1
|
Fang J, Tang Y, Gong C, Huang Z, Feng Y, Liu G, Tang Y, Li W. Prediction of Cytochrome P450 Substrates Using the Explainable Multitask Deep Learning Models. Chem Res Toxicol 2024; 37:1535-1548. [PMID: 39196814 DOI: 10.1021/acs.chemrestox.4c00199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Abstract
Cytochromes P450 (P450s or CYPs) are the most important phase I metabolic enzymes in the human body and are responsible for metabolizing ∼75% of the clinically used drugs. P450-mediated metabolism is also closely associated with the formation of toxic metabolites and drug-drug interactions. Therefore, it is of high importance to predict if a compound is the substrate of a given P450 in the early stage of drug development. In this study, we built the multitask learning models to simultaneously predict the substrates of five major drug-metabolizing P450 enzymes, namely, CYP3A4, 2C9, 2C19, 2D6, and 1A2, based on the collected substrate data sets. Compared to the single-task model and conventional machine learning models, the multitask fingerprints and graph neural networks model achieved superior performance with the average AUC values of 90.8% on the test set. Notably, the multitask model demonstrated its good performance on the small amount of substrate data sets such as CYP1A2, 2C9, and 2C19. In addition, the Shapley additive explanation and the attention mechanism were used to reveal specific substructures associated with P450 substrates, which were further confirmed and complemented by the substructure mining tool and the literature.
Collapse
Affiliation(s)
- Jiaojiao Fang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yan Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Changda Gong
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Zejun Huang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yanjun Feng
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
2
|
Nakashima S, Fukami T, Kudo T, Nakano M, Matsui A, Ishiguro N, Nakajima M. Iminium ion metabolites are formed from nintedanib by human CYP3A4. Drug Metab Pharmacokinet 2024; 57:101025. [PMID: 39068856 DOI: 10.1016/j.dmpk.2024.101025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 07/30/2024]
Abstract
Nintedanib is used to treat idiopathic pulmonary fibrosis, systemic sclerosis, interstitial lung disease, and progressive fibrotic interstitial lung disease. It is primarily cleared via hepatic metabolism, hydrolysis, and glucuronidation. In addition, formation of the iminium ion, a possible reactive metabolite, was predicted based on the chemical structure of nintedanib. To obtain a hint which may help to clarify the cause of nintedanib-induced liver injury, we investigated whether iminium ions were formed in the human liver. To detect unstable iminium ions using liquid chromatography-tandem mass spectrometry (LC-MS/MS), potassium cyanide was added to the reaction mixture as a trapping agent. Human liver and intestinal microsomes were incubated with nintedanib in the presence of NADPH to form two iminium ion metabolites on the piperazine ring. Their formation is strongly inhibited by ketoconazole, a potent cytochrome P450 (CYP) 3A4 inhibitor. Among the recombinant P450s, only CYP3A4 formed cyanide adducts. The role of CYP3A4 was supported by the positive correlation between CYP3A4 protein abundance, as determined by LC-MS-based proteomics, and the formation of cyanide adducts in 25 individual human liver microsomes. In conclusion, we have demonstrated that iminium ion metabolites are formed from nintedanib by CYP3A4 as potential reactive metabolites.
Collapse
Affiliation(s)
- Shimon Nakashima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tatsuki Fukami
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Takashi Kudo
- Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan
| | - Masataka Nakano
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan
| | - Akiko Matsui
- Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan
| | - Naoki Ishiguro
- Department of Pharmacokinetics and Nonclinical Safety, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Japan
| | - Miki Nakajima
- Drug Metabolism and Toxicology, Faculty of Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan; WPI Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
3
|
Wang S, Argikar UA, Chatzopoulou M, Cho S, Crouch RD, Dhaware D, Gu TJ, Heck CJS, Johnson KM, Kalgutkar AS, Liu J, Ma B, Miller GP, Rowley JA, Seneviratne HK, Zhang D, Khojasteh SC. Bioactivation and reactivity research advances - 2023 year in review. Drug Metab Rev 2024; 56:247-284. [PMID: 38963129 DOI: 10.1080/03602532.2024.2376023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/28/2024] [Indexed: 07/05/2024]
Abstract
Advances in the field of bioactivation have significantly contributed to our understanding and prediction of drug-induced liver injury (DILI). It has been established that many adverse drug reactions, including DILI, are associated with the formation and reactivity of metabolites. Modern methods allow us to detect and characterize these reactive metabolites in earlier stages of drug development, which helps anticipate and circumvent the potential for DILI. Improved in silico models and experimental techniques that better reflect in vivo environments are enhancing predictive capabilities for DILI risk. Further, studies on the mechanisms of bioactivation, including enzyme interactions and the role of individual genetic differences, have provided valuable insights for drug optimizations. Cumulatively, this progress is continually refining our approaches to drug safety evaluation and personalized medicine.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | | | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Rachel D Crouch
- Department of Pharmacy and Pharmaceutical Sciences, Lipscomb University College of Pharmacy, Nashville, TN, USA
| | | | - Ting-Jia Gu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Carley J S Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Groton, CT, USA
| | - Kevin M Johnson
- Drug Metabolism and Pharmacokinetics, Inotiv, Maryland Heights, MO, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, USA
| |
Collapse
|
4
|
Richter JM, Gunaga P, Yadav N, Bora RO, Bhide R, Rajugowda N, Govindrajulu K, Godesi S, Akuthota N, Rao P, Sivaraman A, Panda M, Kaspady M, Gupta A, Mathur A, Levesque PC, Gulia J, Dokania M, Ramarao M, Kole P, Chacko S, Lentz KA, Sivaprasad Lvj S, Thatipamula RP, Sridhar S, Kamble S, Govindrajan A, Soleman SI, Gordon DA, Wexler RR, Priestley ES. Discovery of BMS-986308: A Renal Outer Medullary Potassium Channel Inhibitor for the Treatment of Heart Failure. J Med Chem 2024; 67:9731-9744. [PMID: 38807539 DOI: 10.1021/acs.jmedchem.4c00893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Recent literature reports highlight the importance of the renal outer medullary potassium (ROMK) channel in renal sodium and potassium homeostasis and emphasize the potential impact that ROMK inhibitors could have as a novel mechanism diuretic in heart failure patients. A series of piperazine-based ROMK inhibitors were designed and optimized to achieve excellent ROMK potency, hERG selectivity, and ADME properties, which led to the identification of compound 28 (BMS-986308). BMS-986308 demonstrated efficacy in the volume-loaded rat diuresis model as well as promising in vitro and in vivo profiles and was therefore advanced to clinical development.
Collapse
Affiliation(s)
- Jeremy M Richter
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashantha Gunaga
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Navnath Yadav
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajesh Onkardas Bora
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Rajeev Bhide
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Nagendra Rajugowda
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Kavitha Govindrajulu
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sreenivasulu Godesi
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Nagarjuna Akuthota
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Prasanna Rao
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Aneesh Sivaraman
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoranjan Panda
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Mahammed Kaspady
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Anuradha Gupta
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Paul C Levesque
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Jyoti Gulia
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manoj Dokania
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Manjunath Ramarao
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Prashant Kole
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Silvi Chacko
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Kimberley A Lentz
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Sankara Sivaprasad Lvj
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | | | - Srikanth Sridhar
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Shyam Kamble
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Arun Govindrajan
- Biocon Bristol Myers Squibb Research Center, Syngene International Limited, Bangalore 560099, India
| | - Sharif I Soleman
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - David A Gordon
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - Ruth R Wexler
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| | - E Scott Priestley
- Bristol Myers Squibb Research & Early Development, Princeton, New Jersey 08540, United States
| |
Collapse
|
5
|
Jin L, Cheng S, Ding W, Huang J, van Eldik R, Ji L. Insight into chemically reactive metabolites of aliphatic amine pollutants: A de novo prediction strategy and case study of sertraline. ENVIRONMENT INTERNATIONAL 2024; 186:108636. [PMID: 38593692 DOI: 10.1016/j.envint.2024.108636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
The uncommon metabolic pathways of organic pollutants are easily overlooked, potentially leading to idiosyncratic toxicity. Prediction of their biotransformation associated with the toxic effects is the very purpose that this work focuses, to develop a de novo method to mechanistically predict the reactive toxicity pathways of uncommon metabolites from start aliphatic amine molecules, which employed sertraline triggered by CYP450 enzymes as a model system, as there are growing concerns about the effects on human health posed by antidepressants in the aquatic environment. This de novo prediction strategy combines computational and experimental methods, involving DFT calculations upon sequential growth, in vitro and in vivo assays, dissecting chemically reactive mechanism relevant to toxicity, and rationalizing the fundamental factors. Significantly, desaturation and debenzylation-aromatization as the emerging metabolic pathways of sertraline have been elucidated, with the detection of DNA adducts of oxaziridine metabolite in mice, highlighting the potential reactive toxicity. Molecular orbital analysis supports the reactivity preference for toxicological-relevant C-N desaturation over N-hydroxylation of sertraline, possibly extended to several other aliphatic amines based on the Bell-Evans-Polanyi principle. It was further validated toward some other wide-concerned aliphatic amine pollutants involving atrazine, ε-caprolactam, 6PPD via in silico and in vitro assays, thereby constituting a complete path for de novo prediction from case study to general applications.
