1
|
Neal WM, Pandey P, Khan SI, Khan IA, Chittiboyina AG. Machine learning and traditional QSAR modeling methods: a case study of known PXR activators. J Biomol Struct Dyn 2024; 42:903-917. [PMID: 37059719 DOI: 10.1080/07391102.2023.2196701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/22/2023] [Indexed: 04/16/2023]
Abstract
Pregnane X receptor (PXR), extensively expressed in human tissues related to digestion and metabolism, is responsible for recognizing and detoxifying diverse xenobiotics encountered by humans. To comprehend the promiscuous nature of PXR and its ability to bind a variety of ligands, computational approaches, viz., quantitative structure-activity relationship (QSAR) models, aid in the rapid dereplication of potential toxicological agents and mitigate the number of animals used to establish a meaningful regulatory decision. Recent advancements in machine learning techniques accommodating larger datasets are expected to aid in developing effective predictive models for complex mixtures (viz., dietary supplements) before undertaking in-depth experiments. Five hundred structurally diverse PXR ligands were used to develop traditional two-dimensional (2D) QSAR, machine-learning-based 2D-QSAR, field-based three-dimensional (3D) QSAR, and machine-learning-based 3D-QSAR models to establish the utility of predictive machine learning methods. Additionally, the applicability domain of the agonists was established to ensure the generation of robust QSAR models. A prediction set of dietary PXR agonists was used to externally-validate generated QSAR models. QSAR data analysis revealed that machine-learning 3D-QSAR techniques were more accurate in predicting the activity of external terpenes with an external validation squared correlation coefficient (R2) of 0.70 versus an R2 of 0.52 in machine-learning 2D-QSAR. Additionally, a visual summary of the binding pocket of PXR was assembled from the field 3D-QSAR models. By developing multiple QSAR models in this study, a robust groundwork for assessing PXR agonism from various chemical backbones has been established in anticipation of the identification of potential causative agents in complex mixtures.
Collapse
Affiliation(s)
- William M Neal
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Pankaj Pandey
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Shabana I Khan
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Ikhlas A Khan
- Division of Pharmacognosy, Department of BioMolecular Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| | - Amar G Chittiboyina
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, USA
| |
Collapse
|
2
|
Chen Q, Zhou X, Rehmel J, Steele JP, Svensson KA, Beck JP, Hembre EJ, Hao J. Ensemble Docking Approach to Mitigate Pregnane X Receptor-Mediated CYP3A4 Induction Risk. J Chem Inf Model 2023; 63:173-186. [PMID: 36473234 DOI: 10.1021/acs.jcim.2c01175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three structurally closely related dopamine D1 receptor positive allosteric modulators (D1 PAMs) based on a tetrahydroisoquinoline (THIQ) scaffold were profiled for their CYP3A4 induction potentials. It was found that the length of the linker at the C5 position greatly affected the potentials of these D1 PAMs as CYP3A4 inducers, and the level of induction correlated well with the activation of the pregnane X receptor (PXR). Based on the published PXR X-ray crystal structures, we built a binding model specifically for these THIQ-scaffold-based D1 PAMs in the PXR ligand-binding pocket via an ensemble docking approach and found the model could explain the observed CYP induction disparity. Combined with our previously reported D1 receptor homology model, which identified the C5 position as pointing toward the solvent-exposed space, our PXR-binding model coincidentally suggested that structural modifications at the C5 position could productively modulate the CYP induction potential while maintaining the D1 PAM potency of these THIQ-based PAMs.
