Volume 15 Issue 3
Apr.  2025
Turn off MathJax
Article Contents
Xiaolan Hu, Jian-Lin Wu, Quan He, Zhi-Qi Xiong, Na Li. Strategy for cysteine-targeting covalent inhibitors screening using in-house database based LC-MS/MS and drug repurposing[J]. Journal of Pharmaceutical Analysis, 2025, 15(3): 101045. doi: 10.1016/j.jpha.2024.101045
Citation: Xiaolan Hu, Jian-Lin Wu, Quan He, Zhi-Qi Xiong, Na Li. Strategy for cysteine-targeting covalent inhibitors screening using in-house database based LC-MS/MS and drug repurposing[J]. Journal of Pharmaceutical Analysis, 2025, 15(3): 101045. doi: 10.1016/j.jpha.2024.101045

Strategy for cysteine-targeting covalent inhibitors screening using in-house database based LC-MS/MS and drug repurposing

doi: 10.1016/j.jpha.2024.101045
Funds:

This work was supported by the Science and Technology Development Fund, Macau SAR, China (Grant Nos.: FDCT 0001/2020/AKP and 006/2023/SKL) and Guangxi Science and Technology Major Program, China (Program No.: Guike AA22096022).

  • Received Date: Apr. 07, 2024
  • Accepted Date: Jul. 16, 2024
  • Rev Recd Date: Jul. 14, 2024
  • Publish Date: Jul. 18, 2024
  • Targeted covalent inhibitors, primarily targeting cysteine residues, have attracted great attention as potential drug candidates due to good potency and prolonged duration of action. However, their discovery is challenging. In this research, a database-assisted liquid chromatography-tandem mass spectrometry (LC-MS/MS) strategy was developed to quickly discover potential cysteine-targeting compounds. First, compounds with potential reactive groups were selected and incubated with N-acetyl-cysteine in microsomes. And the precursor ions of possible cysteine-adducts were predicted based on covalent binding mechanisms to establish in-house database. Second, substrate-independent product ions produced from N-acetyl-cysteine moiety were selected. Third, multiple reaction monitoring scan was conducted to achieve sensitive screening for cysteine-targeting compounds. This strategy showed broad applicability, and covalent compounds with diverse structures were screened out, offering structural resources for covalent inhibitors development. Moreover, the screened compounds, norketamine and hydroxynorketamine, could modify synaptic transmission-related proteins in vivo, indicating their potential as covalent inhibitors. This experimental-based screening strategy provides a quick and reliable guidance for the design and discovery of covalent inhibitors.