Collapse
Affiliation(s)
- Lingmin Jin
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, China
| | - Shiyang Cheng
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, China.
| | - Wen Ding
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, China
| | - Jingru Huang
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, China
| | - Rudi van Eldik
- Department of Chemistry and Pharmacy, University of Erlangen-Nuremberg, Egerlandstr. 1, 91058 Erlangen, Germany; Faculty of Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland
| | - Li Ji
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, China.
| |
Collapse
|
6
|
Wodtke R, Laube M, Hauser S, Meister S, Ludwig FA, Fischer S, Kopka K, Pietzsch J, Löser R. Preclinical evaluation of an 18F-labeled N ε-acryloyllysine piperazide for covalent targeting of transglutaminase 2. EJNMMI Radiopharm Chem 2024; 9:1. [PMID: 38165538 PMCID: PMC10761660 DOI: 10.1186/s41181-023-00231-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/15/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Transglutaminase 2 (TGase 2) is a multifunctional protein and has a prominent role in various (patho)physiological processes. In particular, its transamidase activity, which is rather latent under physiological conditions, gains importance in malignant cells. Thus, there is a great need of theranostic probes for targeting tumor-associated TGase 2, and targeted covalent inhibitors appear to be particularly attractive as vector molecules. Such an inhibitor, equipped with a radionuclide suitable for noninvasive imaging, would be supportive for answering the general question on the possibility for functional characterization of tumor-associated TGase 2. For this purpose, the recently developed 18F-labeled Nε-acryloyllysine piperazide [18F]7b, which is a potent and selective irreversible inhibitor of TGase 2, was subject to a detailed radiopharmacological characterization herein. RESULTS An alternative radiosynthesis of [18F]7b is presented, which demands less than 300 µg of the respective trimethylammonio precursor per synthesis and provides [18F]7b in good radiochemical yields (17 ± 7%) and high (radio)chemical purities (≥ 99%). Ex vivo biodistribution studies in healthy mice at 5 and 60 min p.i. revealed no permanent enrichment of 18F-activity in tissues with the exception of the bone tissue. In vivo pretreatment with ketoconazole and in vitro murine liver microsome studies complemented by mass spectrometric analysis demonstrated that bone uptake originates from metabolically released [18F]fluoride. Further metabolic transformations of [18F]7b include mono-hydroxylation and glucuronidation. Based on blood sampling data and liver microsome experiments, pharmacokinetic parameters such as plasma and intrinsic clearance were derived, which substantiated the apparently rapid distribution of [18F]7b in and elimination from the organisms. A TGase 2-mediated uptake of [18F]7b in different tumor cell lines could not be proven. Moreover, evaluation of [18F]7b in melanoma tumor xenograft models based on A375-hS100A4 (TGase 2 +) and MeWo (TGase 2 -) cells by ex vivo biodistribution and PET imaging studies were not indicative for a specific targeting. CONCLUSION [18F]7b is a valuable radiometric tool to study TGase 2 in vitro under various conditions. However, its suitability for targeting tumor-associated TGase 2 is strongly limited due its unfavorable pharmacokinetic properties as demonstrated in rodents. Consequently, from a radiochemical perspective [18F]7b requires appropriate structural modifications to overcome these limitations.
Collapse
Affiliation(s)
- Robert Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany.
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, 04318, Leipzig, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328, Dresden, Germany.
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069, Dresden, Germany.
| |
Collapse
|
7
|
Gunduz M, Argikar UA, Cirello AL, Brown AP, Bonazzi S, Walles M. Species-specific Bioactivation of Morpholines as a Causative of Drug Induced Liver Injury Observed in Monkeys. DRUG METABOLISM AND BIOANALYSIS LETTERS 2024; 17:13-22. [PMID: 38047363 DOI: 10.2174/0118723128260455231104180653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Everolimus, an allosteric mechanistic target of rapamycin (mTOR) inhibitor, recently demonstrated the therapeutic value of mTOR inhibitors for Central Nervous System (CNS) indications driven by hyperactivation of mTOR. A newer, potent brain-penetrant analog of everolimus, referred to as (1) in this manuscript [(S)-3-methyl-4-(7-((R)-3-methylmorpholino)-2- (thiazol-4-yl)-3H-imidazo[4,5-b]pyridin-5-yl)morpholine,(1)] catalytically inhibits mTOR function in the brain and increases the lifespan of mice with neuronal mTOR hyperactivation. INTRODUCTION Early evaluation of the safety of 1 was conducted in cynomolgus monkeys in which oral doses were administered to three animals in a rising-dose fashion (from 2 to 30 mg/kg/day). 1 produced severe toxicity including the evidence of hepatic toxicity, along with non-dose proportional increases in drug exposure. Investigations of cross-species hepatic bioactivation of 1 were conducted to assess whether the formation of reactive drug metabolites was associated with the mechanism of liver toxicity. METHODS 1 contained two morpholine rings known as structural alerts and can potentially form reactive intermediates through oxidative metabolism. Bioactivation of 1 was investigated in rat, human and monkey liver microsomes fortified with trapping agents such as methoxylamine or potassium cyanide. RESULTS Our results suggest that bioactivation of the morpholine moieties to reactive intermediates may have been involved in the mechanism of liver toxicity observed with 1. Aldehyde intermediates trappable by methoxylamine were identified in rat and monkey liver microsomal studies. In addition, a total of four cyano conjugates arising from the formation of iminium ion intermediates were observed and identified. These findings may potentially explain the observed monkey toxicity. Interestingly, methoxylamine or cyano adducts of 1 were not observed in human liver microsomes. CONCLUSION The bioactivation of 1 appears to be species-specific. Circumstantial evidence for the toxicity derived from 1 point to the formation of iminium ion intermediates trappable by cyanide in monkey liver microsomes. The cyano conjugates were only observed in monkey liver microsomes, potentially pointing to cause at least the hepatotoxicity observed in monkeys. In contrast, methoxylamine conjugates were detected in both rat and monkey liver microsomes, with only a trace amount in human liver microsomes. Cyano conjugates were not observed in human liver microsomes, challenging the team on the drugability and progressivity of 1 through drug development. The mechanisms for drug-induced liver toxicity are multifactorial. These results are highly suggestive that the iminium ion may be an important component in the mechanism of liver toxicity 1 observed in the monkey.
Collapse
Affiliation(s)
- Mithat Gunduz
- Department of Pharmacokinetic Sciences, Global Biotransformation, Novartis Biomedical Research, Inc., Cambridge, MA, US
| | - Upendra A Argikar
- Department of Pharmacokinetic Sciences, Global Biotransformation, Novartis Biomedical Research, Inc., Cambridge, MA, US
- Bill & Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Amanda L Cirello
- Department of Pharmacokinetic Sciences, Global Biotransformation, Novartis Biomedical Research, Inc., Cambridge, MA, US
| | - Alan P Brown
- Department of Preclinical Safety, Novartis Biomedical Research, Inc., Translational Medicine, Cambridge, MA, USA
| | - Simone Bonazzi
- Department of Global Discovery Chemistry, Novartis BioMedical Research, Inc., Cambridge, MA, USA
| | - Markus Walles
- Department of Pharmacokinetic Sciences, Global Biotransformation, Novartis Biomedical Research, Inc., Basel, Switzerland
| |
Collapse
|
8
|
He C, Mao Y, Wan H. Preclinical evaluation of chemically reactive metabolites and mitigation of bioactivation in drug discovery. Drug Discov Today 2023; 28:103621. [PMID: 37201781 DOI: 10.1016/j.drudis.2023.103621] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/20/2023]
Abstract
The formation of reactive metabolites (RMs) is thought to be one of the pathogeneses for some idiosyncratic adverse drug reactions (IADRs) which are considered one of the leading causes of some drug attritions and/or recalls. Minimizing or eliminating the formation of RMs via chemical modification is a useful tactic to reduce the risk of IADRs and time-dependent inhibition (TDI) of cytochrome P450 enzymes (CYPs). The RMs should be carefully handled before making a go-no-go decision. Herein, we highlight the role of RMs in the occurrence of IADRs and CYP TDI, the risk of structural alerts, the approaches of RM assessment at the discovery stage and strategies to minimize or eliminate RM liability. Finally, some considerations for developing a RM-positive drug candidate are suggested.
Collapse
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical, No. 279 Wenjing Road, Shanghai 200245, China.
| | - Yuchang Mao
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical, No. 279 Wenjing Road, Shanghai 200245, China
| | - Hong Wan
- Department of DMPK/Bioanalysis, Shanghai Medicilon, No. 585 Chuanda Road, Shanghai 201299, China.
| |
Collapse
|
9
|
Sharma R, Dowling MS, Futatsugi K, Kalgutkar AS. Mitigating a Bioactivation Liability with an Azetidine-Based Inhibitor of Diacylglycerol Acyltransferase 2 (DGAT2) En Route to the Discovery of the Clinical Candidate Ervogastat. Chem Res Toxicol 2023. [PMID: 37148271 DOI: 10.1021/acs.chemrestox.3c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We recently disclosed SAR studies on systemically acting, amide-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2) that addressed metabolic liabilities with the liver-targeted DGAT2 inhibitor PF-06427878. Despite strategic placement of a nitrogen atom in the dialkoxyaromatic ring in PF-06427878 to evade oxidative O-dearylation, metabolic intrinsic clearance remained high due to extensive piperidine ring oxidation as exemplified with compound 1. Piperidine ring modifications through alternate N-linked heterocyclic ring/spacer combination led to azetidine 2 that demonstrated lower intrinsic clearance. However, 2 underwent a facile cytochrome P450 (CYP)-mediated α-carbon oxidation followed by azetidine ring scission, resulting in the formation of ketone (M2) and aldehyde (M6) as stable metabolites in NADPH-supplemented human liver microsomes. Inclusion of GSH or semicarbazide in microsomal incubations led to the formation of Cys-Gly-thiazolidine (M3), Cys-thiazolidine (M5), and semicarbazone (M7) conjugates, which were derived from reaction of the nucleophilic trapping agents with aldehyde M6. Metabolites M2 and M5 were biosynthesized from NADPH- and l-cysteine-fortified human liver microsomal incubations with 2, and proposed metabolite structures were verified using one- and two-dimensional NMR spectroscopy. Replacement of the azetidine substituent with a pyridine ring furnished 8, which mitigated the formation of the electrophilic aldehyde metabolite, and was a more potent DGAT2 inhibitor than 2. Further structural refinements in 8, specifically introducing amide bond substituents with greater metabolic stability, led to the discovery of PF-06865571 (ervogastat) that is currently in phase 2 clinical trials for the treatment of nonalcoholic steatohepatitis.
