1
|
Uetrecht J. DILI prediction in drug development: present and future. Expert Opin Drug Metab Toxicol 2025:1-12. [PMID: 40253704 DOI: 10.1080/17425255.2025.2495955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
INTRODUCTION Idiosyncratic drug-induced liver injury (iDILI) results in significant patient morbidity and significantly increases the risk of drug development. The current methods to screen for iDILI risk are inadequate. AREAS COVERED The general mechanism of iDILI and the current methods to screen for iDILI are reviewed. Then the potential for new biomarkers is explored. EXPERT OPINION Better biomarkers of iDILI risk should be based on the mechanism of iDILI. In general, it is an adaptive immune response, specifically CD8+ cytotoxic T cells, that is responsible for hepatocyte cell death, not direct toxicity of the drug. Therefore, in vitro cytotoxicity assays represent an artifact not the mechanism of iDILI. Activation of the adaptive immune response leading to iDILI requires an innate immune response, in particular activation of antigen presenting cells. The innate immune response is immediate and unlikely to be idiosyncratic. For example, studies have found that incubation of hepatocytes with drugs causes the release of molecules that activate THP-1-derived macrophages. The response of hepatocytes, the release of damage-associated molecular pattern molecules (DAMPs), especially in extracellular vesicles, and the response of antigen presenting cells (APCs) are likely to provide better biomarkers of iDILI risk.
Collapse
Affiliation(s)
- Jack Uetrecht
- Faculty of Pharmacy, University of Toronto, Toronto, ON, USA
| |
Collapse
|
2
|
He C, Mao Y, Wan H. In-depth understanding of the structure-based reactive metabolite formation of organic functional groups. Drug Metab Rev 2025:1-43. [PMID: 40008940 DOI: 10.1080/03602532.2025.2472076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a leading cause of drug attrition and/or withdrawal. The formation of reactive metabolites is widely accepted as a key factor contributing to idiosyncratic DILI. Therefore, identifying reactive metabolites has become a critical focus during lead optimization, and a combination of GSH-/cyano-trapping and cytochrome P450 inactivation studies is recommended to identify compounds with the potential to generate reactive metabolites. Daily dose, clinical indication, detoxication pathways, administration route, and treatment duration are the most considerations when deprioritizing candidates that generate reactive metabolites. Removing the structural alerts is considered a pragmatic strategy for mitigating the risk associated with reactive metabolites, although this approach may sometimes exclude otherwise potent molecules. In this context, an in-depth insight into the structure-based reactive metabolite formation of organic functional groups can significantly aid in the rational design of drug candidates with improved safety profiles. The primary goal of this review is to delve into an analysis of the bioactivation mechanisms of organic functional groups and their potential detrimental effects with recent examples to assist medicinal chemists and metabolism scientists in designing safer drug candidates with a higher likelihood of success.
Collapse
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Yuchang Mao
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Hong Wan
- WHDex Consulting AB, Mölndal, Sweden
| |
Collapse
|
3
|
Faramarzi S, Bassan A, Cross KP, Yang X, Myatt GJ, Volpe DA, Stavitskaya L. Novel (Q)SAR models for prediction of reversible and time-dependent inhibition of cytochrome P450 enzymes. Front Pharmacol 2025; 15:1451164. [PMID: 40012840 PMCID: PMC11860084 DOI: 10.3389/fphar.2024.1451164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/27/2024] [Indexed: 02/28/2025] Open
Abstract
The 2020 FDA drug-drug interaction (DDI) guidance includes a consideration for metabolites with structural alerts for potential mechanism-based inhibition (MBI) and describes how this information may be used to determine whether in vitro studies need to be conducted to evaluate the inhibitory potential of a metabolite on CYP enzymes. To facilitate identification of structural alerts, an extensive literature search was performed and alerts for mechanism-based inhibition of cytochrome P450 enzymes (CYP) were collected. Furthermore, five quantitative structure-activity relationship (QSAR) models were developed to predict not only time-dependent inhibition of CYP3A4, an enzyme that metabolizes approximately 50% of all marketed drugs, but also reversible inhibition of 3A4, 2C9, 2C19 and 2D6. The non-proprietary training database for the QSAR models contains data for 10,129 chemicals harvested from FDA drug approval packages and published literature. The cross-validation performance statistics for the new CYP QSAR models range from 78% to 84% sensitivity and 79%-84% normalized negative predictivity. Additionally, the performance of the newly developed QSAR models was assessed using external validation sets. Overall performance statistics showed up to 75% in sensitivity and up to 80% in normalized negative predictivity. The newly developed models will provide a faster and more effective evaluation of potential drug-drug interaction caused by metabolites.
Collapse
Affiliation(s)
- Sadegh Faramarzi
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | | | | | - Xinning Yang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | | | - Donna A. Volpe
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Lidiya Stavitskaya
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
4
|
Nishio Y, Lindsley CW, Bender AM. Classics in Chemical Neuroscience: Tianeptine. ACS Chem Neurosci 2024; 15:3863-3873. [PMID: 39382192 PMCID: PMC11587517 DOI: 10.1021/acschemneuro.4c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
Tianeptine (1) is an unusual antidepressant in that its mechanism of action appears to be independent from any activity at serotonin receptors or monoamine transporters. In fact, tianeptine has been shown to be a moderately potent agonist for the mu opioid receptor (MOR) and to a lesser extent the delta opioid receptor (DOR). Additionally, tianeptine's efficacy may be related to its action on glutamate-mediated pathways of neuroplasticity. Regardless of which neurotransmitter system is primarily responsible for the observed efficacy, the MOR agonist activity is problematic with respect to abuse liability. Increasing numbers of case reports have demonstrated that tianeptine is indeed being used recreationally at doses far beyond what are considered therapeutically relevant or safe, and scheduling reclassifications or outright bans on tianeptine products are ongoing around the world. It is the aim of this review to discuss the medicinal chemistry and pharmacology of tianeptine and to summarize this intriguing discrepancy between tianeptine's historical use as a safe and effective antidepressant and its emerging potential for abuse.
Collapse
Affiliation(s)
- Yu Nishio
- Warren
Center for Neuroscience Drug Discovery, Department of Pharmacology, Department of Chemistry, and Department of
Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Warren
Center for Neuroscience Drug Discovery, Department of Pharmacology, Department of Chemistry, and Department of
Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Aaron M. Bender
- Warren
Center for Neuroscience Drug Discovery, Department of Pharmacology, Department of Chemistry, and Department of
Biochemistry, Vanderbilt University, Nashville, Tennessee 37232, United States
| |
Collapse
|
5
|
Smith DA. Teratogenicity is more likely a function of primary and secondary pharmacology than caused by chemically reactive metabolites: a critical evaluation of 40 years of scientific research. Xenobiotica 2024; 54:599-608. [PMID: 38913781 DOI: 10.1080/00498254.2024.2366302] [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: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/26/2024]
Abstract
The number of therapeutic drugs known to be human teratogens is actually relatively small. This may reflect the rigorous animal testing and well defined labelling. Some of these drugs were identified to have reactive metabolites and this has been postulated, historically, to be their teratogenic mechanism. These drugs include thalidomide, various anticonvulsants and retinoic acid derivatives.Many of these experiments were conducted in a period where chemically reactive metabolites were being intensely investigated and associated with all forms of toxicity. The legacy of this is that these examples are routinely cited as well established mechanisms.Examination of mechanism leads to the conclusion that the teratogenicity in humans of these compounds is likely due to the primary and secondary pharmacology of the parent drug and stable circulating metabolites and that association of reactive metabolites to this toxicity is unwarranted.
Collapse
|
6
|
Claesson A. Use of Structural Alerts for Reactive Metabolites in the Application SpotRM. Chem Res Toxicol 2024; 37:1231-1245. [PMID: 39088358 DOI: 10.1021/acs.chemrestox.4c00205] [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: 08/03/2024]
Abstract
Reactive metabolite (RM) formation is widely accepted as playing a crucial role in causing idiosyncratic adverse drug reactions (IADRs), where the liver is most affected. An important goal of drug design is to avoid selection of drug candidates giving rise to RMs and therefore risk causing problems later on involving IADRs. The simplest, initial approach is to avoid test structures that have substructures known or strongly suspected to be associated with IADRs. However, as is evident from the many case reports of IADRs, in most cases a clear association with any (bio)chemical mechanism is lacking, which makes it hard to establish any structure-toxicity relationship. Separate studies of RM formation, in vitro and in vivo, have led to likely evidence and to establishing many structural alerts (SAs) that can be used for fast selection/deselection of planned test compounds. As a background to a discussion of the concept, 25 kinase inhibitor drugs with known problems of hepatotoxicity were probed against a set of SAs contained in the application SpotRM. A clear majority of the probed drugs show liabilities as evident by being flagged by more than one of the fairly established types of SAs. At the same time, no clear SAs were found in three drugs, which is discussed in the broader context of usefulness and selection tactics of SAs in drug design.
Collapse
Affiliation(s)
- Alf Claesson
- Awametox AB, Lilldalsvägen 17 A, SE-14461 Rönninge, Sweden
| |
Collapse
|
7
|
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
|
8
|
Sabat M, Carney DW, Hernandez-Torres G, Gibson TS, Balakrishna D, Zou H, Xu R, Chen CH, de Jong R, Dougan DR, Qin L, Bigi-Botterill SV, Chambers A, Miura J, Johnson LK, Ermolieff J, Johns D, Selimkhanov J, Kwok L, DeMent K, Proffitt C, Vu P, Lindsey EA, Ivetac T, Jennings A, Wang H, Manam P, Santos C, Fullenwider C, Manohar R, Flick AC. Design and Discovery of a Potent and Selective Inhibitor of Integrin αvβ1. J Med Chem 2024; 67:10306-10320. [PMID: 38872300 DOI: 10.1021/acs.jmedchem.4c00743] [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: 06/15/2024]
Abstract
Selective inhibition of the RGD (Arg-Gly-Asp) integrin αvβ1 has been recently identified as an attractive therapeutic approach for the treatment of liver fibrosis given its function, target expression, and safety profile. Our identification of a non-RGD small molecule lead followed by focused, systematic changes to the core structure utilizing a crystal structure, in silico modeling, and a tractable synthetic approach resulted in the identification of a potent small molecule exhibiting a remarkable affinity for αvβ1 relative to several other integrin isoforms measured. Azabenzimidazolone 25 demonstrated antifibrotic efficacy in an in vivo rat liver fibrosis model and represents a tool compound capable of further exploring the biological consequences of selective αvβ1 inhibition.
