disease (PD). Conduritol B epoxide (CBE), a mechanism‐based irreversible inhibitor of GBA, is used to generate cell and animal models for investigations on GD and PD. However, CBE may have additional glycosidase targets besides GBA. Here, we present the first in vivo target engagement study for CBE, employing a suite of activity‐based probes to visualize catalytic pocket occupancy of candidate off‐target glycosidases. Only at significantly higher CBE concentrations, nonlysosomal glucosylceramidase (GBA2) and lysosomal α‐glucosidase were identified as major off‐targets in cells and zebrafish larvae. A tight, but acceptable window for selective inhibition of GBA in the brain of mice was observed. On the other hand, cyclophellitol, a closer glucose mimic, was found to inactivate with equal affinity GBA and GBA2 and therefore is not suitable to generate genuine GD‐like models. Enzymes Glucocerebrosidase (http://www.chem.qmul.ac.uk/iubmb/enzyme/EC3/2/1/45.html), nonlysosomal β‐glucocerebrosidase (http://www.chem.qmul.ac.uk/iubmb/enzyme/EC3/2/1/45.html); cytosolic β‐glucosidase (http://www.chem.qmul.ac.uk/iubmb/enzyme/EC3/2/1/21.html); α‐glucosidases (http://www.chem.qmul.ac.uk/iubmb/enzyme/EC3/2/1/20.html); β‐glucuronidase (http://www.chem.qmul.ac.uk/iubmb/enzyme/EC3/2/1/31.html).
Collapse
Affiliation(s)
- Chi-Lin Kuo
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Wouter W Kallemeijn
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Lindsey T Lelieveld
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Mina Mirzaian
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Iris Zoutendijk
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Ayelet Vardi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Herman S Overkleeft
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Johannes M F G Aerts
- Department of Medical Biochemistry, Leiden Institute of Chemistry, Leiden University, The Netherlands
| | - Marta Artola
- Department of Bio-organic Synthesis, Leiden Institute of Chemistry, Leiden University, The Netherlands
| |
Collapse
|
191
|
D'Souza DC, Cortes-Briones J, Creatura G, Bluez G, Thurnauer H, Deaso E, Bielen K, Surti T, Radhakrishnan R, Gupta A, Gupta S, Cahill J, Sherif MA, Makriyannis A, Morgan PT, Ranganathan M, Skosnik PD. Efficacy and safety of a fatty acid amide hydrolase inhibitor (PF-04457845) in the treatment of cannabis withdrawal and dependence in men: a double-blind, placebo-controlled, parallel group, phase 2a single-site randomised controlled trial. Lancet Psychiatry 2019; 6:35-45. [PMID: 30528676 DOI: 10.1016/s2215-0366(18)30427-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 11/20/2022]
Abstract
BACKGROUND Cannabis is one of the most widely used drugs worldwide. Cannabis use disorder is characterised by recurrent use of cannabis that causes significant clinical and functional impairment. There are no approved pharmacological treatments for cannabis use disorder. One approach is to potentiate endocannabinoid signalling by inhibiting fatty acid amide hydrolase (FAAH), the enzyme that degrades the endocannabinoid anandamide. We aimed to test the efficacy and safety of the FAAH-inhibitor PF-04457845 in reduction of cannabis withdrawal and cannabis use in men who were daily cannabis users. METHODS We did a double-blind, placebo-controlled, parallel group phase 2a trial at one site in men aged 18-55 years with cannabis dependence according to DSM-IV criteria (equivalent to cannabis use disorder in DSM-5). After baseline assessments, participants were randomly assigned (2:1) to receive PF-04457845 (4 mg per day) or placebo using a fixed block size of six participants, stratified by severity of cannabis use and desire to quit. Participants were admitted to hospital for 5 days (maximum 8 days) to achieve abstinence and precipitate cannabis withdrawal, after which they were discharged to continue the remaining 3 weeks of treatment as outpatients. The primary endpoints were treatment-related differences in cannabis withdrawal symptoms during hospital admission, and week 4 (end of treatment) self-reported cannabis use and urine THC-COOH concentrations in the intention-to-treat population. The study is registered at ClinicalTrials.gov, number NCT01618656. FINDINGS Between Sept 12, 2012, and Jan 18, 2016, 46 men were randomly assigned to PF-04457845 and 24 to placebo. Adherence to study medication was 88%, as confirmed by video-calling and pill count, and corroborated by corresponding drug and anandamide concentrations in blood. Relative to placebo, treatment with PF-04457845 was associated with reduced symptoms of cannabis withdrawal (first day of treatment mean symptom score 11·00 [95% CI 7·78-15·57] vs 6·04 [4·43-8·24]; difference 4·96 [0·71-9·21]; padj=0·048; second day of treatment 11·74 [8·28-16·66] vs 6·02 [4·28-8·47]; difference 5·73 [1·13-10·32]; padj=0·035) and related mood symptoms during the inpatient phase. Additionally, treatment with PF-04457845 was associated with lower self-reported cannabis use at 4 weeks (mean 1·27 joints per day [95% CI 0·82-1·97] vs 0·40 [0·25-0·62]; difference 0·88 [0·29-1·46]; p=0·0003) and lower urinary THC-COOH concentrations (mean 657·92 ng/mL [95% CI 381·60-1134·30] vs 265·55 [175·60-401·57]; difference 392·37 [17·55-767·18)]; p=0·009). Eight (17%) patients in the PF-04457845 group and four (17%) in the placebo group discontinued during the treatment period. During the 4-week treatment phase, 20 (43%) of 46 participants in the PF-04457845 group and 11 (46%) of 24 participants in the placebo group had an adverse event. There were no serious adverse events. INTERPRETATION PF-04457845, a novel FAAH inhibitor, reduced cannabis withdrawal symptoms and cannabis use in men, and might represent an effective and safe approach for the treatment of cannabis use disorder. FUNDING United States National Institute of Drug Abuse (NIDA).