Collapse
Affiliation(s)
- Raman Sharma
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Matthew S Dowling
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kentaro Futatsugi
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, 1 Portland St, Cambridge, Massachusetts 02139, United States
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development, and Medical, 1 Portland St, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
10
|
Jackson KD, Argikar UA, Cho S, Crouch RD, Driscoll JP, Heck C, King L, Maw HH, Miller GP, Seneviratne HK, Wang S, Wei C, Zhang D, Khojasteh SC. Bioactivation and Reactivity Research Advances - 2021 year in review. Drug Metab Rev 2022; 54:246-281. [PMID: 35876116 DOI: 10.1080/03602532.2022.2097254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This year's review on bioactivation and reactivity began as a part of the annual review on biotransformation and bioactivation led by Cyrus Khojasteh (Khojasteh et al., 2021, 2020, 2019, 2018, 2017; Baillie et al., 2016). Increased contributions from experts in the field led to the development of a stand alone edition for the first time this year focused specifically on bioactivation and reactivity. Our objective for this review is to highlight and share articles which we deem influential and significant regarding the development of covalent inhibitors, mechanisms of reactive metabolite formation, enzyme inactivation, and drug safety. Based on the selected articles, we created two sections: (1) reactivity and enzyme inactivation, and (2) bioactivation mechanisms and safety (Table 1). Several biotransformation experts have contributed to this effort from academic and industry settings.
Collapse
Affiliation(s)
- Klarissa D Jackson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill & Melinda Gates Medical Research Institute, Cambridge, MA, 02139, USA
| | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Rachel D Crouch
- Department of Pharmaceutical Sciences, Lipscomb University College of Pharmacy and Health Sciences, Nashville, TN, 37203, USA
| | - James P Driscoll
- Department of Drug Metabolism and Pharmacokinetics. Bristol Myers Squibb, Brisbane, CA, 94005, USA
| | - Carley Heck
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut, USA
| | - Lloyd King
- Department of DMPK, UCB Biopharma UK, 216 Bath Road, Slough, SL1 3WE, UK
| | - Hlaing Holly Maw
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, 06877, USA
| | - Grover P Miller
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, 4301 W Markham St Slot 516, Little Rock, Arkansas, 72205, USA
| | - Herana Kamal Seneviratne
- Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Cong Wei
- Drug Metabolism & Pharmacokinetics, Biogen Inc., Cambridge, MA, 02142, USA
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, MS412a, South San Francisco, CA, 94080, USA
| |
Collapse
|
11
|
Wang JY, Li JX, Ning J, Huo XK, Yu ZL, Tian Y, Zhang BJ, Wang Y, Sa D, Li YC, Lv X, Ma XC. Human cytochrome P450 3A-mediated two-step oxidation metabolism of dimethomorph: Implications in the mechanism-based enzyme inactivation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 822:153585. [PMID: 35121040 DOI: 10.1016/j.scitotenv.2022.153585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
Dimethomorph (DMM), an effective and broad-spectrum fungicide applied in agriculture, is toxic to environments and living organisms due to the hazardous nature of its toxic residues. This study aims to investigate the human cytochrome P450 enzyme (CYP)-mediated oxidative metabolism of DMM by combining experimental and computational approaches. Dimethomorph was metabolized predominantly through a two-step oxidation process mediated by CYPs, and CYP3A was identified as the major contributor to DMM sequential oxidative metabolism. Meanwhile, DMM elicited the mechanism-based inactivation (MBI) of CYP3A in a suicide manner, and the iminium ion and epoxide reactive intermediates generated in DMM metabolism were identified as the culprits of MBI. Furthermore, three common pesticides, prochloraz (PCZ), difenoconazole (DFZ) and chlorothalonil (CTL), could significantly inhibit CYP3A-mediated DMM metabolism, and consequently trigger elevated exposure to DMM in vivo. Computational studies elucidated that the differentiation effects in charge distribution and the interaction pattern played crucial roles in DMM-induced MBI of CYP3A4 during sequential oxidative metabolism. Collectively, this study provided a global view of the two-step metabolic activation process of DMM mediated by CYP3A, which was beneficial for elucidating the environmental fate and toxicological mechanism of DMM in humans from a new perspective.
Collapse
Affiliation(s)
- Jia-Yue Wang
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China; Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian 116000, Liaoning, China; Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Jing-Xin Li
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China; School of Public Health, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Jing Ning
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Xiao-Kui Huo
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Zhen-Long Yu
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Yan Tian
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Bao-Jing Zhang
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Yan Wang
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Deng Sa
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Ya-Chen Li
- School of Public Health, Dalian Medical University, Dalian 116000, Liaoning, China
| | - Xia Lv
- College of Integrative Medicine, Dalian Medical University, Dalian 116000, Liaoning, China.
| | - Xiao-Chi Ma
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian 116000, Liaoning, China.
| |
Collapse
|
12
|
Tang LWT, Wei W, Verma RK, Koh SK, Zhou L, Fan H, Chan ECY. Direct and Sequential Bioactivation of Pemigatinib to Reactive Iminium Ion Intermediates Culminate in Mechanism-Based Inactivation of Cytochrome P450 3A. Drug Metab Dispos 2022; 50:529-540. [DOI: 10.1124/dmd.121.000804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/01/2022] [Indexed: 11/22/2022] Open
|
13
|
Duarte de Almeida L, Bourriquen F, Junge K, Beller M. Catalytic Formal Hydroamination of Allylic Alcohols Using Manganese PNP-Pincer Complexes. Adv Synth Catal 2021; 363:4177-4181. [PMID: 34690626 PMCID: PMC8519145 DOI: 10.1002/adsc.202100081] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/05/2021] [Indexed: 01/09/2023]
Abstract
Several manganese-PNP pincer catalysts for the formal hydroamination of allylic alcohols are presented. The resulting γ-amino alcohols are selectively obtained in high yields applying Mn-1 in a tandem process under mild conditions.
Collapse
Affiliation(s)
| | - Florian Bourriquen
- Leibniz-Institut für Katalyse e.V.Albert-Einstein-Str. 29a18059RostockGermany
| | - Kathrin Junge
- Leibniz-Institut für Katalyse e.V.Albert-Einstein-Str. 29a18059RostockGermany
| | - Matthias Beller
- Leibniz-Institut für Katalyse e.V.Albert-Einstein-Str. 29a18059RostockGermany
| |
Collapse
|
14
|
Huang J, Chen Z, Wu J. Recent Progress in Methyl-Radical-Mediated Methylation or Demethylation Reactions. ACS Catal 2021. [DOI: 10.1021/acscatal.1c02010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Jiapian Huang
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education, and Jiangxi Key Laboratory of Green Chemistry, College of Chemistry & Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, P. R. China
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, 1139 Shifu Avenue, Taizhou 318000, China
| | - Zhiyuan Chen
- Key Laboratory of Functional Small Organic Molecules, Ministry of Education, and Jiangxi Key Laboratory of Green Chemistry, College of Chemistry & Chemical Engineering, Jiangxi Normal University, 99 Ziyang Road, Nanchang, Jiangxi 330022, P. R. China
| | - Jie Wu
- School of Pharmaceutical and Materials Engineering & Institute for Advanced Studies, Taizhou University, 1139 Shifu Avenue, Taizhou 318000, China
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
15
|
In Vitro Metabolism of Donepezil in Liver Microsomes Using Non-Targeted Metabolomics. Pharmaceutics 2021; 13:pharmaceutics13070936. [PMID: 34201744 PMCID: PMC8309179 DOI: 10.3390/pharmaceutics13070936] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/17/2022] Open
Abstract
Donepezil is a reversible acetylcholinesterase inhibitor that is currently the most commonly prescribed drug for the treatment of Alzheimer’s disease. In general, donepezil is known as a safe and well-tolerated drug, and it was not associated with liver abnormalities in several clinical trials. However, rare cases of drug-related liver toxicity have been reported since it has become commercially available. Few studies have investigated the metabolic profile of donepezil, and the mechanism of liver damage caused by donepezil has not been elucidated. In this study, the in vitro metabolism of donepezil was investigated using liquid chromatography–tandem mass spectrometry based on a non-targeted metabolomics approach. To identify metabolites, the data were subjected to multivariate data analysis and molecular networking. A total of 21 donepezil metabolites (17 in human liver microsomes, 21 in mice liver microsomes, and 17 in rat liver microsomes) were detected including 14 newly identified metabolites. One potential reactive metabolite was identified in rat liver microsomal incubation samples. Metabolites were formed through four major metabolic pathways: (1) O-demethylation, (2) hydroxylation, (3) N-oxidation, and (4) N-debenzylation. This study indicates that a non-targeted metabolomics approach combined with molecular networking is a reliable tool to identify and detect unknown drug metabolites.
Collapse
|
16
|
Bauer MR, Di Fruscia P, Lucas SCC, Michaelides IN, Nelson JE, Storer RI, Whitehurst BC. Put a ring on it: application of small aliphatic rings in medicinal chemistry. RSC Med Chem 2021; 12:448-471. [PMID: 33937776 PMCID: PMC8083977 DOI: 10.1039/d0md00370k] [Citation(s) in RCA: 177] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/18/2020] [Indexed: 12/15/2022] Open
Abstract
Aliphatic three- and four-membered rings including cyclopropanes, cyclobutanes, oxetanes, azetidines and bicyclo[1.1.1]pentanes have been increasingly exploited in medicinal chemistry for their beneficial physicochemical properties and applications as functional group bioisosteres. This review provides a historical perspective and comparative up to date overview of commonly applied small rings, exemplifying key principles with recent literature examples. In addition to describing the merits and advantages of each ring system, potential hazards and liabilities are also illustrated and explained, including any significant chemical or metabolic stability and toxicity risks.