Collapse
Affiliation(s)
- Mark Sabat
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Daniel W Carney
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Gloria Hernandez-Torres
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony S Gibson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deepika Balakrishna
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Hua Zou
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rui Xu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chien-Hung Chen
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ron de Jong
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Douglas R Dougan
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ling Qin
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Simone V Bigi-Botterill
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Alison Chambers
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Joanne Miura
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lucas K Johnson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jacques Ermolieff
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deidre Johns
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jangir Selimkhanov
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lily Kwok
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Kevin DeMent
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chris Proffitt
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Phong Vu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Erick A Lindsey
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony Ivetac
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andy Jennings
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Haixia Wang
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Padma Manam
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cipriano Santos
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cody Fullenwider
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rohan Manohar
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andrew C Flick
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| |
Collapse
|
9
|
Hu XM, Hou YY, Teng XR, Liu Y, Li Y, Li W, Li Y, Ai CZ. Prediction of cytochrome P450-mediated bioactivation using machine learning models and in vitro validation. Arch Toxicol 2024; 98:1457-1467. [PMID: 38492097 DOI: 10.1007/s00204-024-03701-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/31/2024] [Indexed: 03/18/2024]
Abstract
Cytochrome P450 (P450)-mediated bioactivation, which can lead to the hepatotoxicity through the formation of reactive metabolites (RMs), has been regarded as the major problem of drug failures. Herein, we purposed to establish machine learning models to predict the bioactivation of P450. On the basis of the literature-derived bioactivation dataset, models for Benzene ring, Nitrogen heterocycle and Sulfur heterocycle were developed with machine learning methods, i.e., Random Forest, Random Subspace, SVM and Naïve Bayes. The models were assessed by metrics like "Precision", "Recall", "F-Measure", "AUC" (Area Under the Curve), etc. Random Forest algorithms illustrated the best predictability, with nice AUC values of 0.949, 0.973 and 0.958 for the test sets of Benzene ring, Nitrogen heterocycle and Sulfur heterocycle models, respectively. 2D descriptors like topological indices, 2D autocorrelations and Burden eigenvalues, etc. contributed most to the models. Furthermore, the models were applied to predict the occurrence of bioactivation of an external verification set. Drugs like selpercatinib, glafenine, encorafenib, etc. were predicted to undergo bioactivation into toxic RMs. In vitro, IC50 shift experiment was performed to assess the potential of bioactivation to validate the prediction. Encorafenib and tirbanibulin were observed of bioactivation potential with shifts of 3-6 folds or so. Overall, this study provided a reliable and robust strategy to predict the P450-mediated bioactivation, which will be helpful to the assessment of adverse drug reactions (ADRs) in clinic and the design of new candidates with lower toxicities.
Collapse
Affiliation(s)
- Xin-Man Hu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Yan-Yao Hou
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Xin-Ru Teng
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Yong Liu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, 2 Dagong Road, Panjin, 124221, People's Republic of China
| | - Yu Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China
| | - Wei Li
- Translational Medicine Research Institute, College of Medicine, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, 136 Jiangyangzhong Road, Yangzhou, 225001, People's Republic of China.
| | - Yan Li
- Department of Materials Science and Chemical Engineering, Dalian University of Technology, Dalian, 116023, Liaoning, People's Republic of China
| | - Chun-Zhi Ai
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, 15 Yucai Road, Guilin, 541004, People's Republic of China.
| |
Collapse
|
10
|
Ren H, Li T, Xing J, Li Z, Zhang Y, Yu X, Zheng J. Ti-Catalyzed Formal [2π + 2σ] Cycloadditions of Bicyclo[1.1.0]butanes with 2-Azadienes to Access Aminobicyclo[2.1.1]hexanes. Org Lett 2024; 26:1745-1750. [PMID: 38377354 DOI: 10.1021/acs.orglett.4c00421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Saturated bicyclic amines are increasingly targeted to the pharmaceutical industry as sp3-rich bioisosteres of anilines. Numerous strategies have been established for the preparation of bridgehead aminobicyclics. However, methods to assemble the bridge-amino hydrocarbon skeleton, which serves as a meta-substituted arene bioisostere, are limited. Herein, a general approach to access 2-aminobicyclo[2.1.1]hexanes (aminoBCHs) by titanium-catalyzed formal [2π + 2σ] cycloaddition of bicyclo[1.1.0]butanes and 2-azadienes was developed. Simple derivatization of aminoBCHs leads to various medicinally and agrochemically important analogues.
Collapse
Affiliation(s)
- Haosong Ren
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Tianxiang Li
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jinping Xing
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Zhenyue Li
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Yanxia Zhang
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, P. R. China
| | - Xinhong Yu
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| | - Jun Zheng
- Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P. R. China
| |
Collapse
|
11
|
Wang C, Liu X, Wang Q, Fang WH, Chen X. Unveiling Mechanistic Insights and Photocatalytic Advancements in Intramolecular Photo-(3 + 2)-Cycloaddition: A Comparative Assessment of Two Paradigmatic Single-Electron-Transfer Models. JACS AU 2024; 4:419-431. [PMID: 38425917 PMCID: PMC10900211 DOI: 10.1021/jacsau.3c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 03/02/2024]
Abstract
The synthesis of 1-aminonorbornane (1-aminoNB), a potential aniline bioisostere, through photochemistry or photoredox catalysis signifies a remarkable breakthrough with implications in organic chemistry, pharmaceutical chemistry, and sustainable chemistry. However, an understanding of the underlying mechanisms involved in these reactions remains limited and ambiguous. Herein, we employ high-precision CASPT2//CASSCF calculations to elucidate the intricate mechanisms regulating the intramolecular photo-(3 + 2)-cycloaddition reactions for the synthesis of 1-aminoNB in the presence or absence of the Ir-complex-based photocatalyst. Our investigations delve into radical cascades, stereoselectivity, particularly single-electron-transfer (SET) events, etc. Furthermore, we innovatively introduce and compare two SET models integrating Marcus electron-transfer theory and transition-state theory. These models combined with kinetic data contribute to recognizing the critical control factors in diverse photocatalysis, thereby guiding the design and manipulation of photoredox catalysis as well as the improvement and modification of photocatalysts.
Collapse
Affiliation(s)
- Chu Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, People’s Republic of China
| | - Xiao Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, People’s Republic of China
| | - Qian Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, People’s Republic of China
| | - Wei-Hai Fang
- College of Chemistry, Beijing Normal University, Beijing 100875, People’s Republic of China
| | - Xuebo Chen
- College of Chemistry, Beijing Normal University, Beijing 100875, People’s Republic of China
| |
Collapse
|
12
|
Yano T, Yamada T, Isida H, Ohashi N, Itoh T. 2-cyanopyridine derivatives enable N-terminal cysteine bioconjugation and peptide bond cleavage of glutathione under aqueous and mild conditions. RSC Adv 2024; 14:6542-6547. [PMID: 38390509 PMCID: PMC10882492 DOI: 10.1039/d4ra00437j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024] Open
Abstract
Inspired by the chemical reactivity of apalutamide, we have developed an efficient method for N-terminal cysteine bioconjugation with 2-cyanopyridine derivatives. Systematic investigations of various 2-cyanopyridines revealed that 2-cyanopyridines with electron-withdrawing groups react efficiently with cysteine under aqueous and mild conditions. Moreover, the highly reactive 2-cyanopyridines enable the peptide bond cleavage of glutathione. The utility of our method is demonstrated by its application to the cysteine-selective chemical modification of bioactive peptides.
Collapse
Affiliation(s)
- Tetsuya Yano
- Showa Pharmaceutical University Machida Tokyo 194-8543 Japan
| | - Takahiro Yamada
- Showa Pharmaceutical University Machida Tokyo 194-8543 Japan
| | - Hiroaki Isida
- Showa Pharmaceutical University Machida Tokyo 194-8543 Japan
| | - Nami Ohashi
- Showa Pharmaceutical University Machida Tokyo 194-8543 Japan
| | - Toshimasa Itoh
- Showa Pharmaceutical University Machida Tokyo 194-8543 Japan
| |
Collapse
|
13
|
Khalil SM, MacKenzie KR, Maletic-Savatic M, Li F. Metabolic bioactivation of antidepressants: advance and underlying hepatotoxicity. Drug Metab Rev 2024; 56:97-126. [PMID: 38311829 PMCID: PMC11118075 DOI: 10.1080/03602532.2024.2313967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Many drugs that serve as first-line medications for the treatment of depression are associated with severe side effects, including liver injury. Of the 34 antidepressants discussed in this review, four have been withdrawn from the market due to severe hepatotoxicity, and others carry boxed warnings for idiosyncratic liver toxicity. The clinical and economic implications of antidepressant-induced liver injury are substantial, but the underlying mechanisms remain elusive. Drug-induced liver injury may involve the host immune system, the parent drug, or its metabolites, and reactive drug metabolites are one of the most commonly referenced risk factors. Although the precise mechanism by which toxicity is induced may be difficult to determine, identifying reactive metabolites that cause toxicity can offer valuable insights for decreasing the bioactivation potential of candidates during the drug discovery process. A comprehensive understanding of drug metabolic pathways can mitigate adverse drug-drug interactions that may be caused by elevated formation of reactive metabolites. This review provides a comprehensive overview of the current state of knowledge on antidepressant bioactivation, the metabolizing enzymes responsible for the formation of reactive metabolites, and their potential implication in hepatotoxicity. This information can be a valuable resource for medicinal chemists, toxicologists, and clinicians engaged in the fields of antidepressant development, toxicity, and depression treatment.