Collapse
Affiliation(s)
- Deepak Cyril D'Souza
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Jose Cortes-Briones
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Gina Creatura
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Grai Bluez
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Halle Thurnauer
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Emma Deaso
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Kim Bielen
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Toral Surti
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajiv Radhakrishnan
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Aarti Gupta
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Swapnil Gupta
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - John Cahill
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Mohamed A Sherif
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | | | - Peter T Morgan
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Mohini Ranganathan
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Patrick D Skosnik
- Schizophrenia and Neuropharmacology Research Group, VA Connecticut Healthcare System, West Haven, CT, USA; Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, New Haven, CT, USA; Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
192
|
Kumar S, Sarma P, Kumar H, Prakash A, Medhi B. Modulation of endocannabinoid system: Success lies in the failures. Indian J Pharmacol 2018; 50:155-158. [PMID: 30505050 PMCID: PMC6234706 DOI: 10.4103/ijp.ijp_541_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Subodh Kumar
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Phulen Sarma
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Harish Kumar
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Ajay Prakash
- Department of Pharmacology, PGIMER, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, PGIMER, Chandigarh, India
| |
Collapse
|
193
|
Karakunnel JJ, Bui N, Palaniappan L, Schmidt KT, Mahaffey KW, Morrison B, Figg WD, Kummar S. Reviewing the role of healthy volunteer studies in drug development. J Transl Med 2018; 16:336. [PMID: 30509294 PMCID: PMC6278009 DOI: 10.1186/s12967-018-1710-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND With the exception of genotoxic oncology drugs, first-in-human, Phase 1 clinical studies of investigational drugs have traditionally been conducted in healthy volunteers (HVs). The primary goal of these studies is to investigate the pharmacokinetics and pharmacodynamics of a novel drug candidate, determine appropriate dosing, and document safety and tolerability. MAIN BODY When tailored to specific study objectives, HV studies are beneficial to manufacturers and patients alike and can be applied to both non-oncology and oncology drug development. Enrollment of HVs not only increases study accrual rates for dose-escalation studies but also alleviates the ethical concern of enrolling patients with disease in a short-term study at subtherapeutic doses when other studies (e.g. Phase 2 or Phase 3 studies) may be more appropriate for the patient. The use of HVs in non-oncology Phase 1 clinical trials is relatively safe but nonetheless poses ethical challenges because of the potential risks to which HVs are exposed. In general, most adverse events associated with non-oncology drugs are mild in severity, and serious adverse events are rare, but examples of severe toxicity have been reported. The use of HVs in the clinical development of oncology drugs is more limited but is nonetheless useful for evaluating clinical pharmacology and establishing an appropriate starting dose for studies in cancer patients. During the development of oncology drugs, clinical pharmacology studies in HVs have been used to assess pharmacokinetics, drug metabolism, food effects, potential drug-drug interactions, effects of hepatic and renal impairment, and other pharmacologic parameters vital for clinical decision-making in oncology. Studies in HVs are also being used to evaluate biosimilars versus established anticancer biologic agents. CONCLUSION A thorough assessment of toxicity and pharmacology throughout the drug development process is critical to ensure the safety of HVs. With the appropriate safeguards, HVs will continue to play an important role in future drug development.