Collapse
Affiliation(s)
- Qi Chen
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Xin Zhou
- Drug Disposition, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Jessica Rehmel
- Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Steele
- Quantitative Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Kjell A Svensson
- Neuroscience Discovery, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - James P Beck
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| | - Erik J Hembre
- Discovery Chemistry Research and Technologies, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Junliang Hao
- Discovery Chemistry Research and Technologies, Lilly Biotechnology Center, Eli Lilly and Company, 10290 Campus Point Drive, San Diego, California92121, United States
| |
Collapse
|
3
|
Liu T, Beck JP, Hao J. A concise review on hPXR ligand-recognizing residues and structure-based strategies to alleviate hPXR transactivation risk. RSC Med Chem 2022; 13:129-137. [PMID: 35308029 PMCID: PMC8864553 DOI: 10.1039/d1md00348h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/03/2022] [Indexed: 01/21/2023] Open
Abstract
The human pregnane X receptor (hPXR) regulates the expression of major drug metabolizing enzymes. A wide range of drug candidates bind and activate hPXR, and hence are at risk of increasing drug-drug interactions and reducing clinical efficacy. hPXR structural features that function as hot spots for ligand binding are identified and highlighted in this concise review. Based on literature structure-activity relationship data as case studies, structure-based strategies to mitigate hPXR transactivation are summarized for medicinal chemists.
Collapse
Affiliation(s)
- Tao Liu
- Discovery Chemistry Research & Technologies, Eli Lilly and Company, Lilly Biotechnology Center 10290 Campus Point Drive San Diego CA 92121 USA
| | - James P Beck
- Discovery Chemistry Research & Technologies, Eli Lilly and Company, Lilly Biotechnology Center 10290 Campus Point Drive San Diego CA 92121 USA
| | - Junliang Hao
- Discovery Chemistry Research & Technologies, Eli Lilly and Company, Lilly Biotechnology Center 10290 Campus Point Drive San Diego CA 92121 USA
| |
Collapse
|
4
|
Subbaiah MAM, Meanwell NA. Bioisosteres of the Phenyl Ring: Recent Strategic Applications in Lead Optimization and Drug Design. J Med Chem 2021; 64:14046-14128. [PMID: 34591488 DOI: 10.1021/acs.jmedchem.1c01215] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The benzene moiety is the most prevalent ring system in marketed drugs, underscoring its historic popularity in drug design either as a pharmacophore or as a scaffold that projects pharmacophoric elements. However, introspective analyses of medicinal chemistry practices at the beginning of the 21st century highlighted the indiscriminate deployment of phenyl rings as an important contributor to the poor physicochemical properties of advanced molecules, which limited their prospects of being developed into effective drugs. This Perspective deliberates on the design and applications of bioisosteric replacements for a phenyl ring that have provided practical solutions to a range of developability problems frequently encountered in lead optimization campaigns. While the effect of phenyl ring replacements on compound properties is contextual in nature, bioisosteric substitution can lead to enhanced potency, solubility, and metabolic stability while reducing lipophilicity, plasma protein binding, phospholipidosis potential, and inhibition of cytochrome P450 enzymes and the hERG channel.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon-Bristol Myers Squibb Research and Development Centre, Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bangalore, Karnataka 560099, India
| | - Nicholas A Meanwell
- Department of Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
5
|
Hall A, Chanteux H, Ménochet K, Ledecq M, Schulze MSED. Designing Out PXR Activity on Drug Discovery Projects: A Review of Structure-Based Methods, Empirical and Computational Approaches. J Med Chem 2021; 64:6413-6522. [PMID: 34003642 DOI: 10.1021/acs.jmedchem.0c02245] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This perspective discusses the role of pregnane xenobiotic receptor (PXR) in drug discovery and the impact of its activation on CYP3A4 induction. The use of structural biology to reduce PXR activity on drug discovery projects has become more common in recent years. Analysis of this work highlights several important molecular interactions, and the resultant structural modifications to reduce PXR activity are summarized. The computational approaches undertaken to support the design of new drugs devoid of PXR activation potential are also discussed. Finally, the SAR of empirical design strategies to reduce PXR activity is reviewed, and the key SAR transformations are discussed and summarized. In conclusion, this perspective demonstrates that PXR activity can be greatly diminished or negated on active drug discovery projects with the knowledge now available. This perspective should be useful to anyone who seeks to reduce PXR activity on a drug discovery project.