Collapse
Affiliation(s)
- Saleh M. Khalil
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kevin R. MacKenzie
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine; Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Feng Li
- Center for Drug Discovery, Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
14
|
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
|
15
|
Krumm CS, Landzberg RS, Ramos-Espiritu L, Adura C, Liu X, Acuna M, Xie Y, Xu X, Tillman MC, Li Y, Glickman JF, Ortlund EA, Ginn JD, Cohen DE. High-throughput screening identifies small molecule inhibitors of thioesterase superfamily member 1: Implications for the management of non-alcoholic fatty liver disease. Mol Metab 2023; 78:101832. [PMID: 38403978 PMCID: PMC10663673 DOI: 10.1016/j.molmet.2023.101832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 02/27/2024] Open
Abstract
OBJECTIVE Thioesterase superfamily member 1 (Them1) is a long chain acyl-CoA thioesterase comprising two N-terminal HotDog fold enzymatic domains linked to a C-terminal lipid-sensing steroidogenic acute regulatory transfer-related (START) domain, which allosterically modulates enzymatic activity. Them1 is highly expressed in thermogenic adipose tissue, where it functions to suppress energy expenditure by limiting rates of fatty acid oxidation, and is induced markedly in liver in response to high fat feeding, where it suppresses fatty acid oxidation and promotes glucose production. Them1-/- mice are protected against non-alcoholic fatty liver disease (NAFLD), suggesting Them1 as a therapeutic target. METHODS A high-throughput small molecule screen was performed to identify promising inhibitors targeting the fatty acyl-CoA thioesterase activity of purified recombinant Them1.Counter screening was used to determine specificity for Them1 relative to other acyl-CoA thioesterase isoforms. Inhibitor binding and enzyme inhibition were quantified by biophysical and biochemical approaches, respectively. Following structure-based optimization, lead compounds were tested in cell culture. RESULTS Two lead allosteric inhibitors were identified that selectively inhibited Them1 by binding the START domain. In mouse brown adipocytes, these inhibitors promoted fatty acid oxidation, as evidenced by increased oxygen consumption rates. In mouse hepatocytes, they promoted fatty acid oxidation, but also reduced glucose production. CONCLUSION Them1 inhibitors could prove attractive for the pharmacologic management of NAFLD.
Collapse
Affiliation(s)
- Christopher S Krumm
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065, USA
| | - Renée S Landzberg
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Carolina Adura
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Xu Liu
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mariana Acuna
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yang Xie
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xu Xu
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew C Tillman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yingxia Li
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - J Fraser Glickman
- Fisher Drug Discovery Resource Center, The Rockefeller University, New York, NY 10065, USA
| | - Eric A Ortlund
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - John D Ginn
- Sanders Tri-Institutional Therapeutics Discovery Institute, New York, NY 10065, USA
| | - David E Cohen
- Division of Gastroenterology & Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
16
|
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
|
17
|
Najjar A, Ellison CA, Gregoire S, Hewitt NJ. Practical application of the interim internal threshold of toxicological concern (iTTC): a case study based on clinical data. Arch Toxicol 2023; 97:155-164. [PMID: 36149470 PMCID: PMC9816204 DOI: 10.1007/s00204-022-03371-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/25/2022] [Indexed: 01/19/2023]
Abstract
We present a case study that provides a practical step-by-step example of how the internal Threshold of Toxicological Concern (iTTC) can be used as a tool to refine a TTC-based assessment for dermal exposures to consumer products. The case study uses a theoretical scenario where there are no systemic toxicity data for the case study chemicals (avobenzone, oxybenzone, octocrylene, homosalate, octisalate, octinoxate, and ecamsule). Human dermal pharmacokinetic data following single and repeat dermal exposure to products containing the case study chemicals were obtained from data published by the US FDA. The clinical studies utilized an application procedure that followed maximal use conditions (product applied as 2 mg/cm2 to 75% of the body surface area, 4 times a day). The case study chemicals were first reviewed to determine if they were in the applicability domain of the iTTC, and then, the human plasma concentrations were compared to an iTTC limit of 1 µM. When assessed under maximum usage, the external exposure of all chemicals exceeded the external dose TTC limits. By contrast, the internal exposure to all chemicals, except oxybenzone, was an order of magnitude lower than the 1 µM interim iTTC threshold. This work highlights the importance of understanding internal exposure relative to external dose and how the iTTC can be a valuable tool for assessing low-level internal exposures; additionally, the work demonstrates how to use an iTTC, and highlights considerations and refinement opportunities for the approach.
Collapse
Affiliation(s)
| | - Corie A Ellison
- The Procter & Gamble Company, 8700 Mason Montgomery Road, Cincinnati, OH, 45040, USA.
| | - Sebastien Gregoire
- L'Oreal Research & Innovation, 1, Avenue Eugène Schueller, 93601, Aulnay-sous-Bois, France
| | - Nicola J Hewitt
- Cosmetics Europe, Avenue Herrmann-Debroux 40, 1160, Brussels, Belgium
| |
Collapse
|
18
|
Hawryluk N, Robinson D, Shen Y, Kyne G, Bedore M, Menon S, Canan S, von Geldern T, Townson S, Gokool S, Ehrens A, Koschel M, Lhermitte-Vallarino N, Martin C, Hoerauf A, Hernandez G, Dalvie D, Specht S, Hübner MP, Scandale I. Discovery of Substituted Di(pyridin-2-yl)-1,2,4-thiadiazol-5-amines as Novel Macrofilaricidal Compounds for the Treatment of Human Filarial Infections. J Med Chem 2022; 65:11388-11403. [PMID: 35972896 PMCID: PMC9421654 DOI: 10.1021/acs.jmedchem.2c00960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Filarial diseases, including lymphatic filariasis and onchocerciasis, are considered among the most devastating of all tropical diseases, affecting about 145 million people worldwide. Efforts to control and eliminate onchocerciasis are impeded by a lack of effective treatments that target the adult filarial stage. Herein, we describe the discovery of a series of substituted di(pyridin-2-yl)-1,2,4-thiadiazol-5-amines as novel macrofilaricides for the treatment of human filarial infections.
Collapse
Affiliation(s)
- Natalie Hawryluk
- Bristol Myers Squibb, San Diego, California 92121, United States
| | - Dale Robinson
- Bristol Myers Squibb, San Diego, California 92121, United States
| | - Yixing Shen
- Bristol Myers Squibb, San Diego, California 92121, United States
| | - Graham Kyne
- Zoetis, Kalamazoo, Michigan 49001, United States
| | | | - Sanjay Menon
- Zoetis, Kalamazoo, Michigan 49001, United States
| | - Stacie Canan
- Bristol Myers Squibb, San Diego, California 92121, United States
| | | | - Simon Townson
- Northwick Park Institute for Medical Research, London HA1 3UJ, UK
| | - Suzanne Gokool
- Northwick Park Institute for Medical Research, London HA1 3UJ, UK
| | - Alexandra Ehrens
- Institute for Medical Microbiology, Immunology & Parasitology, University Hospital Bonn, 53127 Bonn, Germany.,German Center for Infection Research (DZIF), partner site Bonn-Cologne, 53127 Bonn, Germany
| | - Marianne Koschel
- Institute for Medical Microbiology, Immunology & Parasitology, University Hospital Bonn, 53127 Bonn, Germany
| | - Nathaly Lhermitte-Vallarino
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum national d'Histoire Naturelle, Paris 75005, France
| | - Coralie Martin
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum national d'Histoire Naturelle, Paris 75005, France
| | - Achim Hoerauf
- Institute for Medical Microbiology, Immunology & Parasitology, University Hospital Bonn, 53127 Bonn, Germany.,German Center for Infection Research (DZIF), partner site Bonn-Cologne, 53127 Bonn, Germany
| | | | - Deepak Dalvie
- Bristol Myers Squibb, San Diego, California 92121, United States
| | - Sabine Specht
- Institute for Medical Microbiology, Immunology & Parasitology, University Hospital Bonn, 53127 Bonn, Germany.,Drugs for Neglected Diseases Initiative, Geneva 1204, Switzerland
| | - Marc Peter Hübner
- Institute for Medical Microbiology, Immunology & Parasitology, University Hospital Bonn, 53127 Bonn, Germany.,German Center for Infection Research (DZIF), partner site Bonn-Cologne, 53127 Bonn, Germany
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative, Geneva 1204, Switzerland
| |
Collapse
|
19
|
Spruill ML, Maletic-Savatic M, Martin H, Li F, Liu X. Spatial analysis of drug absorption, distribution, metabolism, and toxicology using mass spectrometry imaging. Biochem Pharmacol 2022; 201:115080. [PMID: 35561842 PMCID: PMC9744413 DOI: 10.1016/j.bcp.2022.115080] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 12/14/2022]
Abstract
Mass spectrometry imaging (MSI) is emerging as a powerful analytical tool for detection, quantification, and simultaneous spatial molecular imaging of endogenous and exogenous molecules via in situ mass spectrometry analysis of thin tissue sections without the requirement of chemical labeling. The MSI generates chemically specific and spatially resolved ion distribution information for administered drugs and metabolites, which allows numerous applications for studies involving various stages of drug absorption, distribution, metabolism, excretion, and toxicity (ADMET). MSI-based pharmacokinetic imaging analysis provides a histological context and cellular environment regarding dynamic drug distribution and metabolism processes, and facilitates the understanding of the spatial pharmacokinetics and pharmacodynamic properties of drugs. Herein, we discuss the MSI's current technological developments that offer qualitative, quantitative, and spatial location information of small molecule drugs, antibody, and oligonucleotides macromolecule drugs, and their metabolites in preclinical and clinical tissue specimens. We highlight the macro and micro drug-distribution in the whole-body, brain, lung, liver, kidney, stomach, intestine tissue sections, organoids, and the latest applications of MSI in pharmaceutical ADMET studies.