Collapse
Affiliation(s)
| | - Nam Bui
- Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA 94305 USA
| | - Latha Palaniappan
- Department of Medicine, Stanford University School of Medicine, 900 Blake Wilbur Drive, Room W200, 2nd Floor MC 5358, Stanford, CA 94304 USA
| | - Keith T. Schmidt
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Kenneth W. Mahaffey
- Stanford Center for Clinical Research (SCCR), Department of Medicine, Stanford University School of Medicine, 300 Pasteur Drive, Grant S-102, Stanford, CA 94305 USA
| | | | - William D. Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892 USA
| | - Shivaani Kummar
- Stanford Cancer Institute, 875 Blake Wilbur Drive, Stanford, CA 94305 USA
| |
Collapse
|
194
|
Garzinsky D, Zahov S, Ekodo Voundi M, Hanekamp W, Lehr M. Tetrazolylpropan-2-ones as inhibitors of fatty acid amide hydrolase: Studies on structure-activity relationships and metabolic stability. Eur J Med Chem 2018; 160:183-192. [DOI: 10.1016/j.ejmech.2018.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 11/30/2022]
|
195
|
Lafreniere J, Kelly M. Potential for endocannabinoid system modulation in ocular pain and inflammation: filling the gaps in current pharmacological options. Neuronal Signal 2018; 2:NS20170144. [PMID: 32714590 PMCID: PMC7373237 DOI: 10.1042/ns20170144] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 02/06/2023] Open
Abstract
Challenges in the management of ocular pain are an underappreciated topic. Currently available therapeutics lack both efficacy and clear guidelines for their use, with many also possessing unacceptable side effects. Promising novel agents would offer analgesic, anti-inflammatory, and possibly neuroprotective actions; have favorable ocular safety profiles; and show potential in managing neuropathic pain. Growing evidence supports a link between the endocannabinoid system (ECS) and a range of physiological and disease processes, notably those involving inflammation and pain. Both preclinical and clinical data suggest analgesic and anti-inflammatory actions of cannabinoids and ECS-modifying drugs in chronic pain conditions, including those of neuropathic origin. This review will examine existing evidence for the anatomical and physiological basis of ocular pain, specifically, ocular surface disease and the development of chronic ocular pain. The mechanism of action, efficacy, and limitations of currently available treatments will be discussed, and current knowledge related to ECS-modulation of ocular pain and inflammatory disease will be summarized. A perspective will be provided on the future directions of ECS research in terms of developing cannabinoid therapeutics for ocular pain.
Collapse
Affiliation(s)
| | - Melanie E.M. Kelly
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Ophthalmology and Visual Sciences, Dalhousie University, Halifax, NS, Canada
- Department of Anesthesia, Pain Management and Perioperative Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
196
|
Archibald K, Tsaioun K, Kenna JG, Pound P. Better science for safer medicines: the human imperative. J R Soc Med 2018; 111:433-438. [PMID: 30439294 PMCID: PMC6295948 DOI: 10.1177/0141076818812783] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
| | - Katya Tsaioun
- Evidence-Based Toxicology Collaboration, John Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | | |
Collapse
|
197
|
Abstract
Fatty acid amides represent a diverse and underappreciated family of lipids found in vertebrates and invertebrates. The most recognized, most studied, and best understood members of the fatty acid amide family are N-arachidonoylethanolamine (anandamide) and oleamide. Over 70 other fatty acid amides have been identified from biological systems and these non-anandamide and non-oleamide fatty acid amides are not well understood: their cellular functions, transport, biosynthesis, and degradation are, at best, partially elucidated. Most of the fatty acid amides are “orphan” ligands for “orphan” or unknown receptors. Interest in the fatty acid amides will wane without a more complete understanding of their function in vivo and most of these lipids will be mentioned in a few sentences in reviews on ananamide and/or olemide. In this commentary, we suggest that one strategy to dramatically increase our understanding of any member of the fatty acid amide family is the design, synthesis, and proper use of binding-based profiling probes (BBPPs) based on the structure of a specific fatty acid amide. A BBPP is an analog of a fatty acid amide that enables the controlled covalent attachment of the probe to a fatty acid amide-binding protein and, also, possesses a chemical moiety that will allow the purification and/or detection of the BBPP-labeled proteins. The identification of the proteins that specifically bind a fatty acid amide will foster a better understanding of the function, transport, and metabolism of a fatty acid amide.