Collapse
Affiliation(s)
- Adrian Hall
- UCB, Avenue de l'Industrie, Braine-L'Alleud 1420, Belgium
| | | | | | - Marie Ledecq
- UCB, Avenue de l'Industrie, Braine-L'Alleud 1420, Belgium
| | | |
Collapse
|
6
|
Liu Y, Xin H, Yin J, Yin D. Microwave-assisted synthesis of 3-substituted-6-ferrocene methylene-1,2,4-triazolo[3,4-b]-1,3,4-thiadiazoles. TRANSIT METAL CHEM 2018. [DOI: 10.1007/s11243-018-0219-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
7
|
Bechara WS, Khazhieva IS, Rodriguez E, Charette AB. One-pot synthesis of 3,4,5-trisubstituted 1,2,4-triazoles via the addition of hydrazides to activated secondary amides. Org Lett 2015; 17:1184-7. [PMID: 25700199 DOI: 10.1021/acs.orglett.5b00128] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A general approach has been developed for the one-pot synthesis of 3,4,5-trisubstituted 1,2,4-triazoles from secondary amides and hydrazides via triflic anhydride activation followed by microwave-induced cyclodehydration. In addition, the 1,2,4-triazole moiety is shown to be a useful directing group for Ru-catalyzed C-H arylation. Access to 1,2,4-triazolophenanthridine can be achieved from the reaction products using a Pd-catalyzed intramolecular C-H functionalization reaction.
Collapse
Affiliation(s)
- William S Bechara
- Centre in Green Chemistry and Catalysis, Faculty of Arts and Sciences, Department of Chemistry, Université de Montréal , P.O. Box 6128, Station Downtown, Montréal, Québec Canada H3C 3J7
| | | | | | | |
Collapse
|
8
|
Drost RM, Broere DLJ, Hoogenboom J, de Baan SN, Lutz M, de Bruin B, Elsevier CJ. Allylpalladium(II) Histidylidene Complexes and Their Application in
Z
‐Selective Transfer Semihydrogenation of Alkynes. Eur J Inorg Chem 2015. [DOI: 10.1002/ejic.201403104] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ruben M. Drost
- Molecular Inorganic Chemistry, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, the Netherlands, http://hims.uva.nl/research/research‐groups/content/molecular‐ inorganic‐chemistry/molecular‐inorganic‐chemistry.html
| | - Daniël L. J. Broere
- Homogeneous Catalysis, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, The Netherlands
| | - Jorin Hoogenboom
- Molecular Inorganic Chemistry, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, the Netherlands, http://hims.uva.nl/research/research‐groups/content/molecular‐ inorganic‐chemistry/molecular‐inorganic‐chemistry.html
| | - Simone N. de Baan
- Molecular Inorganic Chemistry, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, the Netherlands, http://hims.uva.nl/research/research‐groups/content/molecular‐ inorganic‐chemistry/molecular‐inorganic‐chemistry.html
| | - Martin Lutz
- Crystal and Structural Chemistry, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - B. de Bruin
- Homogeneous Catalysis, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, The Netherlands
| | - C. J. Elsevier
- Molecular Inorganic Chemistry, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Science Park 904, 1090 GD Amsterdam, the Netherlands, http://hims.uva.nl/research/research‐groups/content/molecular‐ inorganic‐chemistry/molecular‐inorganic‐chemistry.html
| |
Collapse
|
9
|
Goldberg FW, Dossetter AG, Scott JS, Robb GR, Boyd S, Groombridge SD, Kemmitt PD, Sjögren T, Gutierrez PM, deSchoolmeester J, Swales JG, Turnbull AV, Wild MJ. Optimization of Brain Penetrant 11β-Hydroxysteroid Dehydrogenase Type I Inhibitors and in Vivo Testing in Diet-Induced Obese Mice. J Med Chem 2014; 57:970-86. [DOI: 10.1021/jm4016729] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
| | | | - James S. Scott
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Graeme R. Robb
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Scott Boyd
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Sam D. Groombridge
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Paul D. Kemmitt
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Tove Sjögren
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | | | | | - John G. Swales
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Andrew V. Turnbull
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| | - Martin J. Wild
- AstraZeneca, Mereside, Alderley Park, Macclesfield SK10 4TG, United Kingdom
| |
Collapse
|
10
|
Scott JS, Goldberg FW, Turnbull AV. Medicinal Chemistry of Inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1 (11β-HSD1). J Med Chem 2013; 57:4466-86. [DOI: 10.1021/jm4014746] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- James S. Scott
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Frederick W. Goldberg
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| | - Andrew V. Turnbull