Collapse
Affiliation(s)
- Matthias R Bauer
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge UK
| | - Paolo Di Fruscia
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge UK
| | - Simon C C Lucas
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge UK
| | | | - Jennifer E Nelson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge UK
| | - R Ian Storer
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca Cambridge UK
| | | |
Collapse
|
17
|
Kannt A, Rajagopal S, Hallur MS, Swamy I, Kristam R, Dhakshinamoorthy S, Czech J, Zech G, Schreuder H, Ruf S. Novel Inhibitors of Nicotinamide- N-Methyltransferase for the Treatment of Metabolic Disorders. Molecules 2021; 26:molecules26040991. [PMID: 33668468 PMCID: PMC7918612 DOI: 10.3390/molecules26040991] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/01/2021] [Accepted: 02/06/2021] [Indexed: 12/29/2022] Open
Abstract
Nicotinamide-N-methyltransferase (NNMT) is a cytosolic enzyme catalyzing the transfer of a methyl group from S-adenosyl-methionine (SAM) to nicotinamide (Nam). It is expressed in many tissues including the liver, adipose tissue, and skeletal muscle. Its expression in several cancer cell lines has been widely discussed in the literature, and recent work established a link between NNMT expression and metabolic diseases. Here we describe our approach to identify potent small molecule inhibitors of NNMT featuring different binding modes as elucidated by X-ray crystallographic studies.
Collapse
Affiliation(s)
- Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology-ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany;
| | - Sridharan Rajagopal
- Jubilant Biosys Limited, #96, Industrial Suburb, 2nd Stage Yeshwanthpur, Bangalore 560022, India; (S.R.); (M.S.H.); (I.S.); (R.K.); (S.D.)
| | - Mahanandeesha S. Hallur
- Jubilant Biosys Limited, #96, Industrial Suburb, 2nd Stage Yeshwanthpur, Bangalore 560022, India; (S.R.); (M.S.H.); (I.S.); (R.K.); (S.D.)
| | - Indu Swamy
- Jubilant Biosys Limited, #96, Industrial Suburb, 2nd Stage Yeshwanthpur, Bangalore 560022, India; (S.R.); (M.S.H.); (I.S.); (R.K.); (S.D.)
| | - Rajendra Kristam
- Jubilant Biosys Limited, #96, Industrial Suburb, 2nd Stage Yeshwanthpur, Bangalore 560022, India; (S.R.); (M.S.H.); (I.S.); (R.K.); (S.D.)
| | - Saravanakumar Dhakshinamoorthy
- Jubilant Biosys Limited, #96, Industrial Suburb, 2nd Stage Yeshwanthpur, Bangalore 560022, India; (S.R.); (M.S.H.); (I.S.); (R.K.); (S.D.)
| | - Joerg Czech
- Sanofi-Aventis Deutschland Gmbh, Industriepark Hoechst, 65926 Frankfurt am Main, Germany; (J.C.); (G.Z.); (H.S.)
| | - Gernot Zech
- Sanofi-Aventis Deutschland Gmbh, Industriepark Hoechst, 65926 Frankfurt am Main, Germany; (J.C.); (G.Z.); (H.S.)
| | - Herman Schreuder
- Sanofi-Aventis Deutschland Gmbh, Industriepark Hoechst, 65926 Frankfurt am Main, Germany; (J.C.); (G.Z.); (H.S.)
| | - Sven Ruf
- Sanofi-Aventis Deutschland Gmbh, Industriepark Hoechst, 65926 Frankfurt am Main, Germany; (J.C.); (G.Z.); (H.S.)
- Correspondence:
| |
Collapse
|
18
|
Newman AH, Ku T, Jordan CJ, Bonifazi A, Xi ZX. New Drugs, Old Targets: Tweaking the Dopamine System to Treat Psychostimulant Use Disorders. Annu Rev Pharmacol Toxicol 2021; 61:609-628. [PMID: 33411583 PMCID: PMC9341034 DOI: 10.1146/annurev-pharmtox-030220-124205] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The abuse of illicit psychostimulants such as cocaine and methamphetamine continues to pose significant health and societal challenges. Despite considerable efforts to develop medications to treat psychostimulant use disorders, none have proven effective, leaving an underserved patient population and unanswered questions about what mechanism(s) of action should be targeted for developing pharmacotherapies. As both cocaine and methamphetamine rapidly increase dopamine (DA) levels in mesolimbic brain regions, leading to euphoria that in some can lead to addiction, targets in which this increased dopaminergic tone may be mitigated have been explored. Further, understanding and targeting mechanisms underlying relapse are fundamental to the success of discovering medications that reduce the reinforcing effects of the drug of abuse, decrease the negative reinforcement or withdrawal/negative affect that occurs during abstinence, or both. Atypical inhibitors of the DA transporter and partial agonists/antagonists at DA D3 receptors are described as two promising targets for future drug development.
Collapse
Affiliation(s)
- Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Therese Ku
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA;
| |
Collapse
|
19
|
Zeng X, Yan W, Zacate SB, Cai A, Wang Y, Yang D, Yang K, Liu W. Copper-Catalyzed Deaminative Difluoromethylation. Angew Chem Int Ed Engl 2020; 59:16398-16403. [PMID: 32495485 DOI: 10.1002/anie.202006048] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/01/2020] [Indexed: 12/17/2022]
Abstract
The difluoromethyl group (CF2 H) is considered to be a lipophilic and metabolically stable bioisostere of an amino (NH2 ) group. Therefore, methods that can rapidly convert an NH2 group into a CF2 H group would be of great value to medicinal chemistry. We report herein an efficient Cu-catalyzed approach for the conversion of alkyl pyridinium salts, which can be readily synthesized from the corresponding alkyl amines, to their alkyl difluoromethane analogues. This method tolerates a broad range of functional groups and can be applied to the late-stage modification of complex amino-containing pharmaceuticals.
Collapse
Affiliation(s)
- Xiaojun Zeng
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Wenhao Yan
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Samson B Zacate
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Aijie Cai
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Yufei Wang
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Dongqi Yang
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Kundi Yang
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| | - Wei Liu
- Department of Chemistry and Biochemistry, Miami University, Oxford, OH, 45056, USA
| |
Collapse
|
20
|
Zeng X, Yan W, Zacate SB, Cai A, Wang Y, Yang D, Yang K, Liu W. Copper‐Catalyzed Deaminative Difluoromethylation. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Xiaojun Zeng
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Wenhao Yan
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Samson B. Zacate
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Aijie Cai
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Yufei Wang
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Dongqi Yang
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Kundi Yang
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| | - Wei Liu
- Department of Chemistry and BiochemistryMiami University Oxford OH 45056 USA
| |
Collapse
|
21
|
Linhares BM, Grembecka J, Cierpicki T. Targeting epigenetic protein-protein interactions with small-molecule inhibitors. Future Med Chem 2020; 12:1305-1326. [PMID: 32551894 PMCID: PMC7421387 DOI: 10.4155/fmc-2020-0082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Epigenetic protein-protein interactions (PPIs) play essential roles in regulating gene expression, and their dysregulations have been implicated in many diseases. These PPIs are comprised of reader domains recognizing post-translational modifications on histone proteins, and of scaffolding proteins that maintain integrities of epigenetic complexes. Targeting PPIs have become focuses for development of small-molecule inhibitors and anticancer therapeutics. Here we summarize efforts to develop small-molecule inhibitors targeting common epigenetic PPI domains. Potent small molecules have been reported for many domains, yet small domains that recognize methylated lysine side chains on histones are challenging in inhibitor development. We posit that the development of potent inhibitors for difficult-to-prosecute epigenetic PPIs may be achieved by interdisciplinary approaches and extensive explorations of chemical space.
Collapse
Affiliation(s)
- Brian M Linhares
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tomasz Cierpicki
- Biophysics Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
22
|
Yi X, Lei S, Liu W, Che F, Yu C, Liu X, Wang Z, Zhou X, Zhang Y. Copper-Catalyzed Radical N-Demethylation of Amides Using N-Fluorobenzenesulfonimide as an Oxidant. Org Lett 2020; 22:4583-4587. [DOI: 10.1021/acs.orglett.0c00863] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xuewen Yi
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Siyu Lei
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Wangsheng Liu
- Division of Chemistry & Biological Chemistry, School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Fengrui Che
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Chunzheng Yu
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Xuesong Liu
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Zonghua Wang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Xin Zhou
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yuexia Zhang
- College of Chemistry and Chemical Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
23
|
Slack RD, Ku T, Cao J, Giancola J, Bonifazi A, Loland CJ, Gadiano A, Lam J, Rais R, Slusher BS, Coggiano M, Tanda G, Newman AH. Structure-Activity Relationships for a Series of (Bis(4-fluorophenyl)methyl)sulfinyl Alkyl Alicyclic Amines at the Dopamine Transporter: Functionalizing the Terminal Nitrogen Affects Affinity, Selectivity, and Metabolic Stability. J Med Chem 2020; 63:2343-2357. [PMID: 31661268 PMCID: PMC9617638 DOI: 10.1021/acs.jmedchem.9b01188] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Atypical dopamine transporter (DAT) inhibitors have shown therapeutic potential in preclinical models of psychostimulant abuse. In rats, 1-(4-(2-((bis(4-fluorophenyl)methyl)sulfinyl)ethyl)-piperazin-1-yl)-propan-2-ol (3b) was effective in reducing the reinforcing effects of both cocaine and methamphetamine but did not exhibit psychostimulant behaviors itself. While further development of 3b is ongoing, diastereomeric separation, as well as improvements in potency and pharmacokinetics were desirable for discovering pipeline drug candidates. Thus, a series of bis(4-fluorophenyl)methyl)sulfinyl)alkyl alicyclic amines, where the piperazine-2-propanol scaffold was modified, were designed, synthesized, and evaluated for binding affinities at DAT, as well as the serotonin transporter and σ1 receptors. Within the series, 14a showed improved DAT affinity (Ki = 23 nM) over 3b (Ki = 230 nM), moderate metabolic stability in human liver microsomes, and a hERG/DAT affinity ratio = 28. While 14a increased locomotor activity relative to vehicle, it was significantly lower than activity produced by cocaine. These results support further investigation of 14a as a potential treatment for psychostimulant use disorders.