  • loading
  • [1]
    J. Singh, R.C. Petter, T.A. Baillie, et al., The resurgence of covalent drugs, Nat. Rev. Drug Discov. 10 (2011) 307-317.
    [2]
    R. Lonsdale, R.A. Ward, Structure-based design of targeted covalent inhibitors, Chem. Soc. Rev. 47 (2018) 3816-3830.
    [3]
    D.S. Hong, M.G. Fakih, J.H. Strickler, et al., KRASG12C inhibition with sotorasib in advanced solid tumors, N. Engl. J. Med. 383 (2020) 1207-1217.
    [4]
    J.A. Woyach, R.R. Furman, T. Liu, et al., Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib, N. Engl. J. Med. 370 (2014) 2286-2294.
    [5]
    V.A. Miller, V. Hirsh, J. Cadranel, et al., Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): A phase 2b/3 randomised trial, Lancet Oncol. 13 (2012) 528-538.
    [6]
    J. M. Strelow, A perspective on the kinetics of covalent and irreversible inhibition. SLAS Discov. 22 (2017) 3-20.
    [7]
    R.A. Bauer, Covalent inhibitors in drug discovery: From accidental discoveries to avoided liabilities and designed therapies, Drug Discov. Today 20 (2015) 1061-1073.
    [8]
    D. Toledo Warshaviak, G. Golan, K.W. Borrelli, et al., Structure-based virtual screening approach for discovery of covalently bound ligands, J. Chem. Inf. Model. 54 (2014) 1941-1950.
    [9]
    J. Lyu, S. Wang, T.E. Balius, et al., Ultra-large library docking for discovering new chemotypes, Nature 566 (2019) 224-229.
    [10]
    A. Fischer, M. Smiesko, M. Sellner, et al., Decision making in structure-based drug discovery: Visual inspection of docking results, J. Med. Chem. 64 (2021) 2489-2500.
    [11]
    N. London, R.M. Miller, S. Krishnan, et al., Covalent docking of large libraries for the discovery of chemical probes, Nat. Chem. Biol. 10 (2014) 1066-1072.
    [12]
    E. Resnick, A. Bradley, J. Gan, et al., Rapid covalent-probe discovery by electrophile-fragment screening, J. Am. Chem. Soc. 141 (2019) 8951-8968.
    [13]
    M.M. McCallum, P. Nandhikonda, J.J. Temmer, et al., High-throughput identification of promiscuous inhibitors from screening libraries with the use of a thiol-containing fluorescent probe, J. Biomol. Screen. 18 (2013) 705-713.
    [14]
    D.A. Erlanson, S.W. Fesik, R.E. Hubbard, et al., Twenty years on: The impact of fragments on drug discovery, Nat. Rev. Drug Discov. 15 (2016) 605-619.
    [15]
    A.J. Maurais, E. Weerapana, Reactive-cysteine profiling for drug discovery, Curr. Opin. Chem. Biol. 50 (2019) 29-36.
    [16]
    Q. Zhang, P. Luo, L. Zheng, et al., 18beta-glycyrrhetinic acid induces ROS-mediated apoptosis to ameliorate hepatic fibrosis by targeting PRDX1/2 in activated HSCs, J. Pharm. Anal. 12 (2022) 570-582.
    [17]
    L. Boike, N.J. Henning, D.K. Nomura, Advances in covalent drug discovery, Nat. Rev. Drug Discov. 21 (2022) 881-898.
    [18]
    S. Pushpakom, F. Iorio, P.A. Eyers, et al., Drug repurposing: Progress, challenges and recommendations, Nat. Rev. Drug Discov. 18 (2019) 41-58.
    [19]
    S. Gong, X. Hu, S. Chen, et al., Dual roles of drug or its metabolite-protein conjugate: Cutting-edge strategy of drug discovery using shotgun proteomics, Med. Res. Rev. 42 (2022) 1704-1734.
    [20]
    J.W.T. Yates, S. Ashton, D. Cross, et al., Irreversible inhibition of EGFR: Modeling the combined pharmacokinetic-pharmacodynamic relationship of osimertinib and its active metabolite AZ5104, Mol. Cancer Ther. 15 (2016) 2378-2387.
    [21]
    S. Gong, Y. Zhuo, S. Chen, et al., Quantification of osimertinib and metabolite-protein modification reveals its high potency and long duration of effects on target organs, Chem. Res. Toxicol. 34 (2021) 2309-2318.
    [22]
    Z. Li, X. Zhang, J. Liao, et al., An ultra-robust fingerprinting method for quality assessment of traditional Chinese medicine using multiple reaction monitoring mass spectrometry, J. Pharm. Anal. 11 (2021) 88-95.
    [23]
    X. Bian, Y. Zhang, N. Li, et al., Ultrasensitive quantification of trace amines based on N-phosphorylation labeling chip 2D LC-QQQ/MS, J. Pharm. Anal. 13 (2023) 315-322.
    [24]
    Y. Zhuo, J.-L. Wu, X. Yan, et al., Strategy for hepatotoxicity prediction induced by drug reactive metabolites using human liver microsome and online 2D-nano-LC-MS analysis, Anal. Chem. 89 (2017) 13167-13175.
    [25]
    X. Hu, J.-L. Wu, W. Miao, et al., Covalent protein modification: An unignorable factor for bisphenol A-induced hepatotoxicity, Environ. Sci. Technol. 56 (2022) 9536-9545.
    [26]
    A. Abdeldayem, Y.S. Raouf, S.N. Constantinescu, et al., Advances in covalent kinase inhibitors, Chem. Soc. Rev. 49 (2020) 2617-2687.
    [27]
    T.J. Monks, M. Butterworth, S.S. Lau, The fate of benzene-oxide, Chem. Biol. Interact. 184 (2010) 201-206.
    [28]