- AstraZeneca Innovative Medicines, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, U.K
| |
Collapse
|
11
|
Shao PP, Ye F, Chakravarty PK, Herrington JB, Dai G, Bugianesi RM, Haedo RJ, Swensen AM, Warren VA, Smith MM, Garcia ML, McManus OB, Lyons KA, Li X, Green M, Jochnowitz N, McGowan E, Mistry S, Sun SY, Abbadie C, Kaczorowski GJ, Duffy JL. Improved Cav2.2 Channel Inhibitors through a gem-Dimethylsulfone Bioisostere Replacement of a Labile Sulfonamide. ACS Med Chem Lett 2013; 4:1064-8. [PMID: 24900606 DOI: 10.1021/ml4002612] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 09/08/2013] [Indexed: 12/22/2022] Open
Abstract
We report the investigation of sulfonamide-derived Cav2.2 inhibitors to address drug-metabolism liabilities with this lead class of analgesics. Modification of the benzamide substituent provided improvements in both potency and selectivity. However, we discovered that formation of the persistent 3-(trifluoromethyl)benzenesulfonamide metabolite was an endemic problem in the sulfonamide series and that the replacement of the center aminopiperidine scaffold failed to prevent this metabolic pathway. This issue was eventually addressed by application of a bioisostere strategy. The new gem-dimethyl sulfone series retained Cav2.2 potency without the liability of the circulating sulfonamide metabolite.
Collapse
Affiliation(s)
- Pengcheng P. Shao
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Feng Ye
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Prasun K. Chakravarty
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - James B. Herrington
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Ge Dai
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Randal M. Bugianesi
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Rodolfo J. Haedo
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Andrew M. Swensen
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Vivien A. Warren
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - McHardy M. Smith
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Maria L. Garcia
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Owen B. McManus
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Kathryn A. Lyons
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Xiaohua Li
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Mitchell Green
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Nina Jochnowitz
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Erin McGowan
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Shruti Mistry
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Shu-Yu Sun
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Catherine Abbadie
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Gregory J. Kaczorowski
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| | - Joseph L. Duffy
- Departments of Medicinal Chemistry, ‡Ion Channels, §Drug Metabolism and
Pharmacokinetics, and ∥Pharmacology, Merck Research Laboratories, Rahway, New Jersey 07065, United States
| |
Collapse
|
12
|
Micksch M, Tenne M, Strassner T. Synthesis of 1,2-Diaryl- and 1-Aryl-2-alkylimidazoles with Sterically Demanding Substituents. European J Org Chem 2013. [DOI: 10.1002/ejoc.201300688] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
13
|
Wyrwoll CS, Holmes MC, Seckl JR. 11β-hydroxysteroid dehydrogenases and the brain: from zero to hero, a decade of progress. Front Neuroendocrinol 2011; 32:265-86. [PMID: 21144857 PMCID: PMC3149101 DOI: 10.1016/j.yfrne.2010.12.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 12/01/2010] [Accepted: 12/01/2010] [Indexed: 12/11/2022]
Abstract
Glucocorticoids have profound effects on brain development and adult CNS function. Excess or insufficient glucocorticoids cause myriad abnormalities from development to ageing. The actions of glucocorticoids within cells are determined not only by blood steroid levels and target cell receptor density, but also by intracellular metabolism by 11β-hydroxysteroid dehydrogenases (11β-HSD). 11β-HSD1 regenerates active glucocorticoids from their inactive 11-keto derivatives and is widely expressed throughout the adult CNS. Elevated hippocampal and neocortical 11β-HSD1 is observed with ageing and causes cognitive decline; its deficiency prevents the emergence of cognitive defects with age. Conversely, 11β-HSD2 is a dehydrogenase, inactivating glucocorticoids. The major central effects of 11β-HSD2 occur in development, as expression of 11β-HSD2 is high in fetal brain and placenta. Deficient feto-placental 11β-HSD2 results in a life-long phenotype of anxiety and cardiometabolic disorders, consistent with early life glucocorticoid programming.