Collapse
Affiliation(s)
- Michelle L Spruill
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Mirjana Maletic-Savatic
- Department of Pediatrics, Baylor College of Medicine, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Feng Li
- Center for Drug Discovery and Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; NMR and Drug Metabolism Core, Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
20
|
Tu D, Ning J, Zou L, Wang P, Zhang Y, Tian X, Zhang F, Zheng J, Ge G. Unique Oxidative Metabolism of Bufalin Generates Two Reactive Metabolites That Strongly Inactivate Human Cytochrome P450 3A. J Med Chem 2022; 65:4018-4029. [DOI: 10.1021/acs.jmedchem.1c01875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jing Ning
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Liwei Zou
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Wang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yani Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiangge Tian
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine Intervention, College of Integrative Medicine, National & Local Joint Engineering Research Center for Drug Development of Neurodegenerative Disease, College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550025, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
21
|
Nguyen T, Gamage TF, Finlay DB, Decker AM, Langston TL, Barrus D, Glass M, Li JX, Kenakin TP, Zhang Y. Development of 3-(4-Chlorophenyl)-1-(phenethyl)urea Analogues as Allosteric Modulators of the Cannabinoid Type-1 Receptor: RTICBM-189 is Brain Penetrant and Attenuates Reinstatement of Cocaine-Seeking Behavior. J Med Chem 2021; 65:257-270. [PMID: 34929081 DOI: 10.1021/acs.jmedchem.1c01432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We have shown that CB1 receptor negative allosteric modulators (NAMs) attenuated the reinstatement of cocaine-seeking behaviors in rats. In an effort to further define the structure-activity relationships and assess the druglike properties of the 3-(4-chlorophenyl)-1-(phenethyl)urea-based CB1 NAMs that we recently reported, we introduced substituents of different electronic properties and sizes to the phenethyl group and evaluated their potency in CB1 calcium mobilization, cAMP, and GTPγS assays. We found that 3-position substitutions such as Cl, F, and Me afforded enhanced CB1 potency, whereas 4-position analogues were generally less potent. The 3-chloro analogue (31, RTICBM-189) showed no activity at >50 protein targets and excellent brain permeation but relatively low metabolic stability in rat liver microsomes. Pharmacokinetic studies in rats confirmed the excellent brain exposure of 31 with a brain/plasma ratio Kp of 2.0. Importantly, intraperitoneal administration of 31 significantly and selectively attenuated the reinstatement of the cocaine-seeking behavior in rats without affecting locomotion.
Collapse
Affiliation(s)
- Thuy Nguyen
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Thomas F Gamage
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - David B Finlay
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ann M Decker
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Tiffany L Langston
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Daniel Barrus
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| | - Michelle Glass
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Jun-Xu Li
- Department of Pharmacology and Toxicology, University at Buffalo, the State University of New York, Buffalo, New York 14214, United States
| | - Terry P Kenakin
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, Research Triangle Park, North Carolina 27709, United States
| |
Collapse
|
22
|
Pal R, Singh K, Khan SA, Chawla P, Kumar B, Akhtar MJ. Reactive metabolites of the anticonvulsant drugs and approaches to minimize the adverse drug reaction. Eur J Med Chem 2021; 226:113890. [PMID: 34628237 DOI: 10.1016/j.ejmech.2021.113890] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 09/01/2021] [Accepted: 09/29/2021] [Indexed: 12/22/2022]
Abstract
Several generations of antiepileptic drugs (AEDs) are available in the market for the treatment of seizures, but these are amalgamated with acute to chronic side effects. The most common side effects of AEDs are dose-related, but some are idiosyncratic adverse drug reactions (ADRs) that transpire due to the formation of reactive metabolite (RM) after the bioactivation process. Because of the adverse reactions patients usually discontinue the medication in between the treatment. The AEDs such as valproic acid, lamotrigine, phenytoin etc., can be categorized under such types because they form the RM which may prevail with life-threatening adverse effects or immune-mediated reactions. Hepatotoxicity, teratogenicity, cutaneous hypersensitivity, dizziness, addiction, serum sickness reaction, renal calculi, metabolic acidosis are associated with the metabolites of drugs such as arene oxide, N-desmethyldiazepam, 2-(1-hydroxyethyl)-2-methylsuccinimide, 2-(sulphamoy1acetyl)-phenol, E-2-en-VPA and 4-en-VPA and carbamazepine-10,11-epoxide, etc. The major toxicities are associated with the moieties that are either capable of forming RM or the functional groups may itself be too reactive prior to the metabolism. These functional groups or fragment structures are typically known as structural alerts or toxicophores. Therefore, minimizing the bioactivation potential of lead structures in the early phases of drug discovery by a modification to low-risk drug molecules is a priority for the pharmaceutical companies. Additionally, excellent potency and pharmacokinetic (PK) behaviour help in ensuring that appropriate (low dose) candidate drugs progress into the development phase. The current review discusses about RMs in the anticonvulsant drugs along with their mechanism vis-a-vis research efforts that have been taken to minimize the toxic effects of AEDs therapy.
Collapse
Affiliation(s)
- Rohit Pal
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur, G.T. Road, Moga, 142001, Punjab, India
| | - Karanvir Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur, G.T. Road, Moga, 142001, Punjab, India
| | - Shah Alam Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, National University of Science and Technology, PO 620, PC 130, Azaiba, Bousher, Muscat, Oman
| | - Pooja Chawla
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur, G.T. Road, Moga, 142001, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur, G.T. Road, Moga, 142001, Punjab, India.
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ghal Kalan, Ferozpur, G.T. Road, Moga, 142001, Punjab, India; Department of Pharmaceutical Chemistry, College of Pharmacy, National University of Science and Technology, PO 620, PC 130, Azaiba, Bousher, Muscat, Oman.
| |
Collapse
|
23
|
Del Amo EM, Hammid A, Tausch M, Toropainen E, Sadeghi A, Valtari A, Puranen J, Reinisalo M, Ruponen M, Urtti A, Sauer A, Honkakoski P. Ocular metabolism and distribution of drugs in the rabbit eye: Quantitative assessment after intracameral and intravitreal administrations. Int J Pharm 2021; 613:121361. [PMID: 34896561 DOI: 10.1016/j.ijpharm.2021.121361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 10/19/2022]
Abstract
Quantitation of ocular drug metabolism is important, but only sparse data is currently available. Herein, the pharmacokinetics of four drugs, substrates of metabolizing enzymes, was investigated in albino rabbit eyes after intracameral and intravitreal administrations. Acetaminophen, brimonidine, cefuroxime axetil, and sunitinib and their corresponding metabolites were quantitated in the cornea, iris-ciliary body, aqueous humor, lens, vitreous humor, and neural retina with LC-MS/MS analytics. Non-compartmental analysis was employed to estimate the pharmacokinetic parameters of the parent drugs and metabolites. The area under the curve (AUC) values of metabolites were 12-70 times lower than the AUC values of the parent drugs in the tissues with the highest enzymatic activity. The ester prodrug cefuroxime axetil was an exception because it was efficiently and quantitatively converted to cefuroxime in the ocular tissues. In contrast to the liver, sulfotransferases, aldehyde oxidase, and cytochrome P450 3A activities were low in the eye and they had negligible impact on ocular drug clearance. With the exception of esterase substrates, metabolism seems to be a minor player in ocular pharmacokinetics. However, metabolites might contribute to ocular toxicity, and drug metabolism in various eye tissues should be investigated and understood thoroughly.
Collapse
Affiliation(s)
- Eva M Del Amo
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland.
| | - Anam Hammid
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | | | - Elisa Toropainen
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Amir Sadeghi
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Annika Valtari
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Jooseppi Puranen
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Mika Reinisalo
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Marika Ruponen
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| | - Arto Urtti
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland; University of Helsinki, Faculty of Pharmacy, Drug Research Program, Yliopistonkatu 3, 00014 Helsinki, Finland; Saint-Petersburg State University, Institute of Chemistry, Universitetskiy Prospekt, 26, Petergoff 198504, Russian Federation
| | - Achim Sauer
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach, Germany
| | - Paavo Honkakoski
- University of Eastern Finland, School of Pharmacy, Biopharmaceutics, Yliopistonranta 1, 70210 Kuopio, Finland
| |
Collapse
|
24
|
Abstract
The use of artificial intelligence methods in drug safety began in the early 2000s with applications such as predicting bacterial mutagenicity and hERG inhibition. The field has been endlessly expanding ever since and the models have become more complex. These approaches are now integrated into molecule risk assessment processes along with in vitro and in vivo methods. Today, artificial intelligence can be used in every phase of drug discovery and development, from profiling chemical libraries in early discovery, to predicting off-target effects in the mid-discovery phase, to assessing potential mutagenic impurities in development and degradants as part of life cycle management. This chapter provides an overview of artificial intelligence in drug safety and describes its application throughout the entire discovery and development process.
Collapse
|
25
|
Jaladanki CK, Khatun S, Gohlke H, Bharatam PV. Reactive Metabolites from Thiazole-Containing Drugs: Quantum Chemical Insights into Biotransformation and Toxicity. Chem Res Toxicol 2021; 34:1503-1517. [PMID: 33900062 DOI: 10.1021/acs.chemrestox.0c00450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Drugs containing thiazole and aminothiazole groups are known to generate reactive metabolites (RMs) catalyzed by cytochrome P450s (CYPs). These RMs can covalently modify essential cellular macromolecules and lead to toxicity and induce idiosyncratic adverse drug reactions. Molecular docking and quantum chemical hybrid DFT study were carried out to explore the molecular mechanisms involved in the biotransformation of thiazole (TZ) and aminothiazole (ATZ) groups leading to RM epoxide, S-oxide, N-oxide, and oxaziridine. The energy barrier required for the epoxidation is 13.63 kcal/mol, that is lower than that of S-oxidation, N-oxidation, and oxaziridine formation (14.56, 17.90, and 20.20, kcal/mol respectively). The presence of the amino group in ATZ further facilitates all the metabolic pathways, for example, the barrier for the epoxidation reaction is reduced by ∼2.5 kcal/mol. Some of the RMs/their isomers are highly electrophilic and tend to form covalent bonds with nucleophilic amino acids, finally leading to the formation of metabolic intermediate complexes (MICs). The energy profiles of these competitive pathways have also been explored.