Collapse
Affiliation(s)
- Rachel D. Slack
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Therese Ku
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Jianjing Cao
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - JoLynn Giancola
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Claus J. Loland
- Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Alexandra Gadiano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Jenny Lam
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Rana Rais
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, 855 North Wolfe Street, Baltimore, MD 21205, United States
| | - Mark Coggiano
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Gianluigi Tanda
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, United States
| |
Collapse
|
24
|
Is there enough evidence to classify cycloalkyl amine substituents as structural alerts? Biochem Pharmacol 2020; 174:113796. [PMID: 31926938 DOI: 10.1016/j.bcp.2020.113796] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/07/2020] [Indexed: 12/21/2022]
Abstract
Basic amine substituents provide several pharmacokinetic benefits relative to acidic and neutral functional groups, and have been extensively utilized as substituents of choice in drug design. On occasions, basic amines have been associated with off-target pharmacology via interactions with aminergic G-protein coupled receptors, ion-channels, kinases, etc. Structural features associated with the promiscuous nature of basic amines have been well-studied, and can be mitigated in a preclinical drug discovery environment. In addition to the undesirable secondary pharmacology, α-carbon oxidation of certain secondary or tertiary cycloalkyl amines can generate electrophilic iminium and aldehyde metabolites, potentially capable of covalent adduction to proteins or DNA. Consequently, cycloalkyl amines have been viewed as structural alerts (SAs), analogous to functional groups such as anilines, furans, thiophenes, etc., which are oxidized to reactive metabolites that generate immunogenic haptens by covalently binding to host proteins. Detailed survey of the literature, however, suggests that cases where preclinical or clinical toxicity has been explicitly linked to the metabolic activation of a cycloalkyl amine group are extremely rare. Moreover, there is a distinct possibility for the formation of electrophilic iminium/amino-aldehyde metabolites with numerous cycloalkyl amine-containing marketed drugs, since stable ring cleavage products have been characterized as metabolites in human mass balance studies. In the present work, a critical analysis of the evidence for and against the role of iminium ions/aldehydes as mediators of toxicity is discussed with a special emphasis on often time overlooked detoxication pathways of these reactive species to innocuous metabolites.
Collapse
|
25
|
Abstract
Cumulative research over several decades has implicated the involvement of reactive metabolites in many idiosyncratic adverse drug reactions (IADRs). Consequently, "avoidance" strategies have been inserted into drug discovery paradigms, which include the exclusion of structural alerts and possible termination of reactive metabolite-positive compounds. Several noteworthy examples where reactive metabolite-related liabilities have been resolved through structure-metabolism studies are presented herein. Considerable progress has also been made in addressing the limitations of the avoidance strategy and further refining the process of managing reactive metabolite issues in drug development. These efforts primarily stemmed from the observation that numerous drugs, which contain structural alerts and/or form reactive metabolites, are devoid of ADRs. The Perspective also dwells into an analysis of the structural alert/reactive metabolite concept with a discussion of risk mitigation tactics to support the progression of reactive metabolite-positive drug candidates.
Collapse
Affiliation(s)
- Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
26
|
Dalvie D, Di L. Aldehyde oxidase and its role as a drug metabolizing enzyme. Pharmacol Ther 2019; 201:137-180. [PMID: 31128989 DOI: 10.1016/j.pharmthera.2019.05.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/27/2019] [Indexed: 11/29/2022]
Abstract
Aldehyde oxidase (AO) is a cytosolic enzyme that belongs to the family of structurally related molybdoflavoproteins like xanthine oxidase (XO). The enzyme is characterized by broad substrate specificity and marked species differences. It catalyzes the oxidation of aromatic and aliphatic aldehydes and various heteroaromatic rings as well as reduction of several functional groups. The references to AO and its role in metabolism date back to the 1950s, but the importance of this enzyme in the metabolism of drugs has emerged in the past fifteen years. Several reviews on the role of AO in drug metabolism have been published in the past decade indicative of the growing interest in the enzyme and its influence in drug metabolism. Here, we present a comprehensive monograph of AO as a drug metabolizing enzyme with emphasis on marketed drugs as well as other xenobiotics, as substrates and inhibitors. Although the number of drugs that are primarily metabolized by AO are few, the impact of AO on drug development has been extensive. We also discuss the effect of AO on the systemic exposure and clearance these clinical candidates. The review provides a comprehensive analysis of drug discovery compounds involving AO with the focus on developmental candidates that were reported in the past five years with regards to pharmacokinetics and toxicity. While there is only one known report of AO-mediated clinically relevant drug-drug interaction (DDI), a detailed description of inhibitors and inducers of AO known to date has been presented here and the potential risks associated with DDI. The increasing recognition of the importance of AO has led to significant progress in predicting the site of AO-mediated metabolism using computational methods. Additionally, marked species difference in expression of AO makes it is difficult to predict human clearance with high confidence. The progress made towards developing in vivo, in vitro and in silico approaches for predicting AO metabolism and estimating human clearance of compounds that are metabolized by AO have also been discussed.
Collapse
Affiliation(s)
- Deepak Dalvie
- Drug Metabolism and Pharmacokinetics, Celgene Corporation, 10300, Campus Point Drive, San Diego, CA 92121, USA.
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, UK
| |
Collapse
|
27
|
Roldán R, Hernández K, Joglar J, Bujons J, Parella T, Fessner W, Clapés P. Aldolase-Catalyzed Asymmetric Synthesis of N-Heterocycles by Addition of Simple Aliphatic Nucleophiles to Aminoaldehydes. Adv Synth Catal 2019; 361:2673-2687. [PMID: 31680790 PMCID: PMC6813633 DOI: 10.1002/adsc.201801530] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/22/2019] [Indexed: 11/08/2022]
Abstract
Nitrogen heterocycles are structural motifs found in many bioactive natural products and of utmost importance in pharmaceutical drug development. In this work, a stereoselective synthesis of functionalized N-heterocycles was accomplished in two steps, comprising the biocatalytic aldol addition of ethanal and simple aliphatic ketones such as propanone, butanone, 3-pentanone, cyclobutanone, and cyclopentanone to N-Cbz-protected aminoaldehydes using engineered variants of d-fructose-6-phosphate aldolase from Escherichia coli (FSA) or 2-deoxy-d-ribose-5-phosphate aldolase from Thermotoga maritima (DERA Tma ) as catalysts. FSA catalyzed most of the additions of ketones while DERA Tma was restricted to ethanal and propanone. Subsequent treatment with hydrogen in the presence of palladium over charcoal, yielded low-level oxygenated N-heterocyclic derivatives of piperidine, pyrrolidine and N-bicyclic structures bearing fused cyclobutane and cyclopentane rings, with stereoselectivities of 96-98 ee and 97:3 dr in isolated yields ranging from 35 to 79%.
Collapse
Affiliation(s)
- Raquel Roldán
- Dept. Biological Chemistry. Instituto de Química Avanzada de Cataluña IQAC-CSICJordi Girona 18–2608034BarcelonaSpain
| | - Karel Hernández
- Dept. Biological Chemistry. Instituto de Química Avanzada de Cataluña IQAC-CSICJordi Girona 18–2608034BarcelonaSpain
| | - Jesús Joglar
- Dept. Biological Chemistry. Instituto de Química Avanzada de Cataluña IQAC-CSICJordi Girona 18–2608034BarcelonaSpain
| | - Jordi Bujons
- Dept. Biological Chemistry. Instituto de Química Avanzada de Cataluña IQAC-CSICJordi Girona 18–2608034BarcelonaSpain
| | - Teodor Parella
- Servei de Ressonància Magnètica Nuclear.Universitat Autònoma de BarcelonaBellaterraSpain
| | - Wolf‐Dieter Fessner
- Institut für Organische Chemie und BiochemieTechnische Universität DarmstadtPetersenstraße 22D-64287DarmstadtGermany
| | - Pere Clapés
- Dept. Biological Chemistry. Instituto de Química Avanzada de Cataluña IQAC-CSICJordi Girona 18–2608034BarcelonaSpain
| |
Collapse
|
28
|
Yao K, Yuan Q, Qu X, Liu Y, Liu D, Zhang W. Pd-catalyzed asymmetric allylic substitution cascade using α-(pyridin-1-yl)-acetamides formed in situ as nucleophiles. Chem Sci 2019; 10:1767-1772. [PMID: 30842843 PMCID: PMC6369409 DOI: 10.1039/c8sc04626c] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/01/2018] [Indexed: 12/13/2022] Open
Abstract
A Pd-catalyzed asymmetric allylic substitution cascade reaction, using α-(pyridin-1-yl)-acetamides (formed in situ) as nucleophiles, has been developed, generating chiral piperidine-containing amino acid derivatives via a one-pot procedure in high yields and with up to 96% ee. The products can be easily converted into potential bioactive compounds, unnatural chiral amino acids and dipeptides.