Collapse
Affiliation(s)
- David J Merkler
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, USA.,Center for Drug Discovery and Innovation, University of South Florida, Tampa, FL USA.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL USA
| |
Collapse
|
198
|
Pound P, Ritskes-Hoitinga M. Is it possible to overcome issues of external validity in preclinical animal research? Why most animal models are bound to fail. J Transl Med 2018; 16:304. [PMID: 30404629 PMCID: PMC6223056 DOI: 10.1186/s12967-018-1678-1] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/31/2018] [Indexed: 12/12/2022] Open
Abstract
Background The pharmaceutical industry is in the midst of a productivity crisis and rates of translation from bench to bedside are dismal. Patients are being let down by the current system of drug discovery; of the several 1000 diseases that affect humans, only a minority have any approved treatments and many of these cause adverse reactions in humans. A predominant reason for the poor rate of translation from bench to bedside is generally held to be the failure of preclinical animal models to predict clinical efficacy and safety. Attempts to explain this failure have focused on problems of internal validity in preclinical animal studies (e.g. poor study design, lack of measures to control bias). However there has been less discussion of another key factor that influences translation, namely the external validity of preclinical animal models. Review of problems of external validity External validity is the extent to which research findings derived in one setting, population or species can be reliably applied to other settings, populations and species. This paper argues that the reliable translation of findings from animals to humans will only occur if preclinical animal studies are both internally and externally valid. We review several key aspects that impact external validity in preclinical animal research, including unrepresentative animal samples, the inability of animal models to mimic the complexity of human conditions, the poor applicability of animal models to clinical settings and animal–human species differences. We suggest that while some problems of external validity can be overcome by improving animal models, the problem of species differences can never be overcome and will always undermine external validity and the reliable translation of preclinical findings to humans. Conclusion We conclude that preclinical animal models can never be fully valid due to the uncertainties introduced by species differences. We suggest that even if the next several decades were spent improving the internal and external validity of animal models, the clinical relevance of those models would, in the end, only improve to some extent. This is because species differences would continue to make extrapolation from animals to humans unreliable. We suggest that to improve clinical translation and ultimately benefit patients, research should focus instead on human-relevant research methods and technologies.
Collapse
Affiliation(s)
- Pandora Pound
- Safer Medicines Trust, PO Box 122, Kingsbridge, TQ7 9AX, UK.
| | - Merel Ritskes-Hoitinga
- SYRCLE, Department for Health Evidence, Radboud University Medical Center, PO Box 9101, Route 133, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
199
|
Kodani SD, Wan D, Wagner KM, Hwang SH, Morisseau C, Hammock BD. Design and Potency of Dual Soluble Epoxide Hydrolase/Fatty Acid Amide Hydrolase Inhibitors. ACS OMEGA 2018; 3:14076-14086. [PMID: 30411058 PMCID: PMC6210075 DOI: 10.1021/acsomega.8b01625] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 09/04/2018] [Indexed: 06/08/2023]
Abstract
Fatty acid amide hydrolase (FAAH) is responsible for regulating concentrations of the endocannabinoid arachidonoyl ethanolamide. Multiple FAAH inhibitors have been developed for clinical trials and have failed to demonstrate efficacy at treating pain, despite promising preclinical data. One approach toward increasing the efficacy of FAAH inhibitors is to concurrently inhibit other targets responsible for regulating pain. Here, we designed dual inhibitors targeting the enzymes FAAH and soluble epoxide hydrolase (sEH), which are targets previously shown to synergize at reducing inflammatory and neuropathic pain. Exploration of the sEH/FAAH inhibitor structure-activity relationship started with PF-750, a FAAH inhibitor (IC50 = 19 nM) that weakly inhibited sEH (IC50 = 640 nM). Potency was optimized resulting in an inhibitor with improved potency on both targets (11, sEH IC50 = 5 nM, FAAH IC50 = 8 nM). This inhibitor demonstrated good target selectivity, pharmacokinetic properties (AUC = 1200 h nM, t 1/2 = 4.9 h in mice), and in vivo target engagement.
Collapse
|
200
|
Jiang M, van der Stelt M. Activity-Based Protein Profiling Delivers Selective Drug Candidate ABX-1431, a Monoacylglycerol Lipase Inhibitor, To Control Lipid Metabolism in Neurological Disorders. J Med Chem 2018; 61:9059-9061. [PMID: 30354159 PMCID: PMC6204598 DOI: 10.1021/acs.jmedchem.8b01405] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Monoacylglycerol
lipase (MGLL or MAGL) is a critical point of regulation
of both endocannabinoid and eicosanoid signaling pathways in the brain,
thereby providing novel therapeutic opportunities for neurological
and neurodegenerative diseases. In this issue Cisar et al. disclose
the discovery, optimization, and initial preclinical profiling of
ABX-1431, a covalent, irreversible MGLL inhibitor. Activity-based
protein profiling was key to the discovery of ABX-1431. ABX-1431 is
a first-in-class experimental drug that was well-tolerated and safe
in phase 1 clinical studies. Data from an exploratory phase 1b study
indicate that it has the potential to treat symptoms of adult patients
with syndrome of Gilles de la Tourette. ABX-1431 is currently entering
clinical phase 2 studies for this neurological disorder as well as
for other indications, such as neuromyeltis optica and multiple sclerosis.
Collapse
Affiliation(s)
- Ming Jiang
- Department of Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2333 CC Leiden , The Netherlands
| | - Mario van der Stelt
- Department of Molecular Physiology, Leiden Institute of Chemistry , Leiden University , 2333 CC Leiden , The Netherlands
| |
Collapse
|