Collapse
Affiliation(s)
- Chaitanya K Jaladanki
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector -67, S. A. S. Nagar (Mohali), 160 062 Punjab, India
| | - Samima Khatun
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector -67, S. A. S. Nagar (Mohali), 160 062 Punjab, India
| | - Holger Gohlke
- Institut für Pharmazeutische und Medizinische Chemie, Heinrich-Heine-Universität Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.,Forschungszentrum Jülich GmbH, John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), and Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Wilhelm-Johnen-Straße, 52425 Jülich, Germany
| | - Prasad V Bharatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector -67, S. A. S. Nagar (Mohali), 160 062 Punjab, India
| |
Collapse
|
26
|
Hammond S, Thomson P, Meng X, Naisbitt D. In-Vitro Approaches to Predict and Study T-Cell Mediated Hypersensitivity to Drugs. Front Immunol 2021; 12:630530. [PMID: 33927714 PMCID: PMC8076677 DOI: 10.3389/fimmu.2021.630530] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/17/2021] [Indexed: 01/11/2023] Open
Abstract
Mitigating the risk of drug hypersensitivity reactions is an important facet of a given pharmaceutical, with poor performance in this area of safety often leading to warnings, restrictions and withdrawals. In the last 50 years, efforts to diagnose, manage, and circumvent these obscure, iatrogenic diseases have resulted in the development of assays at all stages of a drugs lifespan. Indeed, this begins with intelligent lead compound selection/design to minimize the existence of deleterious chemical reactivity through exclusion of ominous structural moieties. Preclinical studies then investigate how compounds interact with biological systems, with emphasis placed on modeling immunological/toxicological liabilities. During clinical use, competent and accurate diagnoses are sought to effectively manage patients with such ailments, and pharmacovigilance datasets can be used for stratification of patient populations in order to optimise safety profiles. Herein, an overview of some of the in-vitro approaches to predict intrinsic immunogenicity of drugs and diagnose culprit drugs in allergic patients after exposure is detailed, with current perspectives and opportunities provided.
Collapse
Affiliation(s)
- Sean Hammond
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
- ApconiX, Alderley Park, Alderley Edge, United Kingdom
| | - Paul Thomson
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| | - Dean Naisbitt
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
27
|
Ye Y, Guo X, He X, Zhang M, He H, Qiu D, Guo Z. High-resolution mass spectrometry-based approach for the identification and profiling of the metabolites of taletrectinib formed in liver microsomes. Drug Test Anal 2021; 13:1118-1126. [PMID: 33527739 DOI: 10.1002/dta.3008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/25/2021] [Accepted: 01/27/2021] [Indexed: 11/08/2022]
Abstract
Taletrectinib is a potent, orally active, and selective ROS1/NTRK kinase inhibitor. The aim of this study was to study the metabolism of taletrectinib in rat, dog, and human liver microsomes. The biotransformation of taletrectinib was carried out using rat, dog, and human liver microsomes supplemented with nicotinamide adenine dinucleotide phosphate tetrasodium salt (NADPH) and uridine diphosphate glucuronic acid (UDPGA). The microsomal incubations were conducted at 37°C for 60 min. The formed metabolites were identified by ultrahigh performance liquid chromatography coupled to high-resolution tandem mass spectrometry (UHPLC-HRMS) using electrospray ionization in the positive ion mode. They were identified by accurate masses and MS/MS spectra and based on their fragmentation pathways. With UHPLC-HRMS, a total of 10 metabolites including one glucuronide conjugate (M7) were structurally identified. M9 and M10 were unambiguously identified as taletrectinib alcohol and taletrectinib ketone, respectively, using reference standards. The phase I metabolic pathways of taletrectinib involved N-dealkylation, O-dealkylation, oxidative deamination, and oxygenation; the phase II metabolic pathways referred to glucuronidation. The current study investigated the in vitro metabolic fate of taletrectinib in animals and human species, which would bring us considerable benefits for the subsequent studies focusing on the pharmacological effect and toxicity of this drug.
Collapse
Affiliation(s)
- Yongbin Ye
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Xiaojuan Guo
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Xin He
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Mingwan Zhang
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Huiqing He
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Dafa Qiu
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| | - Ziwen Guo
- Department of Hematology, Zhongshan Hospital of Sun Yat-Sen University & Zhongshan City People's Hospital, Zhongshan, China
| |
Collapse
|
28
|
Wedlake AJ, Allen TEH, Goodman JM, Gutsell S, Kukic P, Russell PJ. Confidence in Inactive and Active Predictions from Structural Alerts. Chem Res Toxicol 2020; 33:3010-3022. [PMID: 33295767 DOI: 10.1021/acs.chemrestox.0c00332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Having a measure of confidence in computational predictions of biological activity from in silico tools is vital when making predictions for new chemicals, for example, in chemical risk assessment. Where predictions of biological activity are used as an indicator of a potential hazard, false-negative predictions are the most concerning prediction; however, assigning confidence in inactive predictions is particularly challenging. How can one confidently identify the absence of activating features? In this study, we present methods for assigning confidence to both active and inactive predictions from structural alerts for protein-binding molecular initiating events (MIEs). Structural alerts were derived through an iterative statistical method. Confidence in the activity predictions is assigned by measuring the Tanimoto similarity between Morgan fingerprints of chemicals in the test set to relevant chemicals in the training set, and suitable cutoff values have been defined to give different confidence categories. To avoid a potential compound series bias in the test set and hence overestimate the performance of the method, we measured the biological activity of 27 compounds with 24 proteins, which gave us an additional 648 experimental measurements; many of the measurements are currently nonexistent in the literature and databases. This data set was complemented with newly measured biological activities published in ChEMBL25 and formed a combined independent validation data set. Applying the confidence categories to the computational predictions for the new data leads to the identification of chemicals for which one should be confident of either an inactive or active prediction, allowing model predictions to be used responsibly.
Collapse
Affiliation(s)
- Andrew J Wedlake
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Timothy E H Allen
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom.,MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, United Kingdom
| | - Jonathan M Goodman
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Steve Gutsell
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| | - Paul J Russell
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire MK44 1LQ, United Kingdom
| |
Collapse
|
29
|
Sodano TM, Combee LA, Stephenson CRJ. Recent Advances and Outlook for the Isosteric Replacement of Anilines. ACS Med Chem Lett 2020; 11:1785-1788. [PMID: 33062152 DOI: 10.1021/acsmedchemlett.9b00687] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Many promising drug candidates and pharmaceutical compounds fail due to idiosyncratic adverse drug reactions (IADRs), often arising from the formation of reactive metabolites. Among the "structural alerts" responsible, anilines are well-known to undergo deleterious metabolic processing, yet isosteric replacement strategies remain limited. Herein we discuss current art and potential new avenues of saturated isosteres to mitigate aniline-related toxicities.
Collapse
Affiliation(s)
- Taylor M. Sodano
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Logan A. Combee
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Corey R. J. Stephenson
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
30
|
Agnew-Francis KA, Williams CM. Squaramides as Bioisosteres in Contemporary Drug Design. Chem Rev 2020; 120:11616-11650. [DOI: 10.1021/acs.chemrev.0c00416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Kylie A. Agnew-Francis
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Craig M. Williams
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
31
|
Application of a fluorous derivatization method for characterization of glutathione-trapped reactive metabolites with liquid chromatography-tandem mass spectrometry analysis. J Chromatogr A 2020; 1622:461160. [PMID: 32450990 DOI: 10.1016/j.chroma.2020.461160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 01/05/2023]
Abstract
The glutathione (GSH) trapping assay is commonly utilized for the screening and characterization of reactive metabolites produced by drug metabolism. This study describes a fluorous derivatization method for a more sensitive and selective analysis of reactive metabolites trapped by GSH using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In this study, the GSH-trapped reactive metabolites, which were obtained after incubation of the test compounds with human liver microsome (HLM) in the presence of GSH and NADPH, were derivatized using the perfluoroalkylamine reagent through oxazolone chemistry. Since this reaction enabled the selective modification of the α-carboxyl group in GSH, the structural compositions of the metabolites were not affected by the derivatization. Furthermore, the selective analysis of the resulting derivatives could be performed using perfluoroalkyl-modified stationary phase LC separation via the interaction between the perfluoroalkyl-containing compounds, such as fluorous affinity, followed by detection with the precursor ion and/or enhanced product ion scan modes in MS/MS. Finally, we demonstrated the applicability of this method by analyzing perfluoroalkyl derivatives of some drug metabolites trapped by GSH in HLM incubation.
Collapse
|
32
|
Norman BH. Drug Induced Liver Injury (DILI). Mechanisms and Medicinal Chemistry Avoidance/Mitigation Strategies. J Med Chem 2020; 63:11397-11419. [PMID: 32511920 DOI: 10.1021/acs.jmedchem.0c00524] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Adverse drug reactions (ADRs) are a common cause of attrition in drug discovery and development and drug-induced liver injury (DILI) is a leading cause of preclinical and clinical drug terminations. This perspective outlines many of the known DILI mechanisms and assessment methods used to evaluate and mitigate DILI risk. Literature assessments and retrospective analyses using verified DILI-associated drugs from the Liver Tox Knowledge Base (LTKB) have been used to derive the predictive value of each end point, along with combination approaches of multiple methods. In vitro assays to assess inhibition of the bile salt export pump (BSEP), mitotoxicity, reactive metabolite (RM) formation, and hepatocyte cytolethality, along with physicochemical properties and clinical dose provide useful DILI predictivity. This Perspective also highlights some of the strategies used by medicinal chemists to reduce DILI risk during the optimization of drug candidates.