Collapse
Affiliation(s)
- Kun Yao
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs , School of Pharmacy , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China . ;
| | - Qianjia Yuan
- School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China
| | - Xingxin Qu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs , School of Pharmacy , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China . ;
| | - Yangang Liu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs , School of Pharmacy , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China . ;
| | - Delong Liu
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs , School of Pharmacy , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China . ;
| | - Wanbin Zhang
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs , School of Pharmacy , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China . ;
- School of Chemistry and Chemical Engineering , Shanghai Jiao Tong University , 800 Dongchuan Road , Shanghai 200240 , P. R. China
| |
Collapse
|
29
|
Pearson D, Garnier M, Luneau A, James AD, Walles M. 19F-NMR-based determination of the absorption, metabolism and excretion of the oral phosphatidylinositol-3-kinase (PI3K) delta inhibitor leniolisib (CDZ173) in healthy volunteers. Xenobiotica 2018; 49:953-960. [DOI: 10.1080/00498254.2018.1523488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- David Pearson
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Maxime Garnier
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Alexandre Luneau
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Alexander David James
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Markus Walles
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
30
|
Foote KM, Nissink JWM, McGuire T, Turner P, Guichard S, Yates JWT, Lau A, Blades K, Heathcote D, Odedra R, Wilkinson G, Wilson Z, Wood CM, Jewsbury PJ. Discovery and Characterization of AZD6738, a Potent Inhibitor of Ataxia Telangiectasia Mutated and Rad3 Related (ATR) Kinase with Application as an Anticancer Agent. J Med Chem 2018; 61:9889-9907. [PMID: 30346772 DOI: 10.1021/acs.jmedchem.8b01187] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The kinase ataxia telangiectasia mutated and rad3 related (ATR) is a key regulator of the DNA-damage response and the apical kinase which orchestrates the cellular processes that repair stalled replication forks (replication stress) and associated DNA double-strand breaks. Inhibition of repair pathways mediated by ATR in a context where alternative pathways are less active is expected to aid clinical response by increasing replication stress. Here we describe the development of the clinical candidate 2 (AZD6738), a potent and selective sulfoximine morpholinopyrimidine ATR inhibitor with excellent preclinical physicochemical and pharmacokinetic (PK) characteristics. Compound 2 was developed improving aqueous solubility and eliminating CYP3A4 time-dependent inhibition starting from the earlier described inhibitor 1 (AZ20). The clinical candidate 2 has favorable human PK suitable for once or twice daily dosing and achieves biologically effective exposure at moderate doses. Compound 2 is currently being tested in multiple phase I/II trials as an anticancer agent.
Collapse
Affiliation(s)
- Kevin M Foote
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - J Willem M Nissink
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Thomas McGuire
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Paul Turner
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Sylvie Guichard
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - James W T Yates
- DMPK, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Alan Lau
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Kevin Blades
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Dan Heathcote
- Discovery Sciences, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Rajesh Odedra
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Gary Wilkinson
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Zena Wilson
- Bioscience, Oncology, IMED Biotech Unit , AstraZeneca , Chesterford Research Park , Little Chesterford, Cambridge CB10 1XL , U.K
| | - Christine M Wood
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| | - Philip J Jewsbury
- Chemistry, Oncology, IMED Biotech Unit , AstraZeneca , Cambridge Science Park, 310 Milton Road , Milton, Cambridge CB4 0WG , U.K
| |
Collapse
|
31
|
Claesson A, Minidis A. Systematic Approach to Organizing Structural Alerts for Reactive Metabolite Formation from Potential Drugs. Chem Res Toxicol 2018; 31:389-411. [DOI: 10.1021/acs.chemrestox.8b00046] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Alf Claesson
- Awametox AB, Lilldalsvägen 17 A, SE-14461 Rönninge, Sweden
| | | |
Collapse
|
32
|
Wang YN, Bheemanaboina RRY, Cai GX, Zhou CH. Novel purine benzimidazoles as antimicrobial agents by regulating ROS generation and targeting clinically resistant Staphylococcus aureus DNA groove. Bioorg Med Chem Lett 2018; 28:1621-1628. [DOI: 10.1016/j.bmcl.2018.03.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 02/28/2018] [Accepted: 03/17/2018] [Indexed: 01/19/2023]
|
33
|
Zhu DP, Xu BH, Du YR, Zhang SJ. SiCl4-catalyzed/PR3-mediated β-C(sp3)−H functionalization of nitrones to α,β-unsaturated imines and aromatic heterocycles. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.03.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
34
|
Metabolomics-assisted metabolite profiling of itraconazole in human liver preparations. J Chromatogr B Analyt Technol Biomed Life Sci 2018. [PMID: 29524695 DOI: 10.1016/j.jchromb.2018.02.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Itraconazole (ITZ) is a first-generation triazole-containing antifungal agent that effectively treats various fungal infections. As ITZ has a better safety profile than that of ketoconazole (KCZ), ITZ has been used worldwide for over 25 years. However, few reports have explored the metabolic profile of ITZ, and the underlying mechanism of ITZ-induced liver injury is not clearly understood. In the present study, we revisited ITZ metabolism in humans, using a non-targeted metabolomics approach, and identified several novel metabolic pathways including O-dearylation, piperazine oxidation, and piperazine-N,N'-deethylation. Furthermore, we explored the formation of reactive ITZ metabolites using trapping agents as surrogates, to assess the possibility of metabolism-mediated toxicity. We found that ITZ and its metabolites did not form any adducts with nucleophiles including glutathione, potassium cyanide, and semicarbazide. The present study expands our knowledge of ITZ metabolism and supports the suggestion that ITZ has a better safety profile than that of KCZ in terms of metabolism-mediated toxicity.
Collapse
|
35
|
Exploring the Metabolism of (+)-[ 18F]Flubatine in Vitro and in Vivo: LC-MS/MS Aided Identification of Radiometabolites in a Clinical PET Study. Molecules 2018; 23:molecules23020464. [PMID: 29461507 PMCID: PMC6017759 DOI: 10.3390/molecules23020464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 01/25/2023] Open
Abstract
Both (+)-[18F]flubatine and its enantiomer (−)-[18F]flubatine are radioligands for the neuroimaging of α4β2 nicotinic acetylcholine receptors (nAChRs) by positron emission tomography (PET). In a clinical study in patients with early Alzheimer’s disease, (+)-[18F]flubatine ((+)-[18F]1) was examined regarding its metabolic fate, in particular by identification of degradation products detected in plasma and urine. The investigations included an in vivo study of (+)-flubatine ((+)-1) in pigs and structural elucidation of formed metabolites by LC-MS/MS. Incubations of (+)-1 and (+)-[18F]1 with human liver microsomes were performed to generate in vitro metabolites, as well as radiometabolites, which enabled an assignment of their structures by comparison of LC-MS/MS and radio-HPLC data. Plasma and urine samples taken after administration of (+)-[18F]1 in humans were examined by radio-HPLC and, on the basis of results obtained in vitro and in vivo, formed radiometabolites were identified. In pigs, (+)-1 was monohydroxylated at different sites of the azabicyclic ring system of the molecule. Additionally, one intermediate metabolite underwent glucuronidation, as also demonstrated in vitro. In humans, a fraction of 95.9 ± 1.9% (n = 10) of unchanged tracer remained in plasma, 30 min after injection. However, despite the low metabolic degradation, both radiometabolites formed in humans could be characterized as (i) a product of C-hydroxylation at the azabicyclic ring system, and (ii) a glucuronide conjugate of the precedingly-formed N8-hydroxylated (+)-[18F]1.
Collapse
|
36
|
Zheng J, Xin Y, Zhang J, Subramanian R, Murray BP, Whitney JA, Warr MR, Ling J, Moorehead L, Kwan E, Hemenway J, Smith BJ, Silverman JA. Pharmacokinetics and Disposition of Momelotinib Revealed a Disproportionate Human Metabolite-Resolution for Clinical Development. Drug Metab Dispos 2018; 46:237-247. [PMID: 29311136 DOI: 10.1124/dmd.117.078899] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022] Open
Abstract
Momelotinib (MMB), a small-molecule inhibitor of Janus kinase (JAK)1/2 and of activin A receptor type 1 (ACVR1), is in clinical development for the treatment of myeloproliferative neoplasms. The pharmacokinetics and disposition of [14C]MMB were characterized in a single-dose, human mass-balance study. Metabolism and the pharmacologic activity of key metabolites were elucidated in multiple in vitro and in vivo experiments. MMB was rapidly absorbed following oral dosing with approximately 97% of the radioactivity recovered, primarily in feces with urine as a secondary route. Mean blood-to-plasma [14C] area under the plasma concentration-time curve ratio was 0.72, suggesting low association of MMB and metabolites with blood cells. [14C]MMB-derived radioactivity was detectable in blood for ≤48 hours, suggesting no irreversible binding of MMB or its metabolites. The major circulating human metabolite, M21 (a morpholino lactam), is a potent inhibitor of JAK1/2 and ACVR1 in vitro. Estimation of pharmacological activity index suggests M21 contributes significantly to the pharmacological activity of MMB for the inhibition of both JAK1/2 and ACVR1. M21 was observed in disproportionately higher amounts in human plasma than in rat or dog, the rodent and nonrodent species used for the general nonclinical safety assessment of this molecule. This discrepancy was resolved with additional nonclinical studies wherein the circulating metabolites and drug-drug interactions were further characterized. The human metabolism of MMB was mediated primarily by multiple cytochrome P450 enzymes, whereas M21 formation involved initial P450 oxidation of the morpholine ring followed by metabolism via aldehyde oxidase.