Collapse
Affiliation(s)
- Caitlin S Wyrwoll
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | | | | |
Collapse
|
14
|
Su X, Vicker N, Thomas MP, Pradaux-Caggiano F, Halem H, Culler MD, Potter BVL. Discovery of adamantyl heterocyclic ketones as potent 11β-hydroxysteroid dehydrogenase type 1 inhibitors. ChemMedChem 2011; 6:1439-51. [PMID: 21608132 PMCID: PMC3170876 DOI: 10.1002/cmdc.201100144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Indexed: 11/11/2022]
Abstract
11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) plays a key role in converting intracellular cortisone to physiologically active cortisol, which is implicated in the development of several phenotypes of metabolic syndrome. Inhibition of 11β-HSD1 activity with selective inhibitors has beneficial effects on various conditions, including diabetes, dyslipidemia and obesity, and therefore constitutes a promising strategy to discover novel therapies for metabolic and cardiovascular diseases. A series of novel adamantyl heterocyclic ketones provides potent and selective inhibitors of human 11β-HSD1. Lead compounds display low nanomolar inhibition against human and mouse 11β-HSD1 and are selective with no activity against 11β-HSD2 and 17β-HSD1. Selected potent 11β-HSD1 inhibitors show moderate metabolic stability upon incubation with human liver microsomes and weak inhibition of human CYP450 enzymes.
Collapse
Affiliation(s)
- Xiangdong Su
- Medicinal Chemistry, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA27AY, UK
| | | | | | | | | | | | | |
Collapse
|
15
|
Penning TM. Human hydroxysteroid dehydrogenases and pre-receptor regulation: insights into inhibitor design and evaluation. J Steroid Biochem Mol Biol 2011; 125:46-56. [PMID: 21272640 PMCID: PMC3104102 DOI: 10.1016/j.jsbmb.2011.01.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 01/18/2011] [Accepted: 01/18/2011] [Indexed: 11/16/2022]
Abstract
Hydroxysteroid dehydrogenases (HSDs) represent a major class of NAD(P)(H) dependent steroid hormone oxidoreductases involved in the pre-receptor regulation of hormone action. This is achieved by HSDs working in pairs so that they can interconvert ketosteroids with hydroxysteroids resulting in a change in ligand potency for nuclear receptors. HSDs belong to two protein superfamilies the aldo-keto reductases and the short-chain dehydrogenase/reductases. In humans, many of the important enzymes have been thoroughly characterized including the elucidation of their three-dimensional structures. Because these enzymes play fundamental roles in steroid hormone action they can be considered to be drug targets for a variety of steroid driven diseases, e.g. metabolic syndrome and obesity, inflammation, and hormone dependent malignancies of the endometrium, prostate and breast. This article will review how fundamental knowledge of these enzymes can be exploited in the development of isoform specific HSD inhibitors from both protein superfamilies. Article from the Special issue on Targeted Inhibitors.
Collapse
Affiliation(s)
- Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6084, USA.