Collapse
Affiliation(s)
- Bryan H Norman
- Norman Drug Discovery Training and Consulting, LLC, 8540 Bluefin Circle, Indianapolis, Indiana 46236, United States
| |
Collapse
|
33
|
Tu DZ, Mao X, Zhang F, He RJ, Wu JJ, Wu Y, Zhao XH, Zheng J, Ge GB. Reversible and Irreversible Inhibition of Cytochrome P450 Enzymes by Methylophiopogonanone A. Drug Metab Dispos 2020; 49:459-469. [PMID: 33811108 DOI: 10.1124/dmd.120.000325] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Methylophiopogonanone A (MOA), an abundant homoisoflavonoid bearing a methylenedioxyphenyl moiety, is one of the major constituents in the Chinese herb Ophiopogon japonicas This work aims to assess the inhibitory potentials of MOA against cytochrome P450 enzymes and to decipher the molecular mechanisms for P450 inhibition by MOA. The results showed that MOA concentration-dependently inhibited CYP1A, 2C8, 2C9, 2C19, and 3A in human liver microsomes (HLMs) in a reversible way, with IC50 values varying from 1.06 to 3.43 μM. By contrast, MOA time-, concentration-, and NADPH-dependently inhibited CYP2D6 and CYP2E1, along with KI and kinact values of 207 µM and 0.07 minute-1 for CYP2D6, as well as 20.9 µM and 0.03 minutes-1 for CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in an HLM incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate. Additionally, the potential risks of herb-drug interactions triggered by MOA or MOA-related products were also predicted. Collectively, our findings verify that MOA is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. SIGNIFICANCE STATEMENT: Methylophiopogonanone A (MOA), an abundant homoisoflavonoid isolated from the Chinese herb Ophiopogon japonicas, is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in a human liver microsome incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate.
Collapse
Affiliation(s)
- Dong-Zhu Tu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xu Mao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jing-Jing Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Yue Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xiao-Hua Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jiang Zheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| |
Collapse
|
34
|
Huang L, Li Y, Pan H, Lu Y, Zhou X, Shi F. Cortex dictamni-induced liver injury in mice: The role of P450-mediated metabolic activation of furanoids. Toxicol Lett 2020; 330:41-52. [PMID: 32437846 DOI: 10.1016/j.toxlet.2020.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/11/2020] [Accepted: 05/05/2020] [Indexed: 01/18/2023]
Abstract
Many furan containing compounds have been reported to be toxic resulted from the metabolic activation of the furan ring to reactive metabolite (RM). Cortex Dictamni (CD), a widely used herbal medicine, has been reported to cause severe even fatal hepatotoxicity. The injurious components and mechanism of CD-induced liver injury remain unclear. Our preliminary study showed that dictamnine, one major furanoid in CD, caused mouse liver injury via its reactive epoxide metabolite. Besides dictamnine, the major components of CD are series of bioactivation-alerting furanoids. Thus, we hypothesize that series of furanoids in CD may undergo metabolic activation and play a key role in CD-induced liver injury. Here, a single oral dose of 60 g/kg ethanol extract of CD (ECD) caused severe hepatocellular necrosis in mice at 24 h post-dose. ECD-induced liver injury showed a dose- and time-dependent manner. The hepatotoxic effects could be completely abolished by P450 nonselective inhibitor 1-aminobenzotriazole (ABT) and strongly modulated by other P450 modulators. The furanoids-concentrated fraction of ECD was responsible for the hepatotoxicity. At least ten furanoids with high abundance in ECD, such as obakunone, dictamnine, fraxinellone, limonin, were found to be metabolized to reactive epoxide or cis-enedione. The RM levels were consistent with the liver injury degree. Multiple furanoids, rather than single one, cooperatively contributed to the hepatotoxicity. ECD-induced liver injury could be reproduced by a mixture of pure furanoids. In summary, this study provides toxic component profiles of CD and demonstrates that P450-mediated bioactivation of multiple furanoids is responsible for CD-induced liver injury.
Collapse
Affiliation(s)
- Linyan Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China; Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi 563003, China
| | - Yi Li
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Hong Pan
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi 563003, China
| | - Yuanfu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China
| | - Xumei Zhou
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi 563003, China.
| | - Fuguo Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563003, China.
| |
Collapse
|
35
|
Cerny MA, Kalgutkar AS, Obach RS, Sharma R, Spracklin DK, Walker GS. Effective Application of Metabolite Profiling in Drug Design and Discovery. J Med Chem 2020; 63:6387-6406. [PMID: 32097005 DOI: 10.1021/acs.jmedchem.9b01840] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
At one time, biotransformation was a descriptive activity in pharmaceutical development, viewed simply as structural elucidation of drug metabolites, completed only once compounds entered clinical development. Herein, we present our strategic approach using structural elucidation to enable chemistry design/SAR development. The approach considers four questions that often present themselves to medicinal chemists optimizing their compounds for candidate selection: (1) What are the important clearance mechanisms that mediate the disposition of my molecule? (2) Can metabolic liabilities be modulated in a favorable way? (3) Does my compound undergo bioactivation to a reactive metabolite? (4) Do any of the metabolites possess activity, either on- or off-target? An additional question necessary to support compound development relates to metabolites in safety testing (MIST) and our approach also addresses this question. The value in structural elucidation is derived from its application to better design molecules, guide their clinical development, and underwrite patient safety.
Collapse
Affiliation(s)
- Matthew A Cerny
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, 1 Portland Street, Cambridge Massachusetts 02139, United States
| | - R Scott Obach
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Raman Sharma
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Douglas K Spracklin
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Gregory S Walker
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
36
|
Yang H, Lou C, Li W, Liu G, Tang Y. Computational Approaches to Identify Structural Alerts and Their Applications in Environmental Toxicology and Drug Discovery. Chem Res Toxicol 2020; 33:1312-1322. [DOI: 10.1021/acs.chemrestox.0c00006] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Hongbin Yang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chaofeng Lou
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
37
|
Yang ZY, He JH, Lu AP, Hou TJ, Cao DS. Application of Negative Design To Design a More Desirable Virtual Screening Library. J Med Chem 2020; 63:4411-4429. [DOI: 10.1021/acs.jmedchem.9b01476] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Zi-Yi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Jun-Hong He
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
| | - Ai-Ping Lu
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P. R. China
| | - Ting-Jun Hou
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P. R. China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, P. R. China
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, SAR, P. R. China
| |
Collapse
|
38
|
Wedlake AJ, Folia M, Piechota S, Allen TEH, Goodman JM, Gutsell S, Russell PJ. Structural Alerts and Random Forest Models in a Consensus Approach for Receptor Binding Molecular Initiating Events. Chem Res Toxicol 2020; 33:388-401. [PMID: 31850746 DOI: 10.1021/acs.chemrestox.9b00325] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A molecular initiating event (MIE) is the gateway to an adverse outcome pathway (AOP), a sequence of events ending in an adverse effect. In silico predictions of MIEs are a vital tool in a modern, mechanism-focused approach to chemical risk assessment. For 90 biological targets representing important human MIEs, structural alert-based models have been constructed with an automated procedure that uses Bayesian statistics to iteratively select substructures. These models give impressive average performance statistics (an average of 92% correct predictions across targets), significantly improving on previous models. Random Forest models have been constructed from physicochemical features for the same targets, giving similarly impressive performance statistics (93% correct predictions). A key difference between the models is interpretation of predictions-the structural alert models are transparent and easy to interpret, while Random Forest models can only identify the most important physicochemical features for making predictions. The two complementary models have been combined in a consensus model, improving performance compared to each individual model (94% correct predictions) and increasing confidence in predictions. Variation in model performance has been explained by calculating a modelability index (MODI), using Tanimoto coefficient between Morgan fingerprints to identify nearest neighbor chemicals. This work is an important step toward building confidence in the use of in silico tools for assessment of toxicity.
Collapse
Affiliation(s)
- Andrew J Wedlake
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , United Kingdom
| | - Maria Folia
- Unilever Safety and Environmental Assurance Centre , Colworth Science Park , Sharnbrook , Bedfordshire , MK44 1LQ , United Kingdom
| | - Sam Piechota
- Unilever Safety and Environmental Assurance Centre , Colworth Science Park , Sharnbrook , Bedfordshire , MK44 1LQ , United Kingdom
| | - Timothy E H Allen
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , United Kingdom.,MRC Toxicology Unit , University of Cambridge , Lancaster Road , Leicester LE19HN , United Kingdom
| | - Jonathan M Goodman
- Centre for Molecular Informatics, Department of Chemistry , University of Cambridge , Lensfield Road , Cambridge , CB2 1EW , United Kingdom
| | - Steve Gutsell
- Unilever Safety and Environmental Assurance Centre , Colworth Science Park , Sharnbrook , Bedfordshire , MK44 1LQ , United Kingdom
| | - Paul J Russell
- Unilever Safety and Environmental Assurance Centre , Colworth Science Park , Sharnbrook , Bedfordshire , MK44 1LQ , United Kingdom
| |
Collapse
|
39
|
Liu N, Liu H, Zhang W, Yao H. Metabolic profiles of ribociclib in rat and human liver microsomes using liquid chromatography combined with electrospray ionization high‐resolution mass spectrometry. Biomed Chromatogr 2020; 34:e4783. [PMID: 31899811 DOI: 10.1002/bmc.4783] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Na Liu
- Department of PharmacyJining No. 1 People's Hospital Jining Shandong Province China
- Jining Medical University Jining Shandong Province China
| | - Hongqiang Liu
- Department of PharmacyJining No. 1 People's Hospital Jining Shandong Province China
- Jining Medical University Jining Shandong Province China
| | - Wucheng Zhang
- Department of Clinical MedicineLinyi People's Hospital Dezhou Shandong Province China
| | - Huijie Yao
- Department of PharmacyJining Beihu Provincial Tourism Resort People's Hospital Jining Shandong Province China
| |
Collapse
|
40
|
Paludetto MN, Stigliani JL, Robert A, Bernardes-Génisson V, Chatelut E, Puisset F, Arellano C. Involvement of Pazopanib and Sunitinib Aldehyde Reactive Metabolites in Toxicity and Drug-Drug Interactions in Vitro and in Patient Samples. Chem Res Toxicol 2019; 33:181-190. [PMID: 31535851 DOI: 10.1021/acs.chemrestox.9b00205] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tyrosine kinase inhibitors (TKI) are targeted anticancer drugs that have been successfully developed over the past 2 decades. To date, many of them (around 70%) require warnings for liver injury and five of them, including pazopanib and sunitinib, have Black Box Warning (BBW) labels. Although TKI-induced hepatotoxicity is the first cause of drug failures in clinical trials, BBW labels, and market withdrawals, the underlying mechanisms remain unclear. However, the recent discovery of new reactive metabolites (RM) with aldehyde structures during pazopanib and sunitinib metabolism offers new perspectives for investigating their involvement in the toxicity of these two TKI. These hard electrophiles have a high reactivity potential toward proteins and are thought to be responsible for cytochrome P450 inactivation, drug-drug interactions (DDI), and liver toxicity. We report here, for the first time, the presence of these aldehyde RM in human plasma samples obtained during drug monitoring. Docking experiments in the CYP3A4 active site were performed and showed that pazopanib and sunitinib fitting in the catalytic site are in accordance with their regioselective oxidation to aldehydes. They also suggested that aldehyde RM may react with lysine and arginine residues. Based on these results, we studied the reactivity of the aldehyde RM toward lysine and arginine residues as potential targets on the protein framework to better understand how these RM could be involved in liver toxicity and drug-drug interactions. Adduct formation with different hepatic and plasma proteins was investigated by LC-MS/MS, and adducts between pazopanib or sunitinib aldehyde derivatives and lysine residues on both CYP3A4 and plasma proteins were indeed shown for the first time.