Collapse
Affiliation(s)
- Jim Zheng
- Gilead Sciences, Inc., Foster City, California
| | - Yan Xin
- Gilead Sciences, Inc., Foster City, California
| | | | | | | | | | | | - John Ling
- Gilead Sciences, Inc., Foster City, California
| | | | - Ellen Kwan
- Gilead Sciences, Inc., Foster City, California
| | | | | | | |
Collapse
|
37
|
Kim JH, Jo JH, Seo KA, Hwang H, Lee HS, Lee S. Non-targeted metabolomics-guided sildenafil metabolism study in human liver microsomes. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1072:86-93. [PMID: 29136555 DOI: 10.1016/j.jchromb.2017.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Metabolomics combined with high-resolution mass spectrometry (HR-MS) and multivariate data analysis has broad applications in the study of xenobiotic metabolism. Although information about xenobiotic metabolism is essential to understand toxic mechanisms, pharmacokinetic parameters and excretion pathways, it is limited to predict all generated metabolites in biological fluids. Here, we revisited sildenafil metabolism in human liver microsomes using a metabolomics approach to achieve a global picture of sildenafil phase 1 metabolism. Finally, 12 phase 1 metabolites were identified in human liver microsomes; M1-M5 were previously known metabolites. The chemical structures of the novel metabolites were elucidated by MS2 fragmentation using an HR-MS system as follows: M6, reduced sildenafil; M7, N,N-deethylation and mono-oxidation; M8, demethanamine, N,N-deethylation and mono-hydroxylation; M9, demethanamine and N,N-deethylation; M10 and M11, mono-oxidation in the piperazine ring after N-demethylation; and M12, mono-oxidation. All metabolites, except M1, were formed by CYP3A4 and CYP3A5. In conclusion, we successfully updated the metabolic pathway of sildenafil in human liver, including 7 novel metabolites using metabolomics combined with HR-MS and multivariate data analysis.
Collapse
Affiliation(s)
- Ju-Hyun Kim
- BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Jun Hyun Jo
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyung-Ah Seo
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, Republic of Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 41061, Republic of Korea
| | - Hye Suk Lee
- BK21 PLUS Team for Creative Leader Program for Pharmacomics-based Future Pharmacy and Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Sangkyu Lee
- BK21 Plus KNU Multi-Omics based Creative Drug Research Team, College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
38
|
Hosogi J, Ohashi R, Maeda H, Tashiro S, Fuse E, Yamamoto Y, Kuwabara T. Monoamine oxidase B oxidizes a novel multikinase inhibitor KW-2449 to its iminium ion and aldehyde oxidase further converts it to the oxo-piperazine form in human. Drug Metab Pharmacokinet 2017; 32:255-264. [DOI: 10.1016/j.dmpk.2017.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/12/2017] [Accepted: 06/15/2017] [Indexed: 01/03/2023]
|
39
|
Gerisch M, Schwarz T, Lang D, Rohde G, Reif S, Genvresse I, Reschke S, van der Mey D, Granvil C. Pharmacokinetics of intravenous pan-class I phosphatidylinositol 3-kinase (PI3K) inhibitor [ 14C]copanlisib (BAY 80-6946) in a mass balance study in healthy male volunteers. Cancer Chemother Pharmacol 2017; 80:535-544. [PMID: 28714036 PMCID: PMC5573760 DOI: 10.1007/s00280-017-3383-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/30/2017] [Indexed: 12/14/2022]
Abstract
PURPOSE To determine the pharmacokinetics of radiolabeled copanlisib (BAY 80-6946) in healthy male volunteers and to investigate the disposition and biotransformation of copanlisib. METHODS A single dose of 12 mg copanlisib containing 2.76 MBq [14C]copanlisib was administered as a 1-h intravenous infusion to 6 volunteers with subsequent sampling up to 34 days. Blood, plasma, urine and feces were collected to monitor total radioactivity, parent compound and metabolites. RESULTS Copanlisib treatment was well tolerated. Copanlisib was rapidly distributed throughout the body with a volume distribution of 1870 L and an elimination half-life of 52.1-h (range 40.4-67.5-h). Copanlisib was the predominant component in human plasma (84% of total radioactivity AUC) and the morpholinone metabolite M1 was the only circulating metabolite (about 5%). Excretion of drug-derived radioactivity based on all 6 subjects was 86% of the dose within a collection interval of 20-34 days with 64% excreted into feces as major route of elimination and 22% into urine. Unchanged copanlisib was the main component excreted into urine (15% of dose) and feces (30% of dose). Excreted metabolites (41% of dose) of copanlisib resulted from oxidative biotransformation. CONCLUSIONS Copanlisib was eliminated predominantly in the feces compared to urine as well as by hepatic biotransformation, suggesting that the clearance of copanlisib would more likely be affected by hepatic impairment than by renal dysfunction. The dual mode of elimination via unchanged excretion of copanlisib and oxidative metabolism decreases the risk of clinically relevant PK-related drug-drug interactions.
Collapse
Affiliation(s)
| | - Thomas Schwarz
- Bayer Aktiengesellschaft, DMPK, 42096, Wuppertal, Germany
| | - Dieter Lang
- Bayer Aktiengesellschaft, DMPK, 42096, Wuppertal, Germany
| | - Gabriele Rohde
- Bayer Aktiengesellschaft, DMPK, 42096, Wuppertal, Germany
| | - Stefanie Reif
- Bayer Aktiengesellschaft, Clinical Sciences, Berlin, Germany
| | | | - Susanne Reschke
- Bayer Aktiengesellschaft, Clinical Sciences, Berlin, Germany
| | | | | |
Collapse
|
40
|
Rabelo VWH, Romeiro NC, Abreu PA. Design strategies of oxidosqualene cyclase inhibitors: Targeting the sterol biosynthetic pathway. J Steroid Biochem Mol Biol 2017; 171:305-317. [PMID: 28479228 DOI: 10.1016/j.jsbmb.2017.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/29/2017] [Accepted: 05/04/2017] [Indexed: 01/04/2023]
Abstract
Targeting the sterol biosynthesis pathway has been explored for the development of new bioactive compounds. Among the enzymes of this pathway, oxidosqualene cyclase (OSC) which catalyzes lanosterol cyclization from 2,3-oxidosqualene has emerged as an attractive target. In this work, we reviewed the most promising OSC inhibitors from different organisms and their potential for the development of new antiparasitic, antifungal, hypocholesterolemic and anticancer drugs. Different strategies have been adopted for the discovery of new OSC inhibitors, such as structural modifications of the natural substrate or the reaction intermediates, the use of the enzyme's structural information to discover compounds with novel chemotypes, modifications of known inhibitors and the use of molecular modeling techniques such as docking and virtual screening to search for new inhibitors. This review brings new perspectives on structural insights of OSC from different organisms and reveals the broad structural diversity of OSC inhibitors which may help evidence lead compounds for further investigations with various therapeutic applications.
Collapse
Affiliation(s)
- Vitor Won-Held Rabelo
- Laboratório de Modelagem Molecular e Pesquisa em Ciências Farmacêuticas, LaMCiFar, Universidade Federal do Rio de Janeiro - Campus Macaé, Av. São José do Barreto, Macaé 27965-045, RJ, Brazil; Programa de Pós-Graduação em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Campus Macaé Professor Aloísio Teixeira, Macaé, RJ, Brazil
| | - Nelilma Correia Romeiro
- Laboratório Integrado de Computação Científica, LICC, Universidade Federal do Rio de Janeiro, Campus Macaé, Macaé, RJ, 27965-045, Brazil
| | - Paula Alvarez Abreu
- Laboratório de Modelagem Molecular e Pesquisa em Ciências Farmacêuticas, LaMCiFar, Universidade Federal do Rio de Janeiro - Campus Macaé, Av. São José do Barreto, Macaé 27965-045, RJ, Brazil.
| |
Collapse
|
41
|
Asano M, Hitaka T, Imada T, Yamada M, Morimoto M, Shinohara H, Hara T, Yamaoka M, Santou T, Nakayama M, Imai Y, Habuka N, Yano J, Wilson K, Fujita H, Hasuoka A. Synthesis and biological evaluation of novel selective androgen receptor modulators (SARMs). Part II: Optimization of 4-(pyrrolidin-1-yl)benzonitrile derivatives. Bioorg Med Chem Lett 2017; 27:1897-1901. [DOI: 10.1016/j.bmcl.2017.03.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 03/14/2017] [Accepted: 03/16/2017] [Indexed: 12/20/2022]
|
42
|
Waldman AJ, Ng TL, Wang P, Balskus EP. Heteroatom-Heteroatom Bond Formation in Natural Product Biosynthesis. Chem Rev 2017; 117:5784-5863. [PMID: 28375000 PMCID: PMC5534343 DOI: 10.1021/acs.chemrev.6b00621] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Natural products that contain functional groups with heteroatom-heteroatom linkages (X-X, where X = N, O, S, and P) are a small yet intriguing group of metabolites. The reactivity and diversity of these structural motifs has captured the interest of synthetic and biological chemists alike. Functional groups containing X-X bonds are found in all major classes of natural products and often impart significant biological activity. This review presents our current understanding of the biosynthetic logic and enzymatic chemistry involved in the construction of X-X bond containing functional groups within natural products. Elucidating and characterizing biosynthetic pathways that generate X-X bonds could both provide tools for biocatalysis and synthetic biology, as well as guide efforts to uncover new natural products containing these structural features.
Collapse
Affiliation(s)
- Abraham J. Waldman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Tai L. Ng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Peng Wang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Emily P. Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| |
Collapse
|
43
|
Huang PQ, Chen H. Ni-Catalyzed cross-coupling reactions of N-acylpyrrole-type amides with organoboron reagents. Chem Commun (Camb) 2017; 53:12584-12587. [DOI: 10.1039/c7cc07457c] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We report the first Ni/bis-NHC-catalyzed cross-coupling of N-acylpyrrole-type amides with arylboronic esters to obtain diarylketones.