| |
Collapse
|
16
|
Maletic M, Leeman A, Szymonifka M, Mundt SS, Zokian HJ, Shah K, Dragovic J, Lyons K, Thieringer R, Vosatka AH, Balkovec J, Waddell ST. Bicyclo[2.2.2]octyltriazole inhibitors of 11β-hydoxysteroid dehydrogenase type 1. Pharmacological agents for the treatment of metabolic syndrome. Bioorg Med Chem Lett 2011; 21:2568-72. [DOI: 10.1016/j.bmcl.2011.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/04/2011] [Accepted: 01/06/2011] [Indexed: 01/22/2023]
|
17
|
Sun W, Maletic M, Mundt SS, Shah K, Zokian H, Lyons K, Waddell ST, Balkovec J. Substituted phenyl triazoles as selective inhibitors of 11β-Hydroxysteroid Dehydrogenase Type 1. Bioorg Med Chem Lett 2011; 21:2141-5. [DOI: 10.1016/j.bmcl.2011.01.125] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/27/2011] [Accepted: 01/28/2011] [Indexed: 11/29/2022]
|
18
|
Sun D, Wang Z, Caille S, DeGraffenreid M, Gonzalez-Lopez de Turiso F, Hungate R, Jaen JC, Jiang B, Julian LD, Kelly R, McMinn DL, Kaizerman J, Rew Y, Sudom A, Tu H, Ursu S, Walker N, Willcockson M, Yan X, Ye Q, Powers JP. Synthesis and optimization of novel 4,4-disubstituted cyclohexylbenzamide derivatives as potent 11β-HSD1 inhibitors. Bioorg Med Chem Lett 2010; 21:405-10. [PMID: 21093258 DOI: 10.1016/j.bmcl.2010.10.129] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2010] [Revised: 10/23/2010] [Accepted: 10/26/2010] [Indexed: 11/20/2022]
Abstract
The synthesis and SAR of a series of 4,4-disubstituted cyclohexylbenzamide inhibitors of 11β-HSD1 are described. Optimization rapidly led to potent, highly selective, and orally bioavailable inhibitors demonstrating efficacy in both rat and non-human primate ex vivo pharmacodynamic models.
Collapse
Affiliation(s)
- Daqing Sun
- Amgen Inc, 1120 Veterans Boulevard, South San Francisco, CA 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
DalZotto C, Michaux J, Martinand-Lurin E, Campagne JM. Chan-Lam-Evans Coupling of Cbz-Protected Histidines. European J Org Chem 2010. [DOI: 10.1002/ejoc.201000591] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
20
|
Siu M, Johnson TO, Wang Y, Nair SK, Taylor WD, Cripps SJ, Matthews JJ, Edwards MP, Pauly TA, Ermolieff J, Castro A, Hosea NA, LaPaglia A, Fanjul AN, Vogel JE. N-(Pyridin-2-yl) arylsulfonamide inhibitors of 11β-hydroxysteroid dehydrogenase type 1: Discovery of PF-915275. Bioorg Med Chem Lett 2009; 19:3493-7. [DOI: 10.1016/j.bmcl.2009.05.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 05/03/2009] [Accepted: 05/04/2009] [Indexed: 11/24/2022]
|
21
|
Discovery and structure–activity relationships of pentanedioic acid diamides as potent inhibitors of 11β-hydroxysteroid dehydrogenase type I. Bioorg Med Chem Lett 2009; 19:2674-8. [DOI: 10.1016/j.bmcl.2009.03.140] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Revised: 03/26/2009] [Accepted: 03/27/2009] [Indexed: 11/24/2022]
|
22
|
Rew Y, McMinn DL, Wang Z, He X, Hungate RW, Jaen JC, Sudom A, Sun D, Tu H, Ursu S, Villemure E, Walker NPC, Yan X, Ye Q, Powers JP. Discovery and optimization of piperidyl benzamide derivatives as a novel class of 11beta-HSD1 inhibitors. Bioorg Med Chem Lett 2009; 19:1797-801. [PMID: 19217779 DOI: 10.1016/j.bmcl.2009.01.058] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2008] [Revised: 01/15/2009] [Accepted: 01/16/2009] [Indexed: 11/16/2022]
Abstract
Discovery and optimization of a piperidyl benzamide series of 11beta-HSD1 inhibitors is described. This series was derived from a cyclohexyl benzamide lead structures to address PXR selectivity, high non-specific protein binding, poor solubility, limited in vivo exposure, and in vitro cytotoxicity issues observed with the cyclohexyl benzamide structures. These efforts led to the discovery of piperidyl benzamide 15 which features improved properties over the cyclohexyl benzamide derivatives.
Collapse
Affiliation(s)
- Yosup Rew
- Amgen Inc., 1120 Veterans Boulevard, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|