Collapse
Affiliation(s)
- Marie-Noëlle Paludetto
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037 , Université de Toulouse , 2 Avenue Hubert Curien, CS53717 , 31037 Toulouse , Cedex 1, France.,Université Paul Sabatier , 31330 Toulouse , France.,Institut Claudius-Regaud, IUCT-O , 31059 Toulouse , Cedex 9, France
| | - Jean-Luc Stigliani
- Université Paul Sabatier , 31330 Toulouse , France.,Laboratoire de Chimie de Coordination du CNRS (LCC-CNRS) , Université de Toulouse , 205 Route de Narbonne, BP 44099 , 31077 Toulouse , Cedex 4, France
| | - Anne Robert
- Laboratoire de Chimie de Coordination du CNRS (LCC-CNRS) , Université de Toulouse , 205 Route de Narbonne, BP 44099 , 31077 Toulouse , Cedex 4, France
| | - Vania Bernardes-Génisson
- Université Paul Sabatier , 31330 Toulouse , France.,Laboratoire de Chimie de Coordination du CNRS (LCC-CNRS) , Université de Toulouse , 205 Route de Narbonne, BP 44099 , 31077 Toulouse , Cedex 4, France
| | - Etienne Chatelut
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037 , Université de Toulouse , 2 Avenue Hubert Curien, CS53717 , 31037 Toulouse , Cedex 1, France.,Université Paul Sabatier , 31330 Toulouse , France.,Institut Claudius-Regaud, IUCT-O , 31059 Toulouse , Cedex 9, France
| | - Florent Puisset
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037 , Université de Toulouse , 2 Avenue Hubert Curien, CS53717 , 31037 Toulouse , Cedex 1, France.,Université Paul Sabatier , 31330 Toulouse , France.,Institut Claudius-Regaud, IUCT-O , 31059 Toulouse , Cedex 9, France
| | - Cécile Arellano
- Centre de Recherches en Cancérologie de Toulouse (CRCT), INSERM UMR1037 , Université de Toulouse , 2 Avenue Hubert Curien, CS53717 , 31037 Toulouse , Cedex 1, France.,Université Paul Sabatier , 31330 Toulouse , France
| |
Collapse
|
41
|
Ji C, Guha M, Zhu X, Whritenour J, Hemkens M, Tse S, Walker GS, Evans E, Khan NK, Finkelstein MB, Callegari E, Obach RS. Enzalutamide and Apalutamide: In Vitro Chemical Reactivity Studies and Activity in a Mouse Drug Allergy Model. Chem Res Toxicol 2019; 33:211-222. [DOI: 10.1021/acs.chemrestox.9b00247] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Changhua Ji
- Drug Safety Research and Development, Pfizer Inc., La Jolla, California 92037, United States
| | - Mausumee Guha
- Drug Safety Research and Development, Pfizer Inc., La Jolla, California 92037, United States
| | - Xu Zhu
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Jessica Whritenour
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Michelle Hemkens
- Drug Safety Research and Development, Pfizer Inc., La Jolla, California 92037, United States
| | - Susanna Tse
- Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Gregory S. Walker
- Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Ellen Evans
- Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut 06340, United States
| | - Nasir K. Khan
- Drug Safety Research and Development, Pfizer Inc., Pearl River, New York 10965, United States
| | - Martin B. Finkelstein
- Drug Safety Research and Development, Pfizer Inc., Pearl River, New York 10965, United States
| | - Ernesto Callegari
- Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| | - R. Scott Obach
- Pharmacokinetics, Dynamics, and Drug Metabolism, Pfizer Inc., Groton, Connecticut 06340, United States
| |
Collapse
|
42
|
Paludetto M, Puisset F, Chatelut E, Arellano C. Identifying the reactive metabolites of tyrosine kinase inhibitors in a comprehensive approach: Implications for drug‐drug interactions and hepatotoxicity. Med Res Rev 2019; 39:2105-2152. [DOI: 10.1002/med.21577] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/06/2019] [Accepted: 03/08/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Marie‐Noëlle Paludetto
- Centre de Recherches en Cancérologie de Toulouse, INSERMUMR1037Université de Toulouse Toulouse Cedex 1 France
- Faculté de PharmacieUniversité Paul Sabatier Toulouse France
- Département PharmacieInstitut Claudius Regaud, IUCT‐O Toulouse France
| | - Florent Puisset
- Centre de Recherches en Cancérologie de Toulouse, INSERMUMR1037Université de Toulouse Toulouse Cedex 1 France
- Faculté de PharmacieUniversité Paul Sabatier Toulouse France
- Département PharmacieInstitut Claudius Regaud, IUCT‐O Toulouse France
| | - Etienne Chatelut
- Centre de Recherches en Cancérologie de Toulouse, INSERMUMR1037Université de Toulouse Toulouse Cedex 1 France
- Faculté de PharmacieUniversité Paul Sabatier Toulouse France
| | - Cécile Arellano
- Centre de Recherches en Cancérologie de Toulouse, INSERMUMR1037Université de Toulouse Toulouse Cedex 1 France
- Faculté de PharmacieUniversité Paul Sabatier Toulouse France
| |
Collapse
|
43
|
LoPachin RM, Geohagen BC, Nordstroem LU. Mechanisms of soft and hard electrophile toxicities. Toxicology 2019; 418:62-69. [PMID: 30826385 PMCID: PMC6494464 DOI: 10.1016/j.tox.2019.02.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/24/2019] [Accepted: 02/10/2019] [Indexed: 12/21/2022]
Abstract
Electron-deficient chemicals (electrophiles) react with compounds that have one or more unshared valence electron pairs (nucleophiles). The resulting covalent reactions between electrophiles and nucleophiles (e.g., Michael addition, SN2 reactions) are important, not only to Organic Chemistry, but also to the fields of Molecular Biology and Toxicology. Specifically, covalent bond formation is the operational basis of many critically important cellular processes; e.g., enzyme function, neurotransmitter release, and membrane-vesicle fusion. Given this context it is understandable that these reactions are also relevant to Toxicology, since a significant number of xenobiotic chemicals are toxic electrophiles that can react with endogenous nucleophilic residues. Therefore, the purpose of this Review is to discuss electrophile-nucleophile chemistry as it pertains to cell injury and resulting organ toxicity. Our discussion will involve an introduction to the Hard and Soft, Acids and Bases (HSAB) theory of Pearson. The HSAB concept provides a framework for calculation of quantum chemical parameters that classify the electrophile and nucleophile covalent components according to their respective electronic nature (softness/hardness) and reactivity (electrophilicity/nucleophilicity). The calculated quantum indices in conjunction with corroborative in vivo, in chemico (cell free) and in vitro research can offer an illuminating approach to mechanistic discovery. Accordingly, we will provide examples that demonstrate how this approach has been used to discern mechanisms and sites of electrophile action.
Collapse
Affiliation(s)
- Richard M LoPachin
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 E. 210th St, Bronx NY 10467, United States.
| | - Brian C Geohagen
- Department of Anesthesiology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 E. 210th St, Bronx NY 10467, United States
| | - Lars U Nordstroem
- The Chemical Synthesis & Biology Core Facility, Albert Einstein College of Medicine, Bronx, NY, United States
| |
Collapse
|
44
|
Grove KJ, Hoque S, Rudewicz PJ. Investigation of amodiaquine localization in liver lobules using matrix-assisted laser desorption/ionization imaging mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2019; 33:252-258. [PMID: 30394607 DOI: 10.1002/rcm.8339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/23/2018] [Accepted: 10/27/2018] [Indexed: 06/08/2023]
Abstract
RATIONALE High mass and high spatial resolution matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) opens new possibilities for the detailed study of the hepatic metabolism of drugs. The spatial and temporal distribution of drug metabolites within liver lobules, anatomical subunits of the liver, may aid in the understanding of the formation of reactive metabolites that bind to liver proteins and cause drug-induced liver injury. METHODS Frozen livers obtained from rats dosed orally with amodiaquine (100 mg/kg) were sectioned at 10 μm and coated with a MALDI matrix. The liver sections were then analyzed using a Fourier transform ion cyclotron resonance mass spectrometer. Images corresponding to amodiaquine and its metabolites were obtained at a spatial resolution of 25 μm. RESULTS Molecular images of amodiaquine within liver lobules have higher intensities near the portal triad and lower intensities near the central vein. CONCLUSIONS This study demonstrates that MALDI IMS can be used to investigate the metabolism of drugs within liver lobules. The results are consistent with existing knowledge of amodiaquine metabolism and reactive metabolite formation.