Collapse
Affiliation(s)
- Pei-Qiang Huang
- Department of Chemistry
- College of Chemistry and Chemical Engineering
- and Fujian Provincial Key Laboratory of Chemical Biology
- Xiamen University
- Xiamen
| | - Hang Chen
- Department of Chemistry
- College of Chemistry and Chemical Engineering
- and Fujian Provincial Key Laboratory of Chemical Biology
- Xiamen University
- Xiamen
| |
Collapse
|
44
|
Wu G, Li Y, Yu X, Gao Y, Chen H. Acetic Acid Accelerated Visible-Light Photoredox CatalyzedN-Demethylation ofN,N-Dimethylaminophenyl Derivatives. Adv Synth Catal 2016. [DOI: 10.1002/adsc.201601108] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Guolin Wu
- College of Chemistry; Fuzhou University; Fuzhou Fujian 350116 China
| | - Yazhen Li
- College of Chemistry; Fuzhou University; Fuzhou Fujian 350116 China
| | - Xuemei Yu
- College of Chemistry; Fuzhou University; Fuzhou Fujian 350116 China
| | - Yu Gao
- College of Chemistry; Fuzhou University; Fuzhou Fujian 350116 China
| | - Haijun Chen
- College of Chemistry; Fuzhou University; Fuzhou Fujian 350116 China
| |
Collapse
|
45
|
Marcantoni E, Petrini M. Recent Developments in the Stereoselective Synthesis of Nitrogen-Containing Heterocycles usingN-Acylimines as Reactive Substrates. Adv Synth Catal 2016. [DOI: 10.1002/adsc.201600644] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Enrico Marcantoni
- School of Science and Technology; Chemistry Division; Università di Camerino; via S. Agostino 1 62032 Camerino Italy
| | - Marino Petrini
- School of Science and Technology; Chemistry Division; Università di Camerino; via S. Agostino 1 62032 Camerino Italy
| |
Collapse
|
46
|
Mbofana CT, Chong E, Lawniczak J, Sanford MS. Iron-Catalyzed Oxyfunctionalization of Aliphatic Amines at Remote Benzylic C-H Sites. Org Lett 2016; 18:4258-61. [PMID: 27529646 PMCID: PMC5356366 DOI: 10.1021/acs.orglett.6b02003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We report the development of an iron-catalyzed method for the selective oxyfunctionalization of benzylic C(sp(3))-H bonds in aliphatic amine substrates. This transformation is selective for benzylic C-H bonds that are remote (i.e., at least three carbons) from the amine functional group. High site selectivity is achieved by in situ protonation of the amine with trifluoroacetic acid, which deactivates more traditionally reactive C-H sites that are α to nitrogen. The scope and synthetic utility of this method are demonstrated via the synthesis and derivatization of a variety of amine-containing, biologically active molecules.
Collapse
Affiliation(s)
- Curren T. Mbofana
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eugene Chong
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - James Lawniczak
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Melanie S. Sanford
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
47
|
Zhu Y, Li L, Zhang G, Wan H, Yang C, Diao X, Chen X, Zhang L, Zhong D. Metabolic characterization of pyrotinib in humans by ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1033-1034:117-127. [PMID: 27541626 DOI: 10.1016/j.jchromb.2016.08.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/23/2016] [Accepted: 08/06/2016] [Indexed: 11/28/2022]
Abstract
Pyrotinib is a novel irreversible tyrosine kinase inhibitor developed for the treatment of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. The results of phase I clinical trial demonstrated that pyrotinib was well tolerated and exhibited potent antitumor activity. As a promising therapeutic agent for HER2-positive breast cancer, it is of great importance to investigate the biotransformation of pyrotinib in humans and identify the major enzymes involved in its metabolism during its early stage of development for safety consideration. For this purpose, a robust analytical method based on ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC/Q-TOF MS) was established to characterize the metabolites of pyrotinib in human plasma, feces, and urine, and identify the primary enzymes responsible for its metabolism. As a result, a total of 24 metabolites were identified, including 16 phase I metabolites resulting from dealkylation, oxidation, dehydrogenation, and carbonylation, and 8 phase II metabolites originating from cysteine and N-acetylcysteine conjugation. Pyrotinib was absorbed into blood by 1h, reached its peak level at 4h, and afterwards underwent slow elimination. The principal metabolites detected in humans (M1, M2, and M5) were products resulting from O-depicoline and pyrrolidine lactam formation, whose structures have been confirmed by the synthetic references. In addition, fecal clearance was the major route of excretion for pyrotinib. Further phenotyping experiment proved that CYP3A4 was the most active enzyme responsible for the biotransformation of pyrotinib, implying the vital necessity of the assessment of the potential CYP3A-mediated drug-drug interactions in humans. Taken together, this study provided valuable metabolic data to explicate the dynamic process of pyrotinib in humans, and important reference basis for its safety evaluation and rational clinical application. The results will also benefit the assessment of the contributions to the overall activity or toxicity from the key metabolites.
Collapse
Affiliation(s)
- Yunting Zhu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Liang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Ge Zhang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai 200122, People's Republic of China
| | - Hong Wan
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai 200122, People's Republic of China
| | - Changyong Yang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai 200122, People's Republic of China
| | - Xingxing Diao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Xiaoyan Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
| | - Lianshan Zhang
- Jiangsu Hengrui Medicine Co., Ltd, Shanghai 200122, People's Republic of China
| | - Dafang Zhong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China.
| |
Collapse
|
48
|
Bonamore A, Calisti L, Calcaterra A, Ismail OH, Gargano M, D'Acquarica I, Botta B, Boffi A, Macone A. A Novel Enzymatic Strategy for the Synthesis of Substituted Tetrahydroisoquinolines. ChemistrySelect 2016. [DOI: 10.1002/slct.201600134] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Alessandra Bonamore
- Dipartimento di Scienze Biochimiche and Istituto di Biologia e Patologia Molecolari (IBPM), CNR; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
- MOLIROM s.r.l; Via Carlo Bartolomeo Piazza 8 Roma 00161 Italy
| | - Lorenzo Calisti
- Dipartimento di Scienze Biochimiche and Istituto di Biologia e Patologia Molecolari (IBPM), CNR; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| | - Andrea Calcaterra
- Dipartimento di Chimica e Tecnologia del Farmaco; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| | - Omar H. Ismail
- Dipartimento di Chimica e Tecnologia del Farmaco; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| | - Maurizio Gargano
- Dipartimento di Scienze Biochimiche and Istituto di Biologia e Patologia Molecolari (IBPM), CNR; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| | - Ilaria D'Acquarica
- Dipartimento di Chimica e Tecnologia del Farmaco; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologia del Farmaco; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
- MOLIROM s.r.l; Via Carlo Bartolomeo Piazza 8 Roma 00161 Italy
| | - Alberto Boffi
- Dipartimento di Scienze Biochimiche and Istituto di Biologia e Patologia Molecolari (IBPM), CNR; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
- MOLIROM s.r.l; Via Carlo Bartolomeo Piazza 8 Roma 00161 Italy
| | - Alberto Macone
- Dipartimento di Scienze Biochimiche and Istituto di Biologia e Patologia Molecolari (IBPM), CNR; Sapienza Università di Roma; Piazzale Aldo Moro 5 Roma 00185 Italy
| |
Collapse
|
49
|
Thompson RA, Isin EM, Ogese MO, Mettetal JT, Williams DP. Reactive Metabolites: Current and Emerging Risk and Hazard Assessments. Chem Res Toxicol 2016; 29:505-33. [DOI: 10.1021/acs.chemrestox.5b00410] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Richard A. Thompson
- DMPK, Respiratory, Inflammation & Autoimmunity iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Emre M. Isin
- DMPK, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, 431 83 Mölndal, Sweden
| | - Monday O. Ogese
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| | - Jerome T. Mettetal
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, 35 Gatehouse Dr, Waltham, Massachusetts 02451, United States
| | - Dominic P. Williams
- Translational Safety, Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, United Kingdom
| |
Collapse
|
50
|
Bolleddula J, Chowdhury SK. Carbon-carbon bond cleavage and formation reactions in drug metabolism and the role of metabolic enzymes. Drug Metab Rev 2015; 47:534-57. [PMID: 26390887 DOI: 10.3109/03602532.2015.1086781] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elimination of xenobiotics from the human body is often facilitated by a transformation to highly water soluble and more ionizable molecules. In general, oxidation-reduction, hydrolysis, and conjugation reactions are common biotransformation reactions that are catalyzed by various metabolic enzymes including cytochrome P450s (CYPs), non-CYPs, and conjugative enzymes. Although carbon-carbon (C-C) bond formation and cleavage reactions are known to exist in plant secondary metabolism, these reactions are relatively rare in mammalian metabolism and are considered exceptions. However, various reactions such as demethylation, dealkylation, dearylation, reduction of alkyl chain, ring expansion, ring contraction, oxidative elimination of a nitrile through C-C bond cleavage, and dimerization, and glucuronidation through C-C bond formation have been reported for drug molecules. Carbon-carbon bond cleavage reactions for drug molecules are primarily catalyzed by CYP enzymes, dimerization is mediated by peroxidases, and C-glucuronidation is catalyzed by UGT1A9. This review provides an overview of C-C bond cleavage and formation reactions in drug metabolism and the metabolic enzymes associated with these reactions.
Collapse
Affiliation(s)
- Jayaprakasam Bolleddula
- a Department of Drug Metabolism and Pharmacokinetics , Takeda Pharmaceuticals International Co. , Cambridge , MA , USA
| | - Swapan K Chowdhury
- a Department of Drug Metabolism and Pharmacokinetics , Takeda Pharmaceuticals International Co. , Cambridge , MA , USA
| |
Collapse
|