    Q. He, D.V. Titov, J. Li, et al., Covalent modification of a cysteine residue in the XPB subunit of the general transcription factor TFIIH through single epoxide cleavage of the transcription inhibitor triptolide, Angew. Chem. Int. Ed 54 (2015) 1859-1863.
    [29]
    F. Du, Z. Liu, X. Li, et al., Metabolic pathways leading to detoxification of triptolide, a major active component of the herbal medicine Tripterygium wilfordii, J. Appl. Toxicol. 34 (2014) 878-884.
    [30]
    Y. Bai, W. Peng, C. Yang, et al., Pharmacokinetics and metabolism of naringin and active metabolite naringenin in rats, dogs, humans, and the differences between species, Front. Pharmacol. 11 (2020), 364.
    [31]
    K. Huang, L. Huang, R.B. van Breemen, Detection of reactive metabolites using isotope-labeled glutathione trapping and simultaneous neutral loss and precursor ion scanning with ultra-high-pressure liquid chromatography triple quadruple mass spectrometry, Anal. Chem. 87 (2015) 3646-3654.
    [32]
    P. Liu, Y. Huang, W. Cai, et al., Profiling of thiol-containing compounds by stable isotope labeling double precursor ion scan mass spectrometry, Anal. Chem. 86 (2014) 9765-9773.
    [33]
    J. Kim, T. Farchione, A. Potter, et al., Esketamine for treatment-resistant depression - first FDA-approved antidepressant in a new class, N. Engl. J. Med. 381 (2019) 1-4.
    [34]
    S. Sreeramulu, S.L. Gande, M. Gobel, et al., Molecular mechanism of inhibition of the human protein complex Hsp90-Cdc37, a kinome chaperone-cochaperone, by triterpene celastrol, Angew. Chem. Int. Ed 48 (2009) 5853-5855.
    [35]
    J. Shi, J. Li, Z. Xu, et al., Celastrol: A review of useful strategies overcoming its limitation in anticancer application, Front. Pharmacol. 11 (2020), 558741.
    [36]
    E.A. Burnham, A.A. Abouda, J.E. Bissada, et al., Interindividual variability in cytochrome P450 3A and 1A activity influences sunitinib metabolism and bioactivation, Chem. Res. Toxicol. 35 (2022) 792-806.
    [37]
    G.M. Amaya, R. Durandis, D.S. Bourgeois, et al., Cytochromes P450 1A2 and 3A4 catalyze the metabolic activation of sunitinib, Chem. Res. Toxicol. 31 (2018) 570-584.
    [38]
    C. Fabbri, S. Kasper, J. Zohar, et al., Drug repositioning for treatment-resistant depression: Hypotheses from a pharmacogenomic study, Prog. Neuropsychopharmacol Biol. Psychiatry 104 (2021), 110050.
    [39]
    M. Ke, H. Shen, L. Wang, et al., Identification, quantification, and site localization of protein posttranslational modifications via mass spectrometry-based proteomics, Adv. Exp. Med. Biol. 919 (2016) 345-382.
    [40]
    R. Kandaswamy, A. McQuillin, S.I. Sharp, et al., Genetic association, mutation screening, and functional analysis of a Kozak sequence variant in the metabotropic glutamate receptor 3 gene in bipolar disorder, JAMA Psychiatry 70 (2013) 591-598.
    [41]
    L. Lyon, J.N.C. Kew, C. Corti, et al., Altered hippocampal expression of glutamate receptors and transporters in GRM2 and GRM3 knockout mice, Synapse 62 (2008) 842-850.
    [42]
    A.G. Walker, C.J. Wenthur, Z. Xiang, et al., Metabotropic glutamate receptor 3 activation is required for long-term depression in medial prefrontal cortex and fear extinction, Proc. Natl. Acad. Sci. USA 112 (2015) 1196-1201.
    [43]
    J.L. Engers, K.A. Bollinger, R.L. Weiner, et al., Design and synthesis of N-aryl phenoxyethoxy pyridinones as highly selective and CNS penetrant mGlu3 NAMs, ACS Med. Chem. Lett. 8 (2017) 925-930.
    [44]
    H. Koike, S. Chaki, Requirement of AMPA receptor stimulation for the sustained antidepressant activity of ketamine and LY341495 during the forced swim test in rats, Behav. Brain Res. 271 (2014) 111-115.
    [45]
    J.M. Dwyer, A.E. Lepack, R.S. Duman, mTOR activation is required for the antidepressant effects of mGluR2/3 blockade, Int. J. Neuropsychopharmacol. 15 (2012) 429-434.
    [46]
    N.B. Senese, M.M. Rasenick, J.R. Traynor, The role of G-proteins and G-protein regulating proteins in depressive disorders, Front. Pharmacol. 9 (2018), 1289.
    [47]
    A.F. Abdel-Magid, Inhibition of O-GlcNAcase (OGA): A potential therapeutic target to treat Alzheimer’s disease, ACS Med. Chem. Lett. 5 (2014) 1270-1271.
    [48]
    C. Yang, S. Kobayashi, K. Nakao, et al., AMPA receptor activation-independent antidepressant actions of ketamine metabolite (S)-norketamine, Biol. Psychiatry 84 (2018) 591-600.
    [49]
    P. Zanos, T.D. Gould, Mechanisms of ketamine action as an antidepressant, Mol. Psychiatry 23 (2018) 801-811.
  • 加载中

Catalog

    通讯作者: 陈斌, bchen63@163.com
    • 1. 

      沈阳化工大学材料科学与工程学院 沈阳 110142

    1. 本站搜索
    2. 百度学术搜索
    3. 万方数据库搜索
    4. CNKI搜索

    Figures(1)

    Article Metrics

    Article views (299) PDF downloads(19) Cited by()
    Proportional views
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return