Collapse
Affiliation(s)
- Kerri J Grove
- Novartis Institutes for Biomedical Research, Emeryville, CA, 94608, USA
| | - Shaila Hoque
- Novartis Institutes for Biomedical Research, Emeryville, CA, 94608, USA
| | | |
Collapse
|
45
|
Bao XY, Zheng Q, Tong Q, Zhu PC, Zhuang Z, Zheng GQ, Wang Y. Danshensu for Myocardial Ischemic Injury: Preclinical Evidence and Novel Methodology of Quality Assessment Tool. Front Pharmacol 2018; 9:1445. [PMID: 30618743 PMCID: PMC6297803 DOI: 10.3389/fphar.2018.01445] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/22/2018] [Indexed: 12/09/2022] Open
Abstract
Background: Danshensu (DSS) possesses unique bioactivity on the cardiovascular system. However, there is a lack of systematical summary of DSS for acute myocardial ischemia injury and no quality assessment tool for the systematical review of cell experiments. Here, we aimed to assess the preclinical evidences and possible mechanisms of DSS for myocardial ischemia injury, and to develop a quality assessment tool for the systematical review of cell experiments. Methods: Thirty-two studies with 473 animals and 134 cells were identified by searching seven databases. All data analysis was performed using RevMan 5.3. CAMARADES 10-item checklist was used to assess the methodological quality of animal experiments. A new 10-item checklist was first developed to assess the methodological quality of cell studies. Results: The score of study quality ranged from 3 to 7 points in animal studies, while the cell studies scored 3–6 points. Meta-analysis showed that DSS had significant effects on reducing myocardial infarct (MI) size in vivo, and increasing cell viability and reducing apoptosis rate in vitro compared with controls (P < 0.01). The possible mechanisms of DSS for MI are improving circulation, antioxidant, anti-apoptosis, anti-inflammatory, promoting angiogenesis, anti-excessive autophagy, anti-calcium overload, and improving energy metabolism. Conclusions: DSS could exert cardioprotective effect on myocardial ischemia injury, and thus is a probable candidate for further clinical trials andtreatment of AMI. In addition, the newly devloped 10-item checklist for assessing methodological quality of cell study that recommened to use the sysmatic review of cell studies.
Collapse
Affiliation(s)
- Xiao-Yi Bao
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qun Zheng
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiang Tong
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Chong Zhu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuang Zhuang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Wang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Staveness D, Sodano TM, Li K, Burnham EA, Jackson KD, Stephenson CRJ. Providing a New Aniline Bioisostere through the Photochemical Production of 1-Aminonorbornanes. Chem 2018; 5:215-226. [PMID: 30873503 DOI: 10.1016/j.chempr.2018.10.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This report describes the photochemical conversion of aminocyclopropanes into 1-aminonorbornanes via formal [3+2] cycloadditions initiated by homolytic fragmentation of amine radical cation intermediates. Aligning with the modern movement toward sp 3 -rich motifs in drug discovery, this strategy provides access to a diverse array of substitution patterns on this saturated carbocyclic framework while offering the robust functional group tolerance (e.g. -OH, -NHBoc) necessary for further derivatization. Evaluating the metabolic stability of selected morpholine-based 1-aminonorbornanes demonstrated a low propensity for oxidative processing and no proclivity toward reactive metabolite formation, suggesting a potential bioisosteric role for 1-aminonorbornanes. Continuous flow processing allowed for efficient operation on gram-scale, providing promise for translation to industrially-relevant scales. This methodology only requires low loadings of a commercially-available, visible light-active photocatalyst and a simple salt, thus it stays true to sustainability goals while readily delivering saturated building blocks that can reduce metabolic susceptibility within drug development programs.
Collapse
Affiliation(s)
- Daryl Staveness
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Taylor M Sodano
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kangjun Li
- Department of Pharmaceutical Sciences, Lipscomb University, Nashville, TN 37204, USA
| | - Elizabeth A Burnham
- Department of Pharmaceutical Sciences, Lipscomb University, Nashville, TN 37204, USA
| | - Klarissa D Jackson
- Department of Pharmaceutical Sciences, Lipscomb University, Nashville, TN 37204, USA
| | | |
Collapse
|
47
|
He C, Wan H. Drug metabolism and metabolite safety assessment in drug discovery and development. Expert Opin Drug Metab Toxicol 2018; 14:1071-1085. [DOI: 10.1080/17425255.2018.1519546] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai, P. R. China
| | - Hong Wan
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co., Ltd., Shanghai, P. R. China
| |
Collapse
|
48
|
Jackson KD, Durandis R, Vergne MJ. Role of Cytochrome P450 Enzymes in the Metabolic Activation of Tyrosine Kinase Inhibitors. Int J Mol Sci 2018; 19:E2367. [PMID: 30103502 PMCID: PMC6121577 DOI: 10.3390/ijms19082367] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Tyrosine kinase inhibitors are a rapidly expanding class of molecular targeted therapies for the treatment of various types of cancer and other diseases. An increasing number of clinically important small molecule tyrosine kinase inhibitors have been shown to undergo cytochrome P450-mediated bioactivation to form chemically reactive, potentially toxic products. Metabolic activation of tyrosine kinase inhibitors is proposed to contribute to the development of serious adverse reactions, including idiosyncratic hepatotoxicity. This article will review recent findings and ongoing studies to elucidate the link between drug metabolism and tyrosine kinase inhibitor-associated hepatotoxicity.
Collapse
Affiliation(s)
- Klarissa D Jackson
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN 37204, USA.
| | - Rebecca Durandis
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN 37204, USA.
| | - Matthew J Vergne
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN 37204, USA.
| |
Collapse
|
49
|
Murugesan D, Ray PC, Bayliss T, Prosser GA, Harrison JR, Green K, Soares de Melo C, Feng TS, Street LJ, Chibale K, Warner DF, Mizrahi V, Epemolu O, Scullion P, Ellis L, Riley J, Shishikura Y, Ferguson L, Osuna-Cabello M, Read KD, Green SR, Lamprecht DA, Finin PM, Steyn AJC, Ioerger TR, Sacchettini J, Rhee KY, Arora K, Barry CE, Wyatt PG, Boshoff HIM. 2-Mercapto-Quinazolinones as Inhibitors of Type II NADH Dehydrogenase and Mycobacterium tuberculosis: Structure-Activity Relationships, Mechanism of Action and Absorption, Distribution, Metabolism, and Excretion Characterization. ACS Infect Dis 2018. [PMID: 29522317 PMCID: PMC5996347 DOI: 10.1021/acsinfecdis.7b00275] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Mycobacterium tuberculosis (MTb) possesses
two nonproton pumping type II NADH dehydrogenase (NDH-2)
enzymes which are predicted to be jointly essential for respiratory
metabolism. Furthermore, the structure of a closely related bacterial
NDH-2 has been reported recently, allowing for the structure-based
design of small-molecule inhibitors. Herein, we disclose MTb whole-cell structure–activity relationships (SARs) for a series of 2-mercapto-quinazolinones which target the ndh encoded NDH-2 with nanomolar potencies. The compounds were inactivated by glutathione-dependent adduct formation as well as quinazolinone oxidation in microsomes. Pharmacokinetic studies demonstrated modest bioavailability and compound exposures. Resistance to the compounds in MTb was conferred by promoter mutations in the alternative nonessential NDH-2 encoded by ndhA in MTb. Bioenergetic analyses revealed a decrease in oxygen consumption rates in response to inhibitor in cells in which membrane potential was uncoupled from ATP production, while inverted membrane vesicles showed mercapto-quinazolinone-dependent inhibition of ATP production when NADH was the electron donor to the respiratory chain. Enzyme kinetic studies further demonstrated noncompetitive inhibition, suggesting binding of this scaffold to an allosteric site. In summary, while the initial MTb SAR showed limited improvement in potency, these results, combined with structural information on the bacterial protein, will aid in the future discovery of new and improved NDH-2 inhibitors.
Collapse
Affiliation(s)
- Dinakaran Murugesan
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Peter C. Ray
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Tracy Bayliss
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Gareth A. Prosser
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Justin R. Harrison
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Kirsteen Green
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Candice Soares de Melo
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Tzu-Shean Feng
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Leslie J. Street
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Kelly Chibale
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- Drug Discovery and Development Centre (H3D), Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| | - Valerie Mizrahi
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| | - Ola Epemolu
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Paul Scullion
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Lucy Ellis
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Jennifer Riley
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Yoko Shishikura
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Liam Ferguson
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Maria Osuna-Cabello
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Kevin D. Read
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Simon R. Green
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Dirk A. Lamprecht
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Peter M. Finin
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
| | - Adrie J. C. Steyn
- Africa Health Research Institute (AHRI), K-RITH Tower Building Level 3, 719 Umbilo Road, Durban, 4001, South Africa
- Department of Microbiology, University of Alabama at Birmingham, 1720 Second Avenue South, Birmingham, Alabama 35294-2170, United States
| | - Thomas R. Ioerger
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Jim Sacchettini
- Department of Computer Science and Engineering, Texas A&M University, College Station, Texas 77843, United States
| | - Kyu Y. Rhee
- Division of Infectious Diseases, Weill Department of Medicine, Weill Cornell Medical College, New York, New York 10065, United States
| | - Kriti Arora
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Clifton E. Barry
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | - Paul G. Wyatt
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Sir James Black Centre, Dundee, DD1 5EH, United Kingdom
| | - Helena I. M. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| |
Collapse
|
50
|